1
|
Li Y, Sun Y, Ma K, Wang S, Wang Z, Huang L. Functional mechanism and clinical implications of LINC00339 in delayed fracture healing. J Orthop Surg Res 2024; 19:511. [PMID: 39192334 PMCID: PMC11348643 DOI: 10.1186/s13018-024-04998-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
OBJECTIVE Delayed fracture healing is a common complication of fractures that significantly impacts human health. This study aimed to explore the role of LINC00339 (lncRNA) in delayed fracture healing to provide new directions for its treatment. METHODS This study included 82 patients with fractures healing in a normal manner and 90 patients experiencing delayed fracture healing. Levels of LINC00339, miR-16-5p, and osteogenic marker-related mRNAs were measured using RT-qPCR. The predictive potential of LINC00339 for delayed fracture healing was validated using ROC curve analysis. The interaction between LINC00339 and miR-16-5p was validated using dual-luciferase reporter assays and RIP experiments. CCK-8 was used to assess cell proliferation, and apoptosis rates were measured by flow cytometry. RESULTS LINC00339 was significantly upregulated in delayed fracture healing patients and exhibited strong predictive ability for this condition. Overexpression of LINC00339 inhibited osteoblast proliferation, promoted apoptosis, and reduced mRNA levels of osteogenic markers (P < 0.05). miR-16-5p was recognized as a target mRNA of LINC00339, with LINC00339 exerting negative regulation on miR-16-5p, while overexpression of miR-16-5p mitigated the inhibitory effects of LINC00339 on fracture healing (P < 0.05). CONCLUSION This research indicated that LINC00339 may serve as a diagnostic marker for delayed fracture healing and revealed the function of the LINC00339/miR-16-5p axis on fracture healing by regulating osteoblasts.
Collapse
Affiliation(s)
- Yuntao Li
- Department of Integrated Traditional Chinese and Western Medicine, Tianjin Hospital, Tianjin, 300211, China
| | - Ya Sun
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ke Ma
- College of Medical Technology, Zibo Vocational Institute, Zibo, 255000, China
| | - Shengqian Wang
- Department of Emergency, Zibo Combined Traditional Chinese and Western Medicine Hospital, Zibo, 255000, China
| | - Zhibiao Wang
- Department of Orthopedics, Rizhao Central Hospital, No. 66, Wanghai Road, Donggang District, Rizhao City, Shandong Province, 276800, China.
| | - Lina Huang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise City, Guangxi, 533000, China.
| |
Collapse
|
2
|
Wei L, Xie Y, Yu P, Zhu Q, Lan X, Xiao J. Bioinformatics analysis and validation of RNA methylation-related genes in osteogenic and adipogenic differentiation of rat bone marrow mesenchymal stem cells. Biochem Biophys Res Commun 2024; 739:150570. [PMID: 39181069 DOI: 10.1016/j.bbrc.2024.150570] [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: 05/29/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The regulatory mechanisms of RNA methylation during the processes of osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) have yet to be fully understood. The objective of our study was to analyze and validate the contribution of RNA methylation regulators to the mechanisms underlying the osteogenic and adipogenic differentiation of rat BMSCs. METHODS We downloaded the GSE186026 from the Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) were screened using the DESeq2 package in R software (version 3.6.3). A total of 50 RNA methylation genes obtained from literature review and summary were intersected with the previous DEGs to obtain RNA methylation genes, which have different expressions (RM-DEGs). Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were utilized to reveal the functional enrichment. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to validate RM-DEGs. Protein-protein interaction network (PPI) analysis and visual analysis were performed using STRING and Cytoscape. RM-DEGs regulatory network was constructed to analyze the top 10 hub genes. The relationship between RM-DEGs, some enriched GO and pathways was also been analyzed. The miRNAs and RM-DEGs regulatory networks were established by using miRWalk and TargetScan. RESULTS As part of our research, we detected varying levels of expression for m6A regulators Mettl3 and Rbm15, as well as m7G regulators Mettl1 and Wdr4, in relation to osteogenic differentiation, along with m6A regulator Fmr1 in adipogenic differentiation. The protein-protein interaction (PPI) networks were constructed for 49 differentially expressed genes (DEGs) related to RNA methylation during the process of osteogenic differentiation, and 13 DEGs for adipogenic differentiation. Moreover, top10 hub genes were calculated. In osteogenic differentiation, Mettl3 regulated the Wnt pathway and Hippo pathway by regulating Smad3, Rbm15 regulated the Notch pathway by Notch1, Mettl1 regulated the PI3K-Akt pathway by Gnb4. In adipogenic differentiation, Fmr1 regulated the PI3K-Akt pathway by Egfr. M6A methylation sites of Smad3, Notch1 and Gