1
|
Shen Y, Huang H, Shen L, Yao W, Wang R, Kang M, Huang J, Xie Y, Yang H. ZBTB16 DRIVES VASCULAR CALCIFICATION THROUGH ACCELERATING VSMCS OSTEOBLASTIC TRANSITION IN CHRONIC KIDNEY DISEASE VIA WNT/Β-CATENIN PATHWAY. Shock 2025; 63:312-319. [PMID: 39450908 DOI: 10.1097/shk.0000000000002488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
ABSTRACT Chronic kidney disease (CKD)-related vascular calcification (VC) is a common degenerative phenomenon of the vessel wall and its pathological basis is the phenotypic transformation of vascular smooth muscle cells (VSMCs). Zinc finger and BR-C (Broad-Complex), ttk (tramtrack), and bab (bric à brac) (BTB) domain containing 16 (ZBTB16) have been reported to be expressed in the aortic tissues in a rat model of VC. This work is conducted to reveal the functions of ZBTB16 on VC in CKD and to probe its involved reaction mechanisms. In vivo CKD rat models were established by adenine and VSMC calcification were stimulated with high phosphate (Pi) in vitro . Renal function indexes were estimated with relevant assay kits. Renal tissues were histologically examined with hematoxylin and eosin staining. Alizarin red and von kossa staining were used to measure arterial calcification. Reverse transcription-quantitative PCR and western blot were used to detect ZBTB16 expression. Western blot, immunohistochemistry, and immunofluorescence staining were used to detect osteogenic markers and smooth muscle cell markers. Western blot was used to measure the expressions of proteins implicated in Wnt/β-catenin pathway. In the blood samples of CKD patients with VC, aortic tissues of CKD rats, and Pi-treated VSMCs, ZBTB16 expression was significantly increased. ZBTB16 knockdown reduced renal dysfunction, calcium deposition and inhibited VSMCs osteoblast differentiation both in vitro and in vivo . Moreover, silencing with ZBTB16 inactivated Wingless-related integration site (Wnt)/β-catenin pathway. LiCl (Wnt/β-catenin agonist) reversed the protective effects of ZBTB16 knockdown on the calcification and osteoblastic transformation in vitro . Together, ZBTB16 silencing may downregulate Wnt/β-catenin pathway to protect against CKD-associated VC via repressing the osteoblastic transformation of VSMCs.
Collapse
Affiliation(s)
| | - Huaxing Huang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Lianglan Shen
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Wubin Yao
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Rong Wang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Meizi Kang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jiashan Huang
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Xie
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongli Yang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
2
|
Lin W, Li Y, Qiu C, Zou B, Gong Y, Zhang X, Tian D, Sherman W, Sanchez F, Wu D, Su KJ, Xiao X, Luo Z, Tian Q, Chen Y, Shen H, Deng H. Mapping the spatial atlas of the human bone tissue integrating spatial and single-cell transcriptomics. Nucleic Acids Res 2025; 53:gkae1298. [PMID: 39817519 PMCID: PMC11736439 DOI: 10.1093/nar/gkae1298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
Bone is a multifaceted tissue requiring orchestrated interplays of diverse cells within specialized microenvironments. Although significant progress has been made in understanding cellular and molecular mechanisms of component cells of bone, revealing their spatial organization and interactions in native bone tissue microenvironment is crucial for advancing precision medicine, as they govern fundamental signaling pathways and functional dependencies among various bone cells. In this study, we present the first integrative high-resolution map of human bone and bone marrow, using spatial and single-cell transcriptomics profiling from femoral tissue. This multi-modal approach discovered a novel bone formation-specialized niche enriched with osteoblastic lineage cells and fibroblasts and unveiled critical cell-cell communications and co-localization patterns between osteoblastic lineage cells and other cells. Furthermore, we discovered a novel spatial gradient of cellular composition, gene expression and signaling pathway activities radiating from the trabecular bone. This comprehensive atlas delineates the intricate bone cellular architecture and illuminates key molecular processes and dependencies among cells that coordinate bone metabolism. In sum, our study provides an essential reference for the field of bone biology and lays the foundation for advanced mechanistic studies and precision medicine approaches in bone-related disorders.
Collapse
Affiliation(s)
- Weiqiang Lin
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Yisu Li
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6823 St. Charles Avenue, Uptown, New Orleans, LA 70118, USA
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Binghao Zou
- Department of Structural and Cellular Biology, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Yun Gong
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Xiao Zhang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Di Tian
- The Molecular Pathology Laboratory, Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - William Sherman
- Department of Orthopaedic Surgery, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Fernando Sanchez
- Department of Orthopaedic Surgery, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Di Wu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Xinyi Xiao
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Zhe Luo
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Yiping Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6823 St. Charles Avenue, Uptown, New Orleans, LA 70118, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Hongwen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Dong R, Ji Z, Wang M, Ma G. Role of macrophages in vascular calcification: From the perspective of homeostasis. Int Immunopharmacol 2025; 144:113635. [PMID: 39566391 DOI: 10.1016/j.intimp.2024.113635] [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: 09/28/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Vascular calcification (VC) is a crucial risk factor for the high morbidity and mortality associated with cardiovascular and cerebrovascular diseases. With the global population aging, the incidence of VC is escalating annually. However, due to its silent clinical process, VC often results in irreversible clinical outcomes. Inflammation is a core element in the VC process, and macrophages are the major inflammatory cells. Due to their diverse origins, microenvironments, and polarization states, macrophages exhibit significant heterogeneity, exerting strong effects on the occurrence, development, and even the regression of VC. In this review, we summarize the origin, distribution, classification, and surface markers of macrophages. Simultaneously, we explore the mechanisms by which macrophages maintain homeostasis or regulate inflammation, including the macrophage-mediated regulation of VC through the release of inflammatory factors, osteogenic genes, extracellular vesicles, and alterations in efferocytosis. Finally, we discuss research targeting inflammation and macrophages to develop novel therapeutic regimens for preventing and treating VC.
Collapse
Affiliation(s)
- Rong Dong
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing 210009, China; Department of Cardiology, Yancheng No. 1 People's Hospital, No. 66 South Renmin Road, Yancheng 224000, China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing 210009, China
| | - Mi Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing 210009, China.
| |
Collapse
|
4
|
Yang S, Chen M, Yang D, Guo X. Perfluorooctanoic acid and its alternatives disrupt the osteogenesis and osteoclastogenesis balance: Evidence from the effects on cell differentiation process. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 959:178331. [PMID: 39754941 DOI: 10.1016/j.scitotenv.2024.178331] [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: 09/11/2024] [Revised: 11/14/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
In the present study, we investigated the effects of a representative of the per- and polyfluoroalkyl substance (PFAS) chemical group, namely perfluorooctanoic acid (PFOA), and its alternatives (perfluorobutanoic acid [PFBA] and the hexafluoropropylene oxide dimer acid [GenX]) on bone homeostasis, a process that mainly depends on osteoblast (OB) and osteoclast (OC) activities at the cellular level. C3H10T1/2 cells and bone marrow macrophages (BMMs) were respectively induced into OBs and OCs, and treated with PFOA, PFBA, and GenX at doses of 0.25, 2.5, and 25 μM. Real-time PCR, ALP activity analysis, ARS staining, and TRAP staining were performed to assess cell differentiation. To explore the indirect effects on OC differentiation, conditioned media were collected from 7-day PFAS-treated C3H10T1/2 cells during differentiation. RANKL/OPG expression and secretion levels in C3H10T1/2 cells were determined, and BMMs induced into OCs were incubated in conditioned media for two days. PFOA suppressed osteogenesis characterized by the decreased mRNA expression of OB-specific molecules, ALP activity, and calcium deposition, and promoted osteoclastogenesis as evidenced by upregulated osteoclastic genes and increased number of TRAP-positive cells. Although the PFOA alternatives did not affect early osteogenesis, calcium deposits were significantly reduced and osteoclastogenesis was facilitated. The results suggested that PFOA and its alternatives could directly disturb the balance between osteogenesis and osteoclastogenesis. In addition, PFOA and its alternatives enhanced the RANKL/OPG ratio during early OB differentiation, and more BMMs stimulated with the conditioned medium differentiated into OCs, indicating the indirect stimulation on OC differentiation by PFOA and its alternatives. These findings highlight the potential skeletal hazards of PFASs, particularly emerging alternatives that might be ignored, offering a toxicological basis for further exploration.
Collapse
Affiliation(s)
- Sijia Yang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Mengyuan Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Di Yang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, No. 38 Xueyuan Road, Beijing, China.
| |
Collapse
|
5
|
Yamashita Y, Hayashi M, Liu A, Sasaki F, Tsuchiya Y, Takayanagi H, Saito M, Nakashima T. Fam102a translocates Runx2 and Rbpjl to facilitate Osterix expression and bone formation. Nat Commun 2025; 16:9. [PMID: 39747056 PMCID: PMC11695619 DOI: 10.1038/s41467-024-55451-z] [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: 02/19/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Bone remodeling maintains the robustness of the bone tissue by balancing bone resorption by osteoclasts and bone formation by osteoblasts. Although these cells together play a crucial role in bone remodeling, only a few reports are available on the common factors involved in the differentiation of the two types of cells. Here, we show family with sequence similarity 102 member A (Fam102a) as a bone-remodeling factor that positively regulates both osteoclast and osteoblast differentiation. Fam102a regulates osteoblast differentiation by controlling recombination signal binding protein for immunoglobulin κ J region-like (Rbpjl). The Fam102a-Rbpjl axis promotes the nuclear translocation of transcription factors and enhances the expression of Osterix, a transcription factor essential for osteoblast differentiation. The deletion of Fam102a or a functional mutation in Rbpjl leads to osteopenia accompanied by reduced osteoblastic bone formation. Thus, the Fam102a-Rbpjl axis plays an important role in osteoblasts and this finding provides insights into bone remodeling.
Collapse
Affiliation(s)
- Yu Yamashita
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Mikihito Hayashi
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| | - Anhao Liu
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Fumiyuki Sasaki
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Yosuke Tsuchiya
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
6
|
Maji S, Aliabouzar M, Quesada C, Chiravuri A, Macpherson A, Pinch A, Kazyak K, Emara Z, Abeid BA, Kent RN, Midekssa FS, Zhang M, Baker BM, Franceschi RT, Fabiilli ML. Ultrasound-generated bubbles enhance osteogenic differentiation of mesenchymal stromal cells in composite collagen hydrogels. Bioact Mater 2025; 43:82-97. [PMID: 39345992 PMCID: PMC11439547 DOI: 10.1016/j.bioactmat.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Hydrogels can improve the delivery of mesenchymal stromal cells (MSCs) by providing crucial biophysical cues that mimic the extracellular matrix. The differentiation of MSCs is dependent on biophysical cues like stiffness and viscoelasticity, yet conventional hydrogels cannot be dynamically altered after fabrication and implantation to actively direct differentiation. We developed a composite hydrogel, consisting of type I collagen and phase-shift emulsion, where osteogenic differentiation of MSCs can be non-invasively modulated using ultrasound. When exposed to ultrasound, the emulsion within the hydrogel was non-thermally vaporized into bubbles, which locally compacted and stiffened the collagen matrix surrounding each bubble. Bubble growth and matrix compaction were correlated, with collagen regions proximal (i.e., ≤ ∼60 μm) to the bubble displaying a 2.5-fold increase in Young's modulus compared to distal regions (i.e., > ∼60 μm). The viability and proliferation of MSCs, which were encapsulated within the composite hydrogel, were not impacted by bubble formation. In vitro and in vivo studies revealed encapsulated MSCs exhibited significantly elevated levels of RUNX2 and osteocalcin, markers of osteogenic differentiation, in collagen regions proximal to the bubble compared to distal regions. Additionally, alkaline phosphatase activity and calcium deposition were enhanced adjacent to the bubble. An opposite trend was observed for CD90, a marker of MSC stemness. Following subcutaneous implantation, bubbles persisted in the hydrogels for two weeks, which led to localized collagen alignment and increases in nuclear asymmetry. These results are a significant step toward controlling the 3D differentiation of MSCs in a non-invasive and on-demand manner.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Mitra Aliabouzar
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Carole Quesada
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Anjali Chiravuri
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aidan Macpherson
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Abigail Pinch
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Karsyn Kazyak
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Ziyad Emara
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Bachir A Abeid
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Firaol S Midekssa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Man Zhang
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Renny T Franceschi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Fang T, Zhang R, Song F, Chu X, Fu Q, Wu Q. miR-468-3p suppresses osteogenic differentiation of BMSCs by targeting Runx2 and inhibits bone formation. J Orthop Surg Res 2024; 19:887. [PMID: 39734217 DOI: 10.1186/s13018-024-05410-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
An improved understanding of the molecular actions underpinning bone marrow mesenchymal stem cell (BMSC) differentiation could highlight new therapeutics for osteoporosis (OP). Current evidence indicates that microRNAs (miRNAs) exert critical roles in many biological systems, including osteoblast differentiation. In this study, we examined miR-468-3p effects on osteogenic differentiation (OD). Distinct miR-468-3p reductions were identified during OD. MiR-468-3p also suppressed BMSC OD in gain- and loss-of-function assays, while it negatively regulated Runx2 as shown by molecular, protein, and bioinformatics approaches. When Runx2 was inhibited by small-interfering RNA (siRNA), the inhibitory effects of miR-468-3p toward BMSC osteogenesis were considerably reversed. Also, silenced miR-468-3p in ovariectomized (OVX) and sham mice augmented bone mass (BM) and bone formation (BF) and improved trabecular (Tb) microarchitecture. Therefore, miR-468-3p is a novel Runx2 regulator with key physiological action in BF and OD.
Collapse
Affiliation(s)
- Tao Fang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266000, China
| | - Ranxi Zhang
- Department of Spine Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266000, China
| | - Feng Song
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266000, China
| | - Xueru Chu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, 266000, China
| | - Qin Fu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Qianqian Wu
- Department of Cardiology, Qingdao Municipal Hospital, 1 Jiaozhou Road, Qingdao, Shandong, 266000, China.
| |
Collapse
|
8
|
Zhang Q, Zhang Y, Li B, Wang C, Yang Z, Guo B, Yue Z. Melatonin promotes the proliferation and differentiation of antler chondrocytes via RUNX2 dependent on the interaction between NOTCH1 and SHH signaling pathways. Cell Biol Int 2024. [PMID: 39737592 DOI: 10.1002/cbin.12272] [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: 08/13/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025]
Abstract
Melatonin (MT), an endogenous hormone secreted by pineal gland, has the sedative, anti-inflammatory and antioxidant functions. However, there are few studies on whether MT affects the proliferation and differentiation of antler chondrocytes. The present study investigated the influences of MT on the proliferation and differentiation of antler chondrocytes, explored its regulation on runt-related transcription factor 2 (RUNX2), NOTCH1 and sonic hedgehog (SHH) signaling, and elucidated their interplays. The results showed that MT promoted the proliferation of antler chondrocytes and induced the differentiation of chondrocytes into hypertrophic chondrocytes as evidenced by the significant increase of collagen type X (COL X), alkaline phosphatase (ALP) and matrix metalloproteinase 13 (MMP13) expression and ALP activity, the well-established markers for hypertrophic chondrocytes, but this effectiveness was neutralized by the addition of MT receptor antagonist. Further analysis indicated that MT activated the NOTCH1 and SHH signaling whose blockage abrogated the inducement of MT on the proliferation and differentiation of antler chondrocytes. SHH was identified as a downstream target of recombination signal binding protein for immunoglobulin kappa J region (RBPJ), a transcription factor of NOTCH1 signaling. Meanwhile, MT stimulated the expression of RUNX2 through activating the SHH signaling whose downstream transcription factor glioma-associated oncogene 1 (GLI1) directly controlled the transcription of RUNX2 through binding to its promoter region. Moreover, repression of GLI1 counteracted the proliferative effect of MT on antler chondrocytes and attenuated the advancement of MT on chondrocyte differentiation, while supplementation of recombinant RUNX2 protein recued above effects. Collectively, MT induced the proliferation and differentiation of antler chondrocytes via RUNX2 dependent on the interaction between NOTCH1 and SHH signaling pathways.
Collapse
Affiliation(s)
- Qiaoling Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yan Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Baiyu Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chenhao Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhanqing Yang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Bin Guo
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhanpeng Yue
- College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
9
|
Pang H, Ding L, Che X. Early effects of α7nAChR regulation on maxillary expansion in mice : A study on osteogenesis and inflammatory factors. J Orofac Orthop 2024:10.1007/s00056-024-00565-8. [PMID: 39729130 DOI: 10.1007/s00056-024-00565-8] [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: 12/11/2023] [Accepted: 10/04/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE We aimed to investigate early effects of regulating alpha‑7 nicotinic acetylcholine receptor (α7nAChR) agonists and antagonists on maxillary expansion in mice. METHODS We allocated 36 six-week-old male C57BL/6J mice into three group: 1) expansion alone, 2) expansion plus the α7nAChR-specific agonist 3‑(2,4-dimethoxybenzylidene)-anabaseine dihydrochloride (GTS-21), and 3) expansion plus alpha-bungarotoxin (α-BTX), a competitive antagonist of α7nAChR. The groups were daily injected with saline, GTS-21 (4 mg/kg/day) or α‑BTX (1 mg/kg/day), respectively, from days 0-7. In addition, a mouse model of maxillary expansion was established. Masson's trichrome staining was used to observe morphological changes and immunohistochemistry was performed to analyze α7nAChR, interleukin (IL)-1β, IL‑6, tumor necrosis factor (TNF)-α, runt-related transcription factor 2 (RUNX2), and osteocalcin (OCN) expression in the midpalatal suture. Microcomputed tomography was used to measure midpalatal suture and palatal basal bone widths. We assessed the normal distribution of our data using the Kolmogorov-Smirnoff test and evaluated the homogeneity of variance by Levene's test, followed by a two-way ANOVA and Bonferroni tests at a significance level of P < 0.05. RESULTS In the GTS-21+expansion group, osteogenesis was more active in the middle palatine suture. New bone was calcified and deposited in the suture and we observed decreased IL-1β, IL‑6, and TNF‑α expression (P < 0.05). In the α‑BTX+expansion group, we observed increased proinflammatory cytokine and decreased RUNX2 and OCN expression and increased midpalatal suture and palatal basal bone widths (P < 0.05). CONCLUSION Using α7nAChR agonists and antagonists to regulate the cholinergic anti-inflammatory pathway, the secretion of inflammatory factors and osteoblast markers during maxillary expansion were altered, indicating the potential for clinical modulation of maxillary palatal suture expansion.
Collapse
Affiliation(s)
- Huiqi Pang
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University, 100050, Beijing, China
| | - Luhua Ding
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University, 100050, Beijing, China
| | - Xiaoxia Che
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University, 100050, Beijing, China.
| |
Collapse
|
10
|
Dong Y, Ma P, Wang S, Wang L, Chen Y, Zhao F, Yang K, Zhang X, Zhao H, Li B, Geng R, Tang TS, Zheng Q, Zheng T. TMCO1-deficient mice exhibit a high incidence of otitis media associated with impaired bone homeostasis in the middle ear. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00471-1. [PMID: 39725295 DOI: 10.1016/j.ajpath.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024]
Abstract
Craniofacial dysmorphism, skeletal anomalies and impaired intellectual development syndrome" (CFSMR1; OMIM#213980) is characterized by craniofacial dysmorphism, skeletal anomalies, and mental retardation. However, reports of hearing issues have been limited. To investigate hearing-related aspects of CFSMR1, Tmco1 knockout mice (Tmco1-/-) exhibiting similar symptoms to human patients were utilized in this study. Otitis media was discovered in approximately 80% of Tmco1-/- mice, which led to moderate conductive hearing loss at 3 months old and further progressed to deafness two months later. Pathology studies of Tmco1-/- mice revealed a thickened middle ear (ME) epithelium and pronounced inflammatory infiltrates in the ME cavity (MEC) and Eustachian tube (ET) of Tmco1-/- OM mice. Micro-CT scan of 5-month-old Tmco1-/- OM mice showed significantly reduced ME volume and ME malformation. Tartrate-resistant acid phosphatase (TRAP) and RUNX2, RANKL expression in ME revealed increased osteoclast activity and significantly decreased bone formation, suggesting potential causes of ME malformation. This study represents the first report of the audiological characteristics and the elucidation of potential mechanisms in Tmco1-/- mice. It enriches our understanding of the phenotypes associated with CFSMR1 in the field of otology and provides a promising model for chronic OM with conductive hearing loss.
Collapse
Affiliation(s)
- Yaning Dong
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China; Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, China
| | - Peng Ma
- Department of Medical Genetics and Cell Biology, School of basic medicine, Binzhou Medical University, Yantai, China
| | - Shuli Wang
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Lan Wang
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Yingying Chen
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Fangfang Zhao
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Keyan Yang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Xiaolin Zhang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Hongchun Zhao
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Bo Li
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Ruishuang Geng
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Tie-Shan Tang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Qingyin Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China; Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China.
| | - Tihua Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China.
| |
Collapse
|
11
|
Nan G, Peng W, Xu S, Wang G, Zhang J. The Development of Prenatal Muscle Satellite Cells (MuSCs) and Their Epigenetic Modifications During Skeletal Muscle Development in Yak Fetus. BIOLOGY 2024; 13:1091. [PMID: 39765758 PMCID: PMC11673279 DOI: 10.3390/biology13121091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
To investigate prenatal muscle satellite cell (MuSC) development and the associated epigenetic modifications in yak. Here, we conducted morphological and protein co-localization analyses of fetal longissimus dorsi muscle at various developmental stages using histology and immunofluorescence staining methods. Our study observed that primary muscle fibers began forming at 40 days of gestation, fully developed by 11 weeks, and secondary muscle fibers were predominantly formed by around 105 days. Throughout development, MuSCs were mainly located between the muscle fiber membrane and the basement membrane, acting as a reserve for the stem cell pool. MuSCs appeared within myotubes only during critical phases of primary and secondary muscle fiber formation. The proliferation of MuSCs gradually decreases until birth. MuSCs with 5mC modification show a trend of increasing first and then decreasing. MuSCs with 5hmC modification also present a dynamic change trend. The 41st day and 11th week are the critical periods for the changes of both. From the 11th week to around the 110th day of gestation, the modification effect of histone H3K4me3 is crucial for MuSCs during the development of the fetal longissimus dorsi muscle. Combined, our data identify key time points for yak fetal skeletal muscle growth and development and demonstrate that DNA methylation and histone modifications in MuSCs are closely related to this process, offering a valuable basis for future research into the molecular mechanisms underlying yak muscle development.
Collapse
Affiliation(s)
| | | | | | - Guowen Wang
- College of Animal Husbandry and Veterinary Science, Qinghai University, Xining 810016, China; (G.N.); (W.P.); (S.X.)
| | - Jun Zhang
- College of Animal Husbandry and Veterinary Science, Qinghai University, Xining 810016, China; (G.N.); (W.P.); (S.X.)
| |
Collapse
|
12
|
Da Silva D, Crous A, Abrahamse H. Synergistic Effects of Photobiomodulation and Differentiation Inducers on Osteogenic Differentiation of Adipose-Derived Stem Cells in Three-Dimensional Culture. Int J Mol Sci 2024; 25:13350. [PMID: 39769115 PMCID: PMC11678880 DOI: 10.3390/ijms252413350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoporosis, a common metabolic bone disorder, leads to increased fracture risk and significant morbidity, particularly in postmenopausal women and the elderly. Traditional treatments often fail to fully restore bone health and may cause side effects, prompting the exploration of regenerative therapies. Adipose-derived stem cells (ADSCs) offer potential for osteoporosis treatment, but their natural inclination toward adipogenic rather than osteogenic differentiation poses a challenge. This study investigates a novel approach combining differentiation inducers (DIs), three-dimensional (3D) hydrogel scaffolds, and photobiomodulation (PBM) to promote osteogenic differentiation of immortalised ADSCs. A dextran-based 3D hydrogel matrix, supplemented with a DI cocktail of dexamethasone, β-glycerophosphate disodium, and ascorbic acid, was used to foster osteogenesis. PBM was applied using near-infrared (825 nm), green (525 nm), and combined wavelengths at fluences of 3 J/cm2, 5 J/cm2, and 7 J/cm2 to enhance osteogenic potential. Flow cytometry identified osteoblast-specific markers, while inverted light microscopy evaluated cellular morphology. Reactive oxygen species assays measured oxidative stress, and quantitative polymerase chain reaction (qPCR) revealed upregulated gene expression linked to osteogenesis. The findings demonstrate that integrating DIs, 3D hydrogels, and PBM effectively drives osteogenic differentiation in immortalised ADSCs. The PBM enhanced osteogenic marker expression, induced morphological changes, and upregulated gene activity, presenting a promising framework for bone regeneration. Future research should assess the stability and functionality of these differentiated cells and explore their applicability in preclinical models of bone injury or degeneration. This integrative approach demonstrated specific efficacy in promoting the osteogenic differentiation of ADSCs, highlighting its potential application in developing targeted treatments for osteoporosis.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (D.D.S.); (A.C.)
| |
Collapse
|
13
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
14
|
Li Y, Luo Y, Huang D, Peng L. Sclerostin as a new target of diabetes-induced osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1491066. [PMID: 39720253 PMCID: PMC11666367 DOI: 10.3389/fendo.2024.1491066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Sclerostin, a protein synthesized by bone cells, is a product of the SOST gene. Sclerostin is a potent soluble inhibitor of the WNT signaling pathway, and is known to inhibit bone formation by inhibiting osteocyte differentiation and function. Currently, sclerostin has been the subject of numerous animal experiments and clinical investigations. By conducting a literature review, we have gained insights into the most recent advancements in research. Patients with both type 1 diabetes and type 2 diabetes have high levels of serum sclerostin. Patients with type 1 diabetes and type 2 diabetes are both more likely to suffer from osteoporosis, and serum sclerostin levels are elevated in osteoporosis. Many studies have confirmed that sclerostin has been implicated in the pathogenesis of osteoporosis, so we speculate that sclerostin plays an important role in osteoporosis through the glucose metabolism pathway, which may promote the osteoporosis of morbidity in type 1 diabetes and type 2 diabetes. Based on this, we propose whether serum sclerostin can predict type 1 diabetes and type 2 diabetes-induced osteoporosis, and whether it can be a new target for the prevention and treatment of type 1 diabetes and type 2 diabetes-induced osteoporosis, providing new ideas for clinicians and researchers.
Collapse
Affiliation(s)
- Yanhua Li
- Department of Endocrinology and Metabolism, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Yaheng Luo
- Department of Endocrinology and Metabolism, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Debin Huang
- Department of Endocrinology and Metabolism, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Lele Peng
- Department of Endocrinology and Metabolism, Want Want Hospital, Changsha, Hunan, China
| |
Collapse
|
15
|
Zhou Y, Lu Y, Xu H, Ji X, Deng Q, Wang X, Zhang Y, Li Q, Lu Y, Rustempasic A, Liu Y, Wang Y. The effect of miR-205a with RUNX2 towards proliferation and differentiation of chicken chondrocytes in thiram-induced tibial dyschondroplasia. Poult Sci 2024; 103:104535. [PMID: 39541878 PMCID: PMC11609359 DOI: 10.1016/j.psj.2024.104535] [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: 07/30/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Tibial dyschondroplasia (TD) is a kind of metabolic bone disease in fast-growing broilers, which seriously restricts the development of poultry industry. Our previous studies have revealed a significant upregulation of miR-205a in TD cartilage tissue, suggesting its potential role as a regulatory factor in the pathogenesis of TD. However, the precise function implications and underlying regulatory mechanism remain elusive. Therefore, this study aims to elucidate the biological functions and regulatory mechanisms of miR-205a in the progression of TD by employing mehtodologies such as qRT-PCR, CCK-8 assay, EdU assays, and flow cytometry. The findings demonstrated that the transfection of miR-205a overexpression plasmid reduced chondrocytes growth and development in TD while enhancing apoptosis; conversely, blocking miR-205a had opposite effects. RUNX2 was identified as a target gene of miR-205a through biosynthesis and dual luciferase assays, and its overexpression helps chondrocytes in TD grow and develop. However, when both miR-205a and RUNX2 were overexpressed, the regulatory effect of RUNX2 was significantly suppressed. In conclusion, miR-205a plays a role in slowing the growth and development of chondrocytes in TD by targeting and reducing RUNX2 expression, which helps to initiate and progress TD.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yuxiang Lu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Hengyong Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xuyang Ji
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Qingqing Deng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xi Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yao Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Qiuhang Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yusheng Lu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Alma Rustempasic
- Faculty of Agriculture and Food Science, University in Sarajevo, Zmaja od Bosne 8, 71000 Sarajevo, Bosnia and Herzegovina
| | - Yiping Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China.
| |
Collapse
|
16
|
Ma A, Zhang Y, Chen J, Sun L, Hong G. Differential expression of osteoblast-like cells on self-organized titanium dioxide nanotubes. J Dent Sci 2024; 19:S26-S37. [PMID: 39807437 PMCID: PMC11725137 DOI: 10.1016/j.jds.2024.09.001] [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: 07/12/2024] [Revised: 09/02/2024] [Indexed: 01/16/2025] Open
Abstract
Background/purpose Titanium dioxide nanotube (TNT) structures have been shown to enhance the early osseointegration of dental implants. Nevertheless, the optimal nanotube diameter for promoting osteogenesis remains unclear due to variations in cell types and manufacture of nanotubes. This study aimed to evaluate the differences in MC3T3-E1 and Saos-2 cells behavior on nanotubes of varying diameters. Materials and methods TNT structures were fabricated by anodizing titanium foil at voltages ranging from 15V to 70V and annealed at 450 °C. Surface morphology and wettability were characterized using field emission scanning electron microscopy and water contact angle measurements, respectively. MC3T3-E1 and Saos-2 cells were cultured to evaluate biocompatibility. Early cell morphology and adhesion were visualized by scanning electron microscopy. Cell proliferation was quantified using CCK-8 assays, and differentiation was assessed through alkaline phosphatase assays. Osteogenesis-related gene expression was analyzed by real-time polymerase chain reaction (PCR), measuring runt-related transcription factor 2 (Runx-2), alkaline phosphatase (ALP), collagen type 1 (COL-1), osteocalcin (OCN), and Osteopontin (OPN) gene levels. Results Our results found that Saos-2 cells may be more suitable for TNT-related studies compared to MC3T3-E1 cells. Notably, the 65V nanotube group, with a diameter of 135.9 ± 15.83 nm, demonstrated the most significant osteogenic effect in our assays. Conclusion We propose that the use and screening of multiple cell lines prior to the evaluation of biomaterials can lead to more accurate in vitro experiments, thereby enhancing the reliability of biomaterial research.
Collapse
Affiliation(s)
- Aobo Ma
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yiding Zhang
- Department of Periodontology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Junduo Chen
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Lu Sun
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
- Department of Prosthodontics, College of Stomatology, Dalian Medical University, Dalian, China
| | - Guang Hong
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
- Department of Prosthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
17
|
Bayram C, Ozturk S, Karaosmanoglu B, Gultekinoglu M, Taskiran EZ, Ulubayram K, Majd H, Ahmed J, Edirisinghe M. Microfluidic Fabrication of Gelatin-Nano Hydroxyapatite Scaffolds for Enhanced Control of Pore Size Distribution and Osteogenic Differentiation of Dental Pulp Stem Cells. Macromol Biosci 2024; 24:e2400279. [PMID: 39388643 DOI: 10.1002/mabi.202400279] [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: 06/13/2024] [Revised: 09/13/2024] [Indexed: 10/12/2024]
Abstract
The combination of gelatin and hydroxyapatite (HA) has emerged as a promising strategy in dental tissue engineering due to its favorable biocompatibility, mechanical properties, and ability to support cellular activities essential for tissue regeneration, rendering them ideal components for hard tissue applications. Besides, precise control over interconnecting porosity is of paramount importance for tissue engineering materials. Conventional methods for creating porous scaffolds frequently encounter difficulties in regulating pore size distribution. This study demonstrates the fabrication of gelatin-nano HA scaffolds with uniform porosity using a T-type junction microfluidic device in a single-step process. Significant improvements in control over the pore size distribution are achieved by regulating the flow parameters, resulting in effective and time-efficient manufacturing comparable in quality to the innovative 3D bioprinting techniques. The overall porosity of the scaffolds exceeded 60%, with a remarkably narrow size distribution. The incorporation of nano-HAinto 3D porous gelatin scaffolds successfully induced osteogenic differentiation in stem cells at both the protein and gene levels, as evidenced by the significant increase in osteocalcin (OCN), an important marker of osteogenic differentiation. The OCN levels are 26 and 43 times higher for gelatin and gelatin-HA scaffolds, respectively, compared to the control group.
Collapse
Affiliation(s)
- Cem Bayram
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, 06800, Turkey
| | - Sukru Ozturk
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, 06100, Turkey
| | - Beren Karaosmanoglu
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Merve Gultekinoglu
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, 06800, Turkey
| | - Ekim Z Taskiran
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Kezban Ulubayram
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, 06100, Turkey
| | - Hamta Majd
- Department of Mechanical Engineering, University College London (UCL), London, WC1E7JE, UK
| | - Jubair Ahmed
- Department of Mechanical Engineering, University College London (UCL), London, WC1E7JE, UK
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London (UCL), London, WC1E7JE, UK
| |
Collapse
|
18
|
Zhou Z, Jin Z, Tian Y, Huangfu C, Fan Z, Liu D. CDK14 is regulated by IGF2BP2 and involved in osteogenic differentiation via Wnt/β-catenin signaling pathway in vitro. Life Sci 2024; 358:123148. [PMID: 39447733 DOI: 10.1016/j.lfs.2024.123148] [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: 06/18/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
AIMS Cyclin-dependent kinase (CDK) family proteins involve in various cellular processes via regulating the cell cycle; however, their expression during osteogenic differentiation and postmenopausal osteoporosis remains poorly understood. MAIN METHODS Using bioinformatics, we screened for CDK14 bound to Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) and explored its expression in vitro with time-gradient model and in a mouse model of postmenopausal osteoporosis, building on prior research. Subsequently, we investigated its effect on osteoblast proliferation, cell cycle dynamics, and osteogenic differentiation by administering CDK14 siRNA and the covalent inhibitor FMF-04-159-2. Furthermore, we examined the interaction between IGF2BP2 and CDK14. Finally, we validated the regulatory role of CDK14 on the Wnt/β-catenin pathway. KEY FINDINGS Our findings demonstrate a time-dependent CDK14 expression patterns during osteogenic differentiation of MC3T3-E1 cell line, with an initial increase followed by gradual decline over time. Notably, CDK14 expression exhibited significant reduction in bone tissue of postmenopausal osteoporosis mouse model. CDK14 inhibition altered osteoblast cell cycle dynamics, significantly reduced cellular proliferation capacity, and impaired osteogenic differentiation ability. IGF2BP2 interacted with CDK14 mRNA, and stabilizing mRNA's structure and inhibiting its degradation. Additionally, CDK14 facilitated Low-density lipoprotein receptor-related protein 6 (LRP6) and Glycogen synthase kinase 3β (GSK3β) phosphorylation, thus regulating β-catenin levels. SIGNIFICANCE These findings provide further insight into the molecular mechanisms governing osteoblast proliferation, differentiation and osteoporosis.
Collapse
Affiliation(s)
- Zimo Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Zhuoru Jin
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Yicheng Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Chenghao Huangfu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Zheng Fan
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Da Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
19
|
Wang J, Li Q, Li H, Liu X, Hu Y, Bai Y, Yang K. A novel RUNX2 splice site mutation in Chinese associated with cleidocranial dysplasia. Heliyon 2024; 10:e40277. [PMID: 39584128 PMCID: PMC11585700 DOI: 10.1016/j.heliyon.2024.e40277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
Pathogenic genes in most patients with cleidocranial dysplasia have been confirmed to be runt-related transcription factor 2 (RUNX2), which controls mutations in specific osteoblast transcription factors and affects skull ossification and suture adhesion. This study aimed to explore the role of RUNX2 mutations. Here, we report a rare case of a splice site mutation in a Chinese population with typical cleidocranial dysplasia symptoms, cranial suture insufficiency, clavicle dysplasia, and dental anomalies. Peripheral blood samples from the proband and her mother were subjected to Sanger sequencing. The expression levels of RUNX2 before and after mutation were verified using digital PCR (dPCR). The results revealed a classic mutation at the fifth base of the intron 5 initiation splicing sequence (NM001024630.4: C.685+5G > A). The mutation rate in the proband was 53 %, while the mother did not have any mutations. The secondary RNA structure of the RUNX2 gene in the progenitor was predicted to change, and the structural free energy was low in the wild-type, with the stem folded first and the structure being relatively stable. After the mutation, the free energy increased. This finding enriches the RUNX2 mutation library of CD-related genes in Chinese individuals.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, No.4 Tiantan Xili, Dong cheng District, Beijing, 100050, China
| | - Qiuying Li
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, No.4 Tiantan Xili, Dong cheng District, Beijing, 100050, China
| | - Hongyu Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Hu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, No.4 Tiantan Xili, Dong cheng District, Beijing, 100050, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, No.4 Tiantan Xili, Dong cheng District, Beijing, 100050, China
| |
Collapse
|
20
|
Song J, Zhang Y, Jin X, Zhu Y, Li Y, Hu M. Eucommia ulmoides Oliver polysaccharide alleviates glucocorticoid-induced osteoporosis by stimulating bone formation via ERK/BMP-2/SMAD signaling. Sci Rep 2024; 14:29647. [PMID: 39609585 PMCID: PMC11604974 DOI: 10.1038/s41598-024-80859-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Osteoporosis (OP) is a metabolic disease characterized by low bone mineral mass owing to osteoblast dysfunction. Eucommia ulmoides Oliver (EuO) is a Chinese herbal medicine traditionally used to treat OP. Here, a polysaccharide purified from the EuO cortex (EuOCP3) was administered to OP mice constructed with dexamethasone (Dex) to investigate its anti-OP activity. EuOCP3 significantly improved Dex-induced bone microarchitecture destruction, increased osteoblast numbers and surface, and stimulated an increase in the expression of osteoblast differentiation markers in the femurs of OP mice. Furthermore, EuOCP3 was applied to MC3T3-E1 cells to further explore its effects on osteoblast differentiation. EuOCP3 significantly promoted osteoblast differentiation and increased the level of phosphorylated extracellular signal-regulated kinase1/2 (ERK1/2) and SMAD1/5/8. The EuOCP3-mediated enhancement of osteoblast differentiation-related proteins and phosphorylated SMAD1/5/8 expression levels was strongly suppressed by an ERK inhibitor (PD98059), which confirmed the critical role of ERK signaling in EuOCP3-induced osteoblast differentiation. In summary, EuOCP3 can stimulate bone formation by improving osteoblast differentiation via ERK/BMP-2/SMAD signaling, indicating the potential use of EuOCP3 as a functional ingredient in food products for anti-OP treatment.
Collapse
Affiliation(s)
- Jiyu Song
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yongfeng Zhang
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xinghui Jin
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yutong Li
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
21
|
Wang Y, Duan H, Zhang Z, Chen L, Li J. Research Progress on the Application of Natural Medicines in Biomaterial Coatings. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5607. [PMID: 39597430 PMCID: PMC11595593 DOI: 10.3390/ma17225607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
With the continuous progress of biomedical technology, biomaterial coatings play an important role in improving the performance of medical devices and promoting tissue repair and regeneration. The application of natural medicine to biological materials has become a hot topic due to its diverse biological activity, low toxicity, and wide range of sources. This article introduces the definition and classification of natural medicines, lists some common natural medicines, such as curcumin, allicin, chitosan, tea polyphenols, etc., and lists some biological activities of some common natural medicines, such as antibacterial, antioxidant, antitumor, and other properties. According to the different characteristics of natural medicines, physical adsorption, chemical grafting, layer-by-layer self-assembly, sol-gel and other methods are combined with biomaterials, which can be used for orthopedic implants, cardiovascular and cerebrovascular stents, wound dressings, drug delivery systems, etc., to exert their biological activity. For example, improving antibacterial properties, promoting tissue regeneration, and improving biocompatibility promote the development of medical health. Although the development of biomaterials has been greatly expanded, it still faces some major challenges, such as whether the combination between the coating and the substrate is firm, whether the drug load is released sustainably, whether the dynamic balance will be disrupted, and so on; a series of problems affects the application of natural drugs in biomaterial coatings. In view of these problems, this paper summarizes some suggestions by evaluating the literature, such as optimizing the binding method and release system; carrying out more clinical application research; carrying out multidisciplinary cooperation; broadening the application of natural medicine in biomaterial coatings; and developing safer, more effective and multi-functional natural medicine coatings through continuous research and innovation, so as to contribute to the development of the biomedical field.
Collapse
Affiliation(s)
| | | | | | - Lan Chen
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (Y.W.); (H.D.); (Z.Z.)
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (Y.W.); (H.D.); (Z.Z.)
| |
Collapse
|
22
|
Chen Z, Jiang M, Mo L, Zhou C, Huang H, Ma C, Wang Z, Fan Y, Chen Z, Fang B, Liu Y. A natural agent, 5-deoxycajanin, mitigates estrogen-deficiency bone loss via modulating osteoclast-osteoblast homeostasis. Int Immunopharmacol 2024; 141:112906. [PMID: 39173403 DOI: 10.1016/j.intimp.2024.112906] [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: 06/05/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Hyperactive osteoclasts and hypoactive osteoblasts usually result in osteolytic conditions such as estrogen-deficiency bone loss. Few natural compounds that both attenuating bone resorption and enhancing bone formation could exert effects on this imbalance. 5-Deoxycajanin (5-D), an isoflavonoid extracted from Cajan leaf with estrogen-like properties, were found to have beneficial pharmacological effects on rebalancing the activities of osteoclasts and osteoblasts. This study revealed that 5-D at the same concentration could inhibit osteoclastogenesis of BMMs and promoted osteoblast differentiation of BMSCs. 5-D not only attenuated the fluorescent formation of RANKL-induced F-actin belts and NFATc1, but also activated ALP and RUNX2 expressions. As to downstream factor expressions, 5-D could block osteoclast-specific genes and proteins including NFATc1 and CTSK, while increased osteogenic genes and proteins including OPG and OCN, as confirmed by Real-time PCR and Western Blotting. Additionally, the network pharmacology and molecular docking identified the involvement of 5-D in the MIF and MAPK signaling pathways and the stable binding between 5-D and MAPK2K1. Further Western blot studies showed that 5-D decreased the phosphorylation of p38 and ERK in osteoclasts, but promoted these phosphorylations in osteoblasts. In a female C57BL/6J mouse model of estrogen deficiency-induced bone loss, 5-D demonstrated efficacy in enhancing BMD through attenuating osteoclast activities and promoting osteogenesis. These results underscore the potential application of 5-D on treating osteolysis resulting from hyperactive osteoclasts and hypoactive osteoblasts, shedding light on modulating osteoclast-osteoblast homeostasis.
Collapse
Affiliation(s)
- Zhiwen Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengyu Jiang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liang Mo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chi Zhou
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haoran Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Yangjiang Hospital of Traditional Chinese Medicine, Yangjiang, China
| | - Chao Ma
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhangzheng Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinuo Fan
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenqiu Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Bin Fang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yuhao Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
23
|
Chotipinit T, Supronsinchai W, Chantarangsu S, Suttamanatwong S. Healing effect of curcumin on tooth extraction sockets in diabetic rats. J Appl Oral Sci 2024; 32:e20240251. [PMID: 39570178 PMCID: PMC11643075 DOI: 10.1590/1678-7757-2024-0251] [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: 06/10/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 11/22/2024] Open
Abstract
OBJECTIVE Diabetes mellitus (DM) delays wound healing, including those following tooth extractions. Curcumin (CCM) can promote soft tissue and bone healing. The present study investigates the healing effects of CCM on tooth extraction sockets in diabetic rats. METHODOLOGY Ninety-six male Wistar rats were divided into the following four groups: Control+Corn Oil (CO), Control+CCM, DM+CO, and DM+CCM. Each group was subdivided into 7-, 14-, and 28-day time point subgroups comprising eight rats. All animals had their maxillary first molars extracted. CCM-treated rats received 100 mg/kg of CCM orally for 7, 14, and 28 days. The lesion area was evaluated using macroscopic analyses, whereas socket healing was assessed by hematoxylin and eosin staining. Keratinocyte growth factor (KGF), Runt-related transcription factor 2 (Runx2), and collagen type I (COL1) expression levels were obtained using quantitative polymerase chain reaction (qPCR). Bone healing was analyzed by means of microcomputed tomography (μCT). RESULTS After 7 days, the groups showed no significant differences in lesion area and by day 14, no lesions were present. CCM treatment increased KGF mRNA expression in diabetic rats; however, diabetic rats showed delayed bone healing unrelated to CCM. CCM treatment resulted in increased Runx2 mRNA expression only in control rats, whereas COL1 mRNA expression remained unaffected by CCM. CONCLUSION CCM shows potential as a soft tissue healing enhancer in diabetic rats and could serve as an additional treatment to promote soft tissue repair in diabetic individuals. Although CCM did not impact alveolar bone healing, it may enhance bone healing in other skeleton regions.
Collapse
Affiliation(s)
- Tipthanan Chotipinit
- Chulalongkorn UniversityFaculty of Graduate SchoolBangkokThailandChulalongkorn University, Faculty of Graduate School, Interdisciplinary Program of Physiology, Bangkok, Thailand.
| | - Weera Supronsinchai
- Chulalongkorn UniversityFaculty of DentistryDepartment of PhysiologyBangkokThailandChulalongkorn University, Faculty of Dentistry, Department of Physiology, Bangkok, Thailand.
| | - Soranun Chantarangsu
- Chulalongkorn UniversityFaculty of DentistryDepartment of Oral PathologyBangkokThailandChulalongkorn University, Faculty of Dentistry, Department of Oral Pathology, Bangkok, Thailand.
| | - Supaporn Suttamanatwong
- Chulalongkorn UniversityFaculty of DentistryDepartment of PhysiologyBangkokThailandChulalongkorn University, Faculty of Dentistry, Department of Physiology, Bangkok, Thailand.
| |
Collapse
|
24
|
Pinho LC, Queirós JA, Santos C, Colaço B, Fernandes MH. Biomimetic In Vitro Model of Canine Periodontal Ligament. Int J Mol Sci 2024; 25:12234. [PMID: 39596299 PMCID: PMC11594677 DOI: 10.3390/ijms252212234] [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: 10/11/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Periodontal disease affects about 80% of dogs, highlighting the importance of addressing periodontitis in veterinary dental care. The periodontal ligament (PDL) is a key structure holding the potential to regenerate the entire periodontal complex. This work presents an in vitro model of canine PDL-derived cell cultures that mimic the PDL's regenerative capacity for both mineralised and soft tissues. Explant outgrowth-derived PDL cells were cultured under standard conditions in osteoinductive medium and with hydroxyapatite nanoparticles (Hap NPs). Cell behaviour was assessed for viability/proliferation, morphology, growth patterns, and the expression of osteogenic and periodontal markers. Osteogenic conditions, either achieved with osteoinducers or an osteoconductive biomaterial, strongly promoted PDL-derived cells' commitment towards the osteogenic phenotype and significantly increased the expression of periodontal markers. These findings suggest that cultured PDL cells replicate the biological function of the PDL, supporting the regeneration of both soft and hard periodontal tissues under normal and demanding healing conditions. This in vitro model will offer a platform for testing new regenerative treatments and materials, ultimately contributing to canine dental care and better outcomes.
Collapse
Affiliation(s)
- Laura C. Pinho
- BoneLab—Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U.Porto, 4200-393 Porto, Portugal;
- LAQV/REQUIMTE, Faculty of Dental Medicine, U.Porto, 4200-393 Porto, Portugal
- CITAB—Centre for Research and Technology of Agro-Environmental and Biological Sciences, Inov4Agro, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal;
- CQE, IMS, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal;
- Instituto Politécnico de Setúbal, EST Setúbal, 2910-761 Setúbal, Portugal
| | - José André Queirós
- Hospital Veterinário Universitário de Paredes, 4580-593 Paredes, Portugal;
| | - Catarina Santos
- CQE, IMS, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal;
- Instituto Politécnico de Setúbal, EST Setúbal, 2910-761 Setúbal, Portugal
| | - Bruno Colaço
- CITAB—Centre for Research and Technology of Agro-Environmental and Biological Sciences, Inov4Agro, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal;
- CECAV–Animal and Veterinary Research Centre, University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Maria Helena Fernandes
- BoneLab—Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U.Porto, 4200-393 Porto, Portugal;
- LAQV/REQUIMTE, Faculty of Dental Medicine, U.Porto, 4200-393 Porto, Portugal
| |
Collapse
|
25
|
Chouaib B, Desoutter A, Cuisinier F, Collart-Dutilleul PY. Dental Pulp Stem Cell Conditioned Medium Enhance Osteoblastic Differentiation and Bone Regeneration. Stem Cell Rev Rep 2024:10.1007/s12015-024-10823-2. [PMID: 39514179 DOI: 10.1007/s12015-024-10823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Cell-free approaches, utilizing mesenchymal stem cell secretome, have promising prospects in various fields of regenerative medicine. In this study, we examined in vitro and in vivo the potential of dental pulp stem cell-conditioned medium (DPSC-CM) for bone regeneration. METHODS The secretome of undifferentiated stem cells from dental pulp were collected, and the effects of this DPSC-CM were assessed for osteodifferentiation of osteoblast-like cells (MG-63) and osteoblasts deriving from DPSC. Cell proliferation, alkaline phosphatase (ALP) activity, gene expression of Runt-related transcription factor 2 (Runx2), Bone Sialoprotein (BSP), Osteocalcin (OCN), and extracellular matrix mineralization were evaluated. The rat caudal vertebrae critical size defect model was to investigate the effect of DPSC-CM in vivo. RESULTS Results showed that DPSC-CM induced cell growth, and increased ALP activity and the expression of key marker genes at an early stage of osteoblastic differentiation compared to control. A rat bone defect model was used to illustrate the effect of DPSC-CM in vivo. The bone density within the defects were improved using conditioned medium, even though there was no significant difference between the control and DPSC-CM groups. The analysis of DPSC-CM by human growth factor antibody array revealed the presence of several factors involved in osteogenesis. CONCLUSION Taken together, these findings indicate that DPSC-CM is a promising therapeutic candidate for bone regenerative therapy, accelerating the maturation of osteoblastic cells. And even though safety and efficiency of DPSC-CM have to be confirmed in preclinical studies, these results represent a first step toward clinical application.
Collapse
Affiliation(s)
| | | | - Frédéric Cuisinier
- LBN, Univ. Montpellier, Montpellier, France
- Faculty of Dentistry, Univ. Montpellier, Montpellier, France
- Service Odontologie, CHU de Montpellier, Montpellier, France
| | - Pierre-Yves Collart-Dutilleul
- LBN, Univ. Montpellier, Montpellier, France.
- Faculty of Dentistry, Univ. Montpellier, Montpellier, France.
- Service Odontologie, CHU de Montpellier, Montpellier, France.
| |
Collapse
|
26
|
Hu J, Song Y, Zhang Y, Yang P, Chen S, Wu Z, Zhang J. Catalpol Enhances Osteogenic Differentiation of Human Periodontal Stem Cells and Modulates Periodontal Tissue Remodeling in an Orthodontic Tooth Movement Rat Model. Drug Des Devel Ther 2024; 18:4943-4960. [PMID: 39525045 PMCID: PMC11546164 DOI: 10.2147/dddt.s482969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose This study examines the effects and mechanisms of catalpol (CAT) on the proliferation and osteogenic differentiation of cultured human periodontal ligament stem cells (hPDLSCs) in vitro and assesses the impact of CAT on periodontal remodeling in vivo using an orthodontic tooth movement (OTM) model in rats. Methods hPDLSCs were cultured in a laboratory setting, and their proliferation and osteogenic differentiation were assessed using the Cell-counting Kit-8 (CCK-8), Alizarin Red Staining (ARS), quantitative calcium assay, alkaline phosphatase (ALP) staining and activity assay, and immunofluorescence assay. Additionally, the expression of collagen type 1 (COL-1), ALP, and runt-related transcription factor-2 (RUNX-2) was evaluated through qRT-PCR and Western blot analysis. To verify the function of the estrogen receptor-α (ER-α)-mediated phosphatidylinositol-3-kinase-protein kinase B (PI3K/AKT) pathway in this mechanism, LY294002 (a PI3K signaling pathway inhibitor) and the ER-α specific inhibitor methyl-piperidine-pyrazole (MPP) were used. The osteogenic markers ER-α, AKT, and p-AKT (phosphoprotein kinase B) were identified through Western blot analysis. Eighteen male Sprague-Dawley rats were assigned to two groups randomly: a CAT group receiving CAT and a control group receiving an equivalent volume of saline. Micro-computed tomography (micro-CT) analysis was employed to evaluate tooth movement and changes in alveolar bone structure. Morphological changes in the periodontal tissues between the roots were investigated using hematoxylin and eosin (HE) staining and tartaric-resistant acid phosphatase (TRAP) staining. The expression of COL-1, RUNX-2, and nuclear factor-κB (NF-κB) ligand (RANKL) was assessed through immunohistochemical staining (IHC) to evaluate periodontal tissue remodeling. Tests were analyzed using GraphPad Prism 8 software. Differences among more than two groups were analyzed by one-way or two-way analysis of variance (ANOVA) followed by the Tukey's test. Values of p < 0.05 were regarded as statistically significant. Results In vitro experiments demonstrated that 10 μM CAT significantly promoted the proliferation, ALP activity, and calcium nodule formation of hPDLSCs, with a notable increase in the expression of COL-1, ALP, RUNX-2, ER-α, and p-AKT. The PI3K/AKT pathway was inhibited by LY294002, and further analysis using MPP suggested that ER-α mediated this effect. In vivo, experiments indicated that CAT enhanced the expression of COL-1 and RUNX-2 on the tension side of rat tooth roots, reduced the number of osteoclasts on the compression side, inhibited RANKL expression, and suppressed OTM. Conclusion CAT can promote hPDLSCs proliferation and osteogenic differentiation in vitro through the ER-α/PI3K/AKT pathway and enhance periodontal tissue remodeling in vivo using OTM models. These findings suggest the potential for the clinical application of catalpol in preventing relapse following OTM.
Collapse
Affiliation(s)
- Jing Hu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Yang Song
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Yuxing Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Peng Yang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Siyu Chen
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Zhaoyan Wu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| | - Jun Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
27
|
Menger MM, Speicher R, Hans S, Histing T, El Kayali MKD, Ehnert S, Menger MD, Ampofo E, Wrublewsky S, Laschke MW. Nlrp3 Deficiency Does Not Substantially Affect Femoral Fracture Healing in Mice. Int J Mol Sci 2024; 25:11788. [PMID: 39519338 PMCID: PMC11546854 DOI: 10.3390/ijms252111788] [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: 09/29/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammation has been recognized as major factor for successful bone regeneration. On the other hand, a prolonged or overshooting inflammatory response can also cause fracture healing failure. The nucleotide-binding oligomerization domain (NOD)-like receptor protein (NLRP)3 inflammasome plays a crucial role in inflammatory cytokine production. However, its role during fracture repair remains elusive. We investigated the effects of Nlrp3 deficiency on the healing of closed femoral fractures in Nlrp3-/- and wildtype mice. The callus tissue was analyzed by means of X-ray, biomechanics, µCT and histology, as well as immunohistochemistry and Western blotting at 2 and 5 weeks after surgery. We found a significantly reduced trabecular thickness at 2 weeks after fracture in the Nlrp3-/- mice when compared to the wildtype animals. However, the amount of bone tissue did not differ between the two groups. Additional immunohistochemical analyses showed a reduced number of CD68-positive macrophages within the callus tissue of the Nlrp3-/- mice at 2 weeks after fracture, whereas the number of myeloperoxidase (MPO)-positive granulocytes was increased. Moreover, we detected a significantly lower expression of vascular endothelial growth factor (VEGF) and a reduced number of microvessels in the Nlrp3-/- mice. The expression of the absent in melanoma (AIM)2 inflammasome was increased more than 2-fold in the Nlrp3-/- mice, whereas the expression of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 was not affected. Our results demonstrate that Nlrp3 deficiency does not markedly affect femoral fracture healing in mice. This is most likely due to the unaltered expression of pro-inflammatory cytokines and pro-osteogenic growth factors.
Collapse
Affiliation(s)
- Maximilian M. Menger
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany; (T.H.)
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Rouven Speicher
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Sandra Hans
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany; (T.H.)
| | - Moses K. D. El Kayali
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany;
| | - Michael D. Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (R.S.); (S.H.); (M.K.D.E.K.); (M.D.M.); (E.A.); (S.W.); (M.W.L.)
| |
Collapse
|
28
|
Liu Y, Wei X, Yang T, Wang X, Li T, Sun M, Jiao K, Jia W, Yang Y, Yan Y, Wang S, Wang C, Liu L, Dai Z, Jiang Z, Jiang X, Li C, Liu G, Cheng Z, Luo Y. Hyaluronic acid methacrylate/Pluronic F127 hydrogel enhanced with spermidine-modified mesoporous polydopamine nanoparticles for efficient synergistic periodontitis treatment. Int J Biol Macromol 2024; 281:136085. [PMID: 39353520 DOI: 10.1016/j.ijbiomac.2024.136085] [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: 04/30/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Bacterial infection, reactive oxygen species (ROS) accumulation, and persistent inflammation pose significant challenges in the treatment of periodontitis. However, the current single-modal strategy makes achieving the best treatment effect difficult. Herein, we developed a double-network hydrogel composed of Pluronic F127 (PF-127) and hyaluronic acid methacrylate (HAMA) loaded with spermidine-modified mesoporous polydopamine nanoparticles (M@S NPs). The PF-127/HAMA/M@S (PH/M@S) hydrogel was injectable and exhibited thermosensitivity and photocrosslinking capabilities, which enable it to adapt to the irregular shape of periodontal pockets. In vitro, the PH/M@S displayed multiple therapeutic effects, such as photothermal antibacterial activity, a high ROS scavenging capacity, and anti-inflammatory effects, which are beneficial for the multimodal treatment of periodontitis. The underlying anti-inflammatory mechanism of this hydrogel involves suppression of the extracellular regulated protein kinase 1/2 and nuclear factor kappa-B signalling pathways. Furthermore, in lipopolysaccharide-stimulated macrophage conditioned media, the PH/M@S effectively restored the osteogenic differentiation potential. In a rat model of periodontitis, the PH/M@S effectively reduced the bacterial load, relieved local inflammation and inhibited alveolar bone resorption. Collectively, these findings highlight the versatile functions of the PH/M@S, including photothermal antibacterial activity, ROS scavenging, and anti-inflammatory effects, indicating that this hydrogel is a promising multifunctional filling material for the treatment of periodontitis.
Collapse
Affiliation(s)
- Yun Liu
- Stomatology Center of Jingyue Campus, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China
| | - Xue Wei
- Ultrasound Diagnostic Center (Doctor of excellence program), The First Hospital of Jilin University, Changchun 130021, China
| | - Tao Yang
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xi Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Ting Li
- Department of Gastroenterology, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130000, China
| | - Maolei Sun
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Stomatology, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Kun Jiao
- Stomatology Center of Jingyue Campus, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China
| | - Wenyuan Jia
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Orthopedics, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Yuheng Yang
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Orthopedics, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Yongzheng Yan
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Orthopedics, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Shaoru Wang
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chang Wang
- Stomatology Center of Jingyue Campus, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China
| | - Liping Liu
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zhihui Dai
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zhen Jiang
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xuanzuo Jiang
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Orthopedics, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Chiyu Li
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Orthopedics, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Guomin Liu
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; Department of Orthopedics, The Second Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Zhiqiang Cheng
- Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China; College of Resources and Environment, Jilin Agriculture University, Changchun 130118, China
| | - Yungang Luo
- Stomatology Center of Jingyue Campus, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Scientific and Technological Innovation Center of Health Products and Medical Materials with Characteristic Resources of Jilin Province, Jilin University, Changchun 130021, China; Jilin Provincial Engineering Laboratory of Bone Regeneration and Tissue Repair Materials, Jilin University, Changchun 130021, China; Jilin Provincial Joint University-Industry Innovation Laboratory for Oral Biomedical Materials, Jilin University, Changchun 130021, China.
| |
Collapse
|
29
|
Ou YC, Yu TM, Li JR, Wu CC, Wang JD, Liao SL, Chen WY, Kuan YH, Chen CJ. Runx2 silencing sensitized human renal cell carcinoma cells to ABT-737 apoptosis. Arch Biochem Biophys 2024; 761:110173. [PMID: 39369835 DOI: 10.1016/j.abb.2024.110173] [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: 06/26/2024] [Revised: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
The prognostic value of Runt-related transcription factor 2 (Runx2) and its involvement in cell growth and motility have been reported in patients diagnosed with renal cell carcinoma (RCC). Since Runx2 may have the potential to be a target for the purpose of antitumor intervention, there is an urgent need to gain insight into its oncogenic properties. Using human 786-O, Caki-1 and ACHN RCC cells as models, the silencing of cellular Runx2 expression brought about a reduction in cyclin D1 and β-catenin expression, cell growth and migration without any significant cell death. Runx2-silenced cells turned into apoptosis vulnerable in the presence of ABT-737, a BH3 mimetic Bcl-2 inhibitor. Data from biochemical and molecular studies have revealed a positive correlation between Runx2 expression and Akt phosphorylation, Mcl-1 expression, and fibronectin expression. Results of genetic silencing studies have indicated the potential involvement of Mcl-1 and fibronectin in the decision of RCC cell ABT-737 resistance and sensitivity. The regulatory roles of the PI3K/Akt axis in the expression of Mcl-1 and fibronectin were suggested by means of the results taken from experiments involving pharmacological study of the PI3K/Akt. Since overexpression and prognostic roles of Runx2, activated Akt, Mcl-1, fibronectin, cyclin D1, and β-catenin have been revealed in RCC, it is important to explore the precise mechanisms underlying Runx2 oncogenic effects. Although the linking details between Runx2 and PI3K/Akt have yet to be identified, our findings suggest that Mcl-1 and fibronectin are downstream effectors of Runx2 via a regulatory axis of the PI3K/Akt and their promotion of cell growth, migration, and ABT-737 resistance in RCC cells.
Collapse
Affiliation(s)
- Yen-Chuan Ou
- Department of Urology, Tungs' Taichung MetroHarbor Hospital, Taichung City, 433, Taiwan.
| | - Tung-Min Yu
- Division of Nephrology, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City, 407, Taiwan; Department of Financial Engineering, Providence University, Taichung City, 433, Taiwan; Department of Data Science and Big Data Analytics, Providence University, Taichung City, 433, Taiwan.
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, 407, Taiwan.
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan.
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung City, 402, Taiwan.
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, 407, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City, 404, Taiwan.
| |
Collapse
|
30
|
Castro‐Vázquez D, Arribas‐Castaño P, García‐López I, Gutiérrez‐Cañas I, Pérez‐García S, Lamana A, Villanueva‐Romero R, Cabrera‐Martín A, Tecza K, Martínez C, Juarranz Y, Gomariz RP, Carrión M. Vasoactive intestinal peptide exerts an osteoinductive effect in human mesenchymal stem cells. Biofactors 2024; 50:1148-1160. [PMID: 38733572 PMCID: PMC11627472 DOI: 10.1002/biof.2062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024]
Abstract
Several neuropeptides present in bone tissues, produced by nerve fibers and bone cells, have been reported to play a role in regulating the fine-tuning of osteoblast and osteoclast functions to maintain bone homeostasis. This study aims to characterize the influence of the neuropeptide vasoactive intestinal peptide (VIP) on the differentiation process of human mesenchymal stem cells (MSCs) into osteoblasts and on their anabolic function. We describe the mRNA and protein expression profile of VIP and its receptors in MSCs as they differentiate into osteoblasts, suggesting the presence of an autocrine signaling pathway in these cells. Our findings reveal that VIP enhances the expression of early osteoblast markers in MSCs under osteogenic differentiation and favors both bone matrix formation and proper cytoskeletal reorganization. Finally, our data suggest that VIP could be exerting a direct modulatory role on the osteoblast to osteoclast signaling by downregulating the receptor activator of nuclear factor-κB ligand/osteoprotegerin ratio. These results highlight the potential of VIP as an osteoinductive differentiation factor, emerging as a key molecule in the maintenance of human bone homeostasis.
Collapse
Affiliation(s)
- David Castro‐Vázquez
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Paula Arribas‐Castaño
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Iván García‐López
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Irene Gutiérrez‐Cañas
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Selene Pérez‐García
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Amalia Lamana
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Raúl Villanueva‐Romero
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Alicia Cabrera‐Martín
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Karolina Tecza
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Carmen Martínez
- Departmental Section of Cell Biology, Faculty of MedicineComplutense University of MadridMadridSpain
| | - Yasmina Juarranz
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Rosa P. Gomariz
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| | - Mar Carrión
- Department of Cell Biology, Faculty of Biological ScienceComplutense University of MadridMadridSpain
| |
Collapse
|
31
|
Arya PN, Saranya I, Selvamurugan N. RUNX2 regulation in osteoblast differentiation: A possible therapeutic function of the lncRNA and miRNA-mediated network. Differentiation 2024; 140:100803. [PMID: 39089986 DOI: 10.1016/j.diff.2024.100803] [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: 05/09/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Osteogenic differentiation is a crucial process in the formation of the skeleton and the remodeling of bones. It relies on a complex system of signaling pathways and transcription factors, including Runt-related transcription factor 2 (RUNX2). Non-coding RNAs (ncRNAs) control the bone-specific transcription factor RUNX2 through post-transcriptional mechanisms to regulate osteogenic differentiation. The most research has focused on microRNAs (miRNAs) and long ncRNAs (lncRNAs) in studying how they regulate RUNX2 for osteogenesis in both normal and pathological situations. This article provides a concise overview of the recent advancements in understanding the critical roles of lncRNA/miRNA/axes in controlling the expression of RUNX2 during bone formation. The possible application of miRNAs and lncRNAs as therapeutic agents for the treatment of disorders involving the bones and bones itself is also covered.
Collapse
Affiliation(s)
- Pakkath Narayanan Arya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Iyyappan Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
32
|
Qu Z, Zhao S, Zhang Y, Wang X, Yan L. Natural Compounds for Bone Remodeling: Targeting osteoblasts and relevant signaling pathways. Biomed Pharmacother 2024; 180:117490. [PMID: 39332184 DOI: 10.1016/j.biopha.2024.117490] [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: 06/23/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
In the process of bone metabolism and bone remodeling, bone marrow mesenchymal stem cells (BM-MSCs) differentiate into osteoblasts (OBs) under certain conditions to enable the formation of new bone, and normal bone reconstruction and pathological bone alteration are closely related to the differentiation and proliferation functions of OBs. Osteogenic differentiation of BM-MSCs involves multiple signaling pathways, which function individually but interconnect intricately to form a complex signaling regulatory network. Natural compounds have fewer adverse effects than chemically synthesized drugs, optimize bone health, and are more suitable for long-term use. In this paper, we focus on OBs, summarize the current research progress of signaling pathways related to OBs differentiation, and review the molecular mechanisms by which chemically synthesized drugs with potential anti-osteoporosis properties regulate OBs-mediated bone formation.
Collapse
Affiliation(s)
- Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Zhang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
33
|
Li B, Wang L, Du M, He H. FTO in oral diseases: Functions, mechanisms, and therapeutic potential. FASEB J 2024; 38:e70115. [PMID: 39436191 DOI: 10.1096/fj.202401406rr] [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: 06/20/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Fat mass and obesity-associated protein (FTO) is the first identified N6-methyladenosine (m6A) demethylase widely distributed in various tissues in adults and children. It plays an essential role in diverse mRNA-associated processes including transcriptional stability, selective splicing, mRNA translocation, and also protein translation. Recently, emerging studies have shown that FTO is involved in the genesis and development of oral diseases. However, the correlation between FTO and oral diseases and its specific regulatory mechanism still needs further study. In this review, we will summarize the discovery, distribution, gene expression, protein structure, biological functions, inhibitors, and quantifying methods of FTO, as well as its regulatory role and mechanism in oral diseases. Notably, FTO genetic variants are strongly associated with periodontal diseases (PDs), temporomandibular joint osteoarthritis (TMJOA), and obstructive sleep apnea (OSA). Besides, the latest studies that describe the relationship between FTO and PDs, head and neck squamous cell carcinoma (HNSCCs), TMJOA, and OSA will be discussed. We elaborate on the regulatory roles of FTO in PDs, HNSCCs, and TMJOA, which are modulated through cell proliferation, cell migration, apoptosis, bone metabolism, and immune response. The review will enrich our understanding of RNA epigenetic modifications in oral diseases and present a solid theoretical foundation for FTO to serve as a novel diagnosis and prognostic biomarker for oral diseases.
Collapse
Affiliation(s)
- Biao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Leilei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingyuan Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hong He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Oliveira BA, Levy D, Paz JL, de Freitas FA, Reichert CO, Rodrigues A, Bydlowski SP. 7-Ketocholesterol Effects on Osteogenic Differentiation of Adipose Tissue-Derived Mesenchymal Stem Cells. Int J Mol Sci 2024; 25:11380. [PMID: 39518932 PMCID: PMC11545361 DOI: 10.3390/ijms252111380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Some oxysterols were shown to promote osteogenic differentiation of mesenchymal stem cells (MSCs). Little is known about the effects of 7-ketocholesterol (7-KC) in this process. We describe its impact on human adipose tissue-derived MSC (ATMSC) osteogenic differentiation. ATMSCs were incubated with 7-KC in osteogenic or adipogenic media. Osteogenic and adipogenic differentiation was evaluated by Alizarin red and Oil Red O staining, respectively. Osteogenic (ALPL, RUNX2, BGLAP) and adipogenic markers (PPARƔ, C/EBPα) were determined by RT-PCR. Differentiation signaling pathways (SHh, Smo, Gli-3, β-catenin) were determined by indirect immunofluorescence. ATMSCs treated with 7-KC in osteogenic media stained positively for Alizarin Red. 7-KC in adipogenic media decreased the number of adipocytes. 7-KC increased ALPL and RUNX2 but not BGLAP expressions. 7-KC decreased expression of PPARƔ and C/EBPα, did not change SHh, Smo, and Gli-3 expression, and increased the expression of β-catenin. In conclusion, 7-KC favors osteogenic differentiation of ATMSCs through the expression of early osteogenic genes (matrix maturation phase) by activating the Wnt/β-catenin signaling pathway, while inhibiting adipogenic differentiation. This knowledge can be potentially useful in regenerative medicine, in treatments for bone diseases.
Collapse
Affiliation(s)
- Beatriz Araújo Oliveira
- Lipids, Oxidation, and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil (D.L.); (F.A.d.F.); (C.O.R.)
| | - Débora Levy
- Lipids, Oxidation, and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil (D.L.); (F.A.d.F.); (C.O.R.)
| | - Jessica Liliane Paz
- Lipids, Oxidation, and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil (D.L.); (F.A.d.F.); (C.O.R.)
| | - Fabio Alessandro de Freitas
- Lipids, Oxidation, and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil (D.L.); (F.A.d.F.); (C.O.R.)
| | - Cadiele Oliana Reichert
- Lipids, Oxidation, and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil (D.L.); (F.A.d.F.); (C.O.R.)
| | - Alessandro Rodrigues
- Department of Earth and Exact Sciences, Universidade Federal de Sao Paulo, Diadema 09972-270, SP, Brazil;
| | - Sérgio Paulo Bydlowski
- Lipids, Oxidation, and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil (D.L.); (F.A.d.F.); (C.O.R.)
- National Institute of Science and Technology for Regenerative Medicine (INCT Regenera), National Council for Scientific and Technological Development (CNPq), Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
35
|
Li B, Wu Y, Ying L, Zhu W, Yang J, Zhou L, Yi L, Jiang T, Jiang H, Song X, Xue W, Liang G, Huang S, Song Z. Synthesis and Antiosteoporotic Characterization of Diselenyl Maleimides: Discovery of a Potent Agent for the Treatment of Osteoporosis by Targeting RANKL. J Med Chem 2024; 67:17226-17242. [PMID: 39299698 DOI: 10.1021/acs.jmedchem.4c01105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
To discover new osteoclast-targeting antiosteoporosis agents, we identified forty-six diselenyl maleimides, which were efficiently prepared using a novel, simple, and metal-free method at room temperature in a short reaction time. Among them, 3k showed the most marked inhibition of osteoclast differentiation with an IC50 value of 0.36 ± 0.03 μM. Moreover, 3k significantly suppressed RANKL-induced osteoclast formation, bone resorption, and osteoclast-specific genes expression in vitro. Mechanistic studies revealed that 3k remarkably blocked the RANKL-induced mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways. In ovariectomized mice, intragastric administration of 3k significantly alleviated bone loss, exhibiting an effect similar to that of alendronate. Surface plasmon resonance assay and microscale thermophoresis assay results suggested that RANKL might be a potential molecular target for 3k. Collectively, the findings presented above provided a novel candidate for further development of bone antiresorptive drugs that target RANKL.
Collapse
Affiliation(s)
- Bin Li
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Yao Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Linkun Ying
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weiwei Zhu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jingyi Yang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Lingling Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Lele Yi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Tianle Jiang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Haofu Jiang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Xiangrui Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Guang Liang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- School of Pharmacy, Hangzhou Medical College, Hangzhou 311399, Zhejiang, China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| |
Collapse
|
36
|
Yang Y, Yang DC, Long XY, Liu X, Lu JW, Zhang ZJ, Shi QQ, Zhou Y, Zou DH. Bioinspired triple-layered membranes for periodontal guided bone regeneration applications. J Mater Chem B 2024; 12:9938-9946. [PMID: 39267586 DOI: 10.1039/d4tb01658k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Barrier membranes have been used for the treatment of alveolar bone loss caused by periodontal diseases or trauma. However, an optimal barrier membrane must satisfy multiple requirements simultaneously, which are challenging to combine into a single material. We herein report the design of a bioinspired membrane consisting of three functional layers. The primary layer is composed of clay nanosheets and chitin, which form a nacre-inspired laminated structure. A calcium phosphate mineral layer is deposited on the inner surface of the nacre-inspired layer, while a poly(lactic acid) layer is coated on the outer surface. The composite membrane integrates good mechanical strength and deformability because of the nacre-inspired structure, facilitating operations during the implant surgery. The mineral layer induces the osteogenic differentiation of bone marrow mesenchymal stem cells and increases the stiffness of the membrane, which is an important factor for the regeneration process. The poly(lactic acid) layer can prevent unwanted mineralization on the outer surface of the membrane in oral environments. Cell experiments reveal that the membrane exhibits good biocompatibility and anti-infiltration capability toward connective tissue/epithelium cells. Furthermore, in vitro analyses show that the membrane does not degrade too fast, allowing enough time for bone regeneration. In vivo experiments prove that the membrane can effectively induce better bone regeneration and higher trabecular bone density in alveolar bone defects. This study demonstrates the potential of this bioinspired triple-layered membrane with hierarchical structures as a promising barrier material for periodontal guided tissue regeneration.
Collapse
Affiliation(s)
- Yang Yang
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
- Department of Periodontology, College & Hospital of Stomatology, Anhui Medical University, Hefei, 230032, China
| | - Deng-Cheng Yang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, China
| | - Xian-Yan Long
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Xiang Liu
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Jing-Wen Lu
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Zhou-Jing Zhang
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Qian-Qian Shi
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Yong Zhou
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
- Department of Dental Implantology, College & Hospital of Stomatology, Anhui Medical University, Hefei, 230032, China
| | - Duo-Hong Zou
- College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, China.
- Department of Oral Surgery, Shanghai Key Laboratory of Stomatology, School of Medicine, National Clinical Research Center of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200011, China
| |
Collapse
|
37
|
Gerdesmeyer L, Tübel J, Obermeier A, Harrasser N, Glowalla C, von Eisenhart-Rothe R, Burgkart R. Extracorporeal Magnetotransduction Therapy as a New Form of Electromagnetic Wave Therapy: From Gene Upregulation to Accelerated Matrix Mineralization in Bone Healing. Biomedicines 2024; 12:2269. [PMID: 39457582 PMCID: PMC11505246 DOI: 10.3390/biomedicines12102269] [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: 09/11/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Electromagnetic field therapy is gaining attention for its potential in treating bone disorders, with Extracorporeal Magnetotransduction Therapy (EMTT) emerging as an innovative approach. EMTT offers a higher oscillation frequency and magnetic field strength compared to traditional Pulsed Electromagnetic Field (PEMF) therapy, showing promise in enhancing fracture healing and non-union recovery. However, the mechanisms underlying these effects remain unclear. RESULTS This study demonstrates that EMTT significantly enhances osteoblast bone formation at multiple levels, from gene expression to extracellular matrix mineralization. Key osteoblastogenesis regulators, including SP7 and RUNX2, and bone-related genes such as COL1A1, ALPL, and BGLAP, were upregulated, with expression levels surpassing those of the control group by over sevenfold (p < 0.001). Enhanced collagen synthesis and mineralization were confirmed by von Kossa and Alizarin Red staining, indicating increased calcium and phosphate deposition. Additionally, calcium imaging revealed heightened calcium influx, suggesting a cellular mechanism for EMTT's osteogenic effects. Importantly, EMTT did not compromise cell viability, as confirmed by live/dead staining and WST-1 assays. CONCLUSION This study is the first to show that EMTT can enhance all phases of osteoblastogenesis and improve the production of critical mineralization components, offering potential clinical applications in accelerating fracture healing, treating osteonecrosis, and enhancing implant osseointegration.
Collapse
Affiliation(s)
- Lennart Gerdesmeyer
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Jutta Tübel
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Andreas Obermeier
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Norbert Harrasser
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- ECOM Excellent Center of Medicine, Arabellastraße 17, 81925 Munich, Germany
| | - Claudio Glowalla
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- BG Unfallklinik Murnau, Professor-Küntscher-Straße 8, 82418 Murnau am Staffelsee, Germany
| | - Rüdiger von Eisenhart-Rothe
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Rainer Burgkart
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
38
|
Roth DM, Piña JO, MacPherson M, Budden C, Graf D. Physiology and Clinical Manifestations of Pathologic Cranial Suture Widening. Cleft Palate Craniofac J 2024; 61:1750-1759. [PMID: 37271984 PMCID: PMC11468227 DOI: 10.1177/10556656231178438] [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: 06/06/2023] Open
Abstract
Cranial sutures are complex structures integrating mechanical forces with osteogenesis which are often affected in craniofacial syndromes. While premature fusion is frequently described, rare pathological widening of cranial sutures is a comparatively understudied phenomenon. This narrative review aims to bring to light the biologically variable underlying causes of widened sutures and persistent fontanelles leading to a common outcome. The authors herein present four syndromes, selected from a literature review, and their identified biological mechanisms in the context of altered suture physiology, exploring the roles of progenitor cell differentiation, extracellular matrix production, mineralization, and bone resorption. This article illustrates the gaps in understanding of complex craniofacial disorders, and the potential for further unification of genetics, cellular biology, and clinical pillars of health science research to improve treatment outcomes for patients.
Collapse
Affiliation(s)
- Daniela M. Roth
- School of Dentistry, University of Alberta, Edmonton, Canada
| | - Jeremie Oliver Piña
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | | | - Curtis Budden
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Daniel Graf
- School of Dentistry, University of Alberta, Edmonton, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| |
Collapse
|
39
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
40
|
Makwana P, Modi U, Dhimmar B, Vasita R. Design and development of in-vitro co-culture device for studying cellular crosstalk in varied tissue microenvironment. BIOMATERIALS ADVANCES 2024; 163:213952. [PMID: 38991495 DOI: 10.1016/j.bioadv.2024.213952] [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: 03/26/2024] [Revised: 06/16/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Despite of being in different microenvironment, breast cancer cells influence the bone cells and persuade cancer metastasis from breast to bone. Multiple co-culture approaches have been explored to study paracrine signaling between these cells and to study the progression of cancer. However, lack of native tissue microenvironment remains a major bottleneck in existing co-culture technologies. Therefore, in the present study, a tumorigenic and an osteogenic microenvironment have been sutured together to create a multi-cellular environment and has been appraised to study cancer progression in bone tissue. The PCL-polystyrene and PCL-collagen fibrous scaffolds were characterized for tumorigenic and osteogenic potential induction on MDA-MB-231 and MC3T3-E1 cells respectively. Diffusion ability of crystal violet, glucose, and bovine serum albumin across the membrane were used to access the potential paracrine interaction facilitated by device. While in co-cultured condition, MDA-MB-231 cells showed EMT phenotype along with secretion of TNFα and PTHrP which lower down the expression of osteogenic markers including alkaline phosphatase, RUNX2, Osteocalcin and Osteoprotegerin. The cancer progression in bone microenvironment demonstrated the role and necessity of creating multiple tissue microenvironment and its contribution in studying multicellular disease progression and therapeutics.
Collapse
Affiliation(s)
- Pooja Makwana
- Biomaterial and Biomimetic Laboratory, School of Life Sciences, Central University of Gujarat, India
| | - Unnati Modi
- Biomaterial and Biomimetic Laboratory, School of Life Sciences, Central University of Gujarat, India
| | - Bindiya Dhimmar
- Biomaterial and Biomimetic Laboratory, School of Life Sciences, Central University of Gujarat, India
| | - Rajesh Vasita
- Biomaterial and Biomimetic Laboratory, School of Life Sciences, Central University of Gujarat, India; Terasaki Institute of Biomedical Innovation, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Tan YL, Ju SH, Wang Q, Zhong R, Gao JH, Wang MJ, Kang YL, Xu MZ. Shuanglongjiegu pill promoted bone marrow mesenchymal stem cell osteogenic differentiation by regulating the miR-217/RUNX2 axis to activate Wnt/β-catenin pathway. J Orthop Surg Res 2024; 19:617. [PMID: 39350234 PMCID: PMC11443779 DOI: 10.1186/s13018-024-05085-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
This study aimed to investigate the effects of Shuanglongjiegu pill (SLJGP) on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and explore its mechanism based on miR-217/RUNX2 axis. Results found that drug-containing serum of SLJGP promoted BMSCs viability with a dose-dependent effect. Under osteogenic differentiation conditions, SLJGP promoted the expression of ALP, OPN, BMP2, RUNX2, and the osteogenic differentiation ability of BMSCs. In addition, SLJGP significantly reduced miR-217 expression, and miR-217 directly targeted RUNX2. After treatment with miR-217 mimic, the promoting effects of SLJGP on proliferation and osteogenic differentiation of BMSCs were significantly inhibited. MiR-217 mimic co-treated with pcDNA-RUNX2 further confirmed that the miR-217/RUNX2 axis was involved in SLJGP to promote osteogenic differentiation of BMSCs. In addition, analysis of Wnt/β-catenin pathway protein expression showed that SLJGP activated the Wnt/β-catenin pathway through miR-217/RUNX2. In conclusion, SLJGP promoted osteogenic differentiation of BMSCs by regulating miR-217/RUNX2 axis and activating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- You-Li Tan
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China.
| | - Shao-Hua Ju
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Qiang Wang
- Department of Rehabilitation of sports medicine, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Rui Zhong
- Department of Orthopedics, Affiliated Sports Hospital of Chengdu Sport University, Chengdu, 610041, China
| | - Ji-Hai Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ming-Jian Wang
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Ya-Lan Kang
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Meng-Zhang Xu
- Department of Neck, Shoulder, Waist, and Leg Pain, Sichuan Province Orthopedic Hospital, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Docshin P, Panshin D, Malashicheva A. Molecular Interplay in Cardiac Fibrosis: Exploring the Functions of RUNX2, BMP2, and Notch. Rev Cardiovasc Med 2024; 25:368. [PMID: 39484128 PMCID: PMC11522771 DOI: 10.31083/j.rcm2510368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac fibrosis, characterized by the excessive deposition of extracellular matrix proteins, significantly contributes to the morbidity and mortality associated with cardiovascular diseases. This article explores the complex interplay between Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2), and Notch signaling pathways in the pathogenesis of cardiac fibrosis. Each of these pathways plays a crucial role in the regulation of cellular functions and interactions that underpin fibrotic processes in the heart. Through a detailed review of current research, we highlight how the crosstalk among RUNX2, BMP2, and Notch not only facilitates our understanding of the fibrotic mechanisms but also points to potential biomolecular targets for intervention. This article delves into the regulatory networks, identifies key molecular mediators, and discusses the implications of these signaling pathways in cardiac structural remodeling. By synthesizing findings from recent studies, we provide insights into the cellular and molecular mechanisms that could guide future research directions, aiming to uncover new therapeutic strategies to manage and treat cardiac fibrosis effectively.
Collapse
Affiliation(s)
- Pavel Docshin
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| | - Daniil Panshin
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| |
Collapse
|
43
|
Pei Y, Liu F, Zhao Y, Lin H, Huang X. Role of hedgehog signaling in the pathogenesis and therapy of heterotopic ossification. Front Cell Dev Biol 2024; 12:1454058. [PMID: 39364140 PMCID: PMC11447292 DOI: 10.3389/fcell.2024.1454058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Heterotopic ossification (HO) is a pathological process that generates ectopic bone in soft tissues. Hedgehog signaling (Hh signaling) is a signaling pathway that plays an important role in embryonic development and involves three ligands: sonic hedgehog (Shh), Indian hedgehog (Ihh) and desert hedgehog (Dhh). Hh signaling also has an important role in skeletal development. This paper discusses the effects of Hh signaling on the process of HO formation and describes several signaling molecules that are involved in Hh-mediated processes: parathyroid Hormone-Related Protein (PTHrP) and Fkbp10 mediate the expression of Hh during chondrogenesic differentiation. Extracellular signal-regulated kinase (ERK), GNAs and Yes-Associated Protein (YAP) interact with Hh signaling to play a role in osteogenic differentiation. Runt-Related Transcription Factor 2 (Runx2), Mohawk gene (Mkx) and bone morphogenetic protein (BMP) mediate Hh signaling during both chondrogenic and osteogenic differentiation. This paper also discusses possible therapeutic options for HO, lists several Hh inhibitors and explores whether they could serve as emerging targets for the treatment of HO.
Collapse
Affiliation(s)
- Yiran Pei
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fangzhou Liu
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yike Zhao
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyan Huang
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
44
|
Freiberger RN, López CAM, Palma MB, Cevallos C, Sviercz FA, Jarmoluk P, García MN, Quarleri J, Delpino MV. HIV Modulates Osteoblast Differentiation via Upregulation of RANKL and Vitronectin. Pathogens 2024; 13:800. [PMID: 39338991 PMCID: PMC11435243 DOI: 10.3390/pathogens13090800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Bone loss is a prevalent characteristic among people with HIV (PWH). We focused on mesenchymal stem cells (MSCs) and osteoblasts, examining their susceptibility to different HIV strains (R5- and X4-tropic) and the subsequent effects on bone tissue homeostasis. Our findings suggest that MSCs and osteoblasts are susceptible to R5- and X4-tropic HIV but do not support productive HIV replication. HIV exposure during the osteoblast differentiation process revealed that the virus could not alter mineral and organic matrix deposition. However, the reduction in runt-related transcription factor 2 (RUNX2) transcription, the increase in the transcription of nuclear receptor activator ligand kappa B (RANKL), and the augmentation of vitronectin deposition strongly suggested that X4- and R5-HIV could affect bone homeostasis. This study highlights the HIV ability to alter MSCs' differentiation into osteoblasts, critical for maintaining bone and adipose tissue homeostasis and function.
Collapse
Affiliation(s)
- Rosa Nicole Freiberger
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Cynthia Alicia Marcela López
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - María Belén Palma
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fleni, Consejo de Investigaciones Científicas y Técnicas (CONICET), Escobar 1625, Argentina
| | - Cintia Cevallos
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Franco Agustin Sviercz
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Patricio Jarmoluk
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Marcela Nilda García
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Jorge Quarleri
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - M Victoria Delpino
- Laboratorio de Inmunopatología Viral, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| |
Collapse
|
45
|
Li K, Huang K, Lu Q, Geng W, Jiang D, Guo A. TRIM16 mitigates impaired osteogenic differentiation and antioxidant response in D-galactose-induced senescent osteoblasts. Eur J Pharmacol 2024; 979:176849. [PMID: 39059569 DOI: 10.1016/j.ejphar.2024.176849] [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: 02/25/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
Senile osteoporosis (SOP), characterized by significant bone loss, poses a substantial threat to elderly skeletal health, with oxidative stress playing a crucial role in its pathogenesis. Although Tripartite Motif 16 (TRIM16) has been identified as a promoter of antioxidant response and osteogenic differentiation, its regulatory role in SOP remains incompletely understood. This study aims to elucidate the underlying mechanism of TRIM16 in mitigating D-galactose (D-gal)-induced senescent osteoblasts. Initially, we observed diminished bone mineral density (BMD) and impaired bone microstructure in naturally aging (24 months) and D-gal-induced (18 months) aged mice through Dual-energy X-ray absorptiometry (DEXA), micro-CT, hematoxylin and eosin staining, and Masson staining. Immunohistochemistry analysis revealed downregulation of TRIM16 and osteogenic differentiation markers (Collagen-1, Runx-2, osteopontin) in femur samples of aged mice. Furthermore, in D-gal-induced senescent MC3T3-E1 osteoblasts, we observed the suppression of osteogenic differentiation and maturity, along with cytoskeleton impairment via Alkaline phosphatase (ALP), Alizarin Red S, and Rhodamine-phalloidin staining. The protein expression of TRIM16, osteogenic differentiation markers, and antioxidant indicators (Nrf-2, HO-1, SOD1) decreased, while the production of reactive oxygen species (ROS) significantly increased. Knockdown and overexpression of TRIM16 using lentivirus in osteoblasts revealed that the downregulation of TRIM16 inhibited osteogenic differentiation and induced oxidative stress. Notably, TRIM16 overexpression partially attenuated D-gal-induced inhibition of osteogenic differentiation and increased oxidative stress. These findings suggest TRIM16 may mitigate impaired osteogenic differentiation and antioxidant response in D-gal-induced senescent osteoblasts, suggesting its potential as a therapeutic target for SOP.
Collapse
Affiliation(s)
- Kai Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ke Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Quanyi Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenbo Geng
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dianming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Ai Guo
- Chongqing Institute of Cadre Health Care Research, The First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China.
| |
Collapse
|
46
|
Costa do Bomfim FR, Gonzalez Sella VR, Thomasini RL, Plapler H. Photobiomodulation Modulates Proliferation and Gene Expression Related to Calcium Signaling in Human Osteoblast Cells. J Lasers Med Sci 2024; 15:e45. [PMID: 39381787 PMCID: PMC11459251 DOI: 10.34172/jlms.2024.45] [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: 04/16/2024] [Accepted: 07/29/2024] [Indexed: 10/10/2024]
Abstract
Introduction: Photobiomodulation with low-level laser treatment can enhance bone formation by stimulating the cell division of osteoblasts and increasing the amount of protein deposition, thus encouraging the formation of new bone. The aim of this study was to evaluate the effects of photobiomodulation with a low-level laser on proliferation and gene expression related to calcium signaling in human osteoblasts. Methods: Osteoblastic cell lines of the hFOB1.19 lineage, human osteoblasts, were grown and assigned into two groups, control (C; n=78 cultured wells) and photobiomodulation (L; n=78 cultured wells) with n=6 per day of the experimental period. Cells were cultured (immature at 34 ºC), and after maturation at 37 ºC, group L cells were exposed to laser irradiation with a low-level laser device (gallium and aluminum arsenide), at a wavelength of 808 nm, a power output of 200 mW, and a power density of 200 mW/cm2. The energy delivered to the cells was 37 J/cm2, with a beam area of 0.02 mm2 and an exposure time of 5 seconds. This treatment was applied daily for a period of 13 days. Following this, the number of cells was counted, and RNA was isolated, measured, and then converted into cDNA for further quantification using a comparative Ct method with real-time polymerase chain reaction. The results were then subjected to statistical analysis through a Mann-Whitney test, with a significance level of P<0.05. Results: The cell count in the L group (37.25x10±4±22.02) was statistically higher compared to the control group (22.75x10±4±7.660) with a P value of 0.0259. The values of 2-ΔΔCt for S100A6, plasma membrane calcium ATPase (PMCA), and calmodulin genes indicated hyper-expression on the thirteenth day, while the osteocalcin gene showed hypo-expression. Conclusion: The study suggests that the photobiomodulation mechanism with a low-level laser may regulate gene expression in human osteoblasts in a dose-dependent and cumulative manner.
Collapse
Affiliation(s)
- Fernando Russo Costa do Bomfim
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
- Laboratory of Molecular Biology, Centro Universitário da Fundação Hermínio Ometto - FHO, Araras, SP, Brazil
| | - Valéria Regina Gonzalez Sella
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Ronaldo Luis Thomasini
- Medicine Faculty, Universidade Federal dos Vales do Jequitinhonha e Mucuri - UFVJM, Diamantina, MG, Brazil
| | - Hélio Plapler
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| |
Collapse
|
47
|
Xu H, Wang L, Zhu X, Zhang H, Chen H, Zhang H. Jintiange capsule ameliorates glucocorticoid-induced osteonecrosis of the femoral head in rats by regulating the activity and differentiation of BMSCs. J Tradit Complement Med 2024; 14:568-580. [PMID: 39262662 PMCID: PMC11384076 DOI: 10.1016/j.jtcme.2024.03.013] [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: 09/12/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 09/13/2024] Open
Abstract
Background and aim A surplus of glucocorticoids (GC) is a main cause of non-traumatic osteonecrosis of the femoral head (ONFH), and Jintiange (JTG), as one of the traditional Chinese medicines (TCM), also plays an instrumental role in the alleviation of bone loss simultaneously. Therefore, JTG was thought to be able to reverse GC-induced ONFH (GC-ONFH) to a certain extent. Experimental procedure In vivo, the effect of JTG on trabeculae in the subchondral bone of the femoral head was investigated using micro-computed tomography (micro-CT), TdT-mediated dUTP nick end labeling (TUNEL) and histological staining; in vitro, proliferation, viability, apoptosis, and senescence of purified bone mesenchymal stem cells (BMSCs) were examined to demonstrate the direct impact of JTG on these cells. Meanwhile after using a series of interventions, the function of JTG on BMSC differentiation could be assessed by measuring of osteogenic and adipogenic markers at levels of protein and mRNA. Results Our final results demonstrated that with the involvement of Wnt/β-catenin pathway, JTG was able to significantly promote osteogenesis, restrain adipogenesis, delay senescence in BMSCs, reduce osteoclast number, weaken apoptosis, and enhance proliferation of osteocytes, all of which could mitigate the progression of subchondral osteonecrosis. Conclusion According to the results of experiments in vitro and vivo, JTG was deemed to relieve the early GC-ONFH using the prevention of destruction of subchondral bone, which was contributed to regulating the differentiation of BMSCs and the number of osteoclasts.
Collapse
Affiliation(s)
- Hui Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lin Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xunpeng Zhu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haigang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongwei Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
48
|
Chen J, Ying Y, Li H, Sha Z, Lin J, Wu Y, Wu Y, Zhang Y, Chen X, Zhang W. Abnormal dental follicle cells: A crucial determinant in tooth eruption disorders (Review). Mol Med Rep 2024; 30:168. [PMID: 39027997 DOI: 10.3892/mmr.2024.13292] [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: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
The dental follicle (DF) plays an indispensable role in tooth eruption by regulating bone remodeling through their influence on osteoblast and osteoclast activity. The process of tooth eruption involves a series of intricate regulatory mechanisms and signaling pathways. Disruption of the parathyroid hormone‑related protein (PTHrP) in the PTHrP‑PTHrP receptor signaling pathway inhibits osteoclast differentiation by DF cells (DFCs), thus resulting in obstructed tooth eruption. Furthermore, parathyroid hormone receptor‑1 mutations are linked to primary tooth eruption failure. Additionally, the Wnt/β‑catenin, TGF‑β, bone morphogenetic protein and Hedgehog signaling pathways have crucial roles in DFC involvement in tooth eruption. DFC signal loss or alteration inhibits osteoclast differentiation, affects osteoblast and cementoblast differentiation, and suppresses DFC proliferation, thus resulting in failed tooth eruptions. Abnormal tooth eruption is also associated with a range of systemic syndromes and genetic diseases, predominantly resulting from pathogenic gene mutations. Among these conditions, the following disorders arise due to genetic mutations that disrupt DFCs and impede proper tooth eruption: Cleidocranial dysplasia associated with Runt‑related gene 2 gene mutations; osteosclerosis caused by CLCN7 gene mutations; mucopolysaccharidosis type VI resulting from arylsulfatase B gene mutations; enamel renal syndrome due to FAM20A gene mutations; and dentin dysplasia caused by mutations in the VPS4B gene. In addition, regional odontodysplasia and multiple calcific hyperplastic DFs are involved in tooth eruption failure; however, they are not related to gene mutations. The specific mechanism for this effect requires further investigation. To the best of our knowledge, previous reviews have not comprehensively summarized the syndromes associated with DF abnormalities manifesting as abnormal tooth eruption. Therefore, the present review aims to consolidate the current knowledge on DFC signaling pathways implicated in abnormal tooth eruption, and their association with disorders of tooth eruption in genetic diseases and syndromes, thereby providing a valuable reference for future related research.
Collapse
Affiliation(s)
- Jiahao Chen
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Ying Ying
- Department of Child Health, Yongkang Women and Children's Health Hospital, Yongkang, Zhejiang 321300, P.R. China
| | - Huimin Li
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhuomin Sha
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Jiaqi Lin
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yongjia Wu
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yange Wu
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yun Zhang
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Xuepeng Chen
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Weifang Zhang
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
49
|
Yi L, Han N, Li Z, Jiang H, Cao Z. Relaxin-2 promotes osteoblastic differentiation mediated by epidermal growth factor and epidermal growth factor receptor signaling. Biotechnol Appl Biochem 2024. [PMID: 39219221 DOI: 10.1002/bab.2661] [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: 12/11/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Loss of osteogenic differentiation potential of osteoblasts has been associated with the pathogenesis of osteoporosis. Thus, stimulation of osteoblastic differentiation is a therapeutic strategy for osteoporosis. Relaxin-2 is a peptide hormone with potent biological functions. However, the effects of Relaxin-2 in osteoblastic differentiation and osteoporosis have not been reported before. Here, we report a novel physiological role of Relaxin-2 in promoting osteoblastic differentiation and mineralization of MC3T3-E1 cells. Our results indicate that exposure to Relaxin-2 upregulated the expression, and elevated the activity of alkaline phosphatase (ALP) when MC3T3-E1 cells were cultured in osteogenic differentiation medium (OM). Additionally, Relaxin-2 upregulated the mRNA levels of osteocalcin (ocn), osteopontin (opn), and collagen type I alpha 1 (Col1a1). The alizarin red S staining assay revealed that Relaxin-2 promoted the mineralization of MC3T3-E1 cells. We also found that Relaxin-2 increased the expression of Runx-2 as well as the epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR). Importantly, silencing of EGF abolished the effects of Relaxin-2 in osteoblastic differentiation and related gene expression. These findings suggest that Relaxin-2 stimulates osteogenic differentiation through activating EGF/EGFR signaling.
Collapse
Affiliation(s)
- Lankai Yi
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Ning Han
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhong Li
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Housen Jiang
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhenhao Cao
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| |
Collapse
|
50
|
Garcia-Perez VI, Hotchkiss KM, Silva-Bermudez P, Hernández MM, Prado-Prone G, Olivares-Navarrete R, Rodil SE, Almaguer-Flores A. Amorphous TiO 2nano-coating on stainless steel to improve its biological response. Biomed Mater 2024; 19:055037. [PMID: 39121890 PMCID: PMC11337115 DOI: 10.1088/1748-605x/ad6dc4] [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: 04/19/2024] [Revised: 07/23/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
This study delves into the potential of amorphous titanium oxide (aTiO2) nano-coating to enhance various critical aspects of non-Ti-based metallic orthopedic implants. These implants, such as medical-grade stainless steel (SS), are widely used for orthopedic devices that demand high strength and durability. The aTiO2nano-coating, deposited via magnetron sputtering, is a unique attempt to improve the osteogenesis, the inflammatory response, and to reduce bacterial colonization on SS substrates. The study characterized the nanocoated surfaces (SS-a TiO2) in topography, roughness, wettability, and chemical composition. Comparative samples included uncoated SS and sandblasted/acid-etched Ti substrates (Ti). The biological effects were assessed using human mesenchymal stem cells (MSCs) and primary murine macrophages. Bacterial tests were carried out with two aerobic pathogens (S. aureusandS. epidermidis) and an anaerobic bacterial consortium representing an oral dental biofilm. Results from this study provide strong evidence of the positive effects of the aTiO2nano-coating on SS surfaces. The coating enhanced MSC osteoblastic differentiation and exhibited a response similar to that observed on Ti surfaces. Macrophages cultured on aTiO2nano-coating and Ti surfaces showed comparable anti-inflammatory phenotypes. Most significantly, a reduction in bacterial colonization across tested species was observed compared to uncoated SS substrates, further supporting the potential of aTiO2nano-coating in biomedical applications. The findings underscore the potential of magnetron-sputtering deposition of aTiO2nano-coating on non-Ti metallic surfaces such as medical-grade SS as a viable strategy to enhance osteoinductive factors and decrease pathogenic bacterial adhesion. This could significantly improve the performance of metallic-based biomedical devices beyond titanium.
Collapse
Affiliation(s)
- Victor I Garcia-Perez
- Laboratorio de Biointerfases, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, Ciudad de México, CDMX 04510, Mexico
| | - Kelly M Hotchkiss
- Department of Biomedical Engineering Commonwealth, College of Engineering, Virginia University, Richmond, VA 23284, United States of America
| | - Phaedra Silva-Bermudez
- Unidad de Ingeniería de Tejidos,Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra. Calzada México-Xochimilco, Ciudad de México 14389, Mexico
| | - Miryam Martínez Hernández
- Laboratorio de Biointerfases, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, Ciudad de México, CDMX 04510, Mexico
| | - Gina Prado-Prone
- Laboratorio de Biointerfases, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, Ciudad de México, CDMX 04510, Mexico
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering Commonwealth, College of Engineering, Virginia University, Richmond, VA 23284, United States of America
| | - Sandra E Rodil
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, Ciudad de México, CDMX 04510, Mexico
| | - Argelia Almaguer-Flores
- Laboratorio de Biointerfases, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México. Circuito exterior s/n, Ciudad Universitaria, Ciudad de México, CDMX 04510, Mexico
| |
Collapse
|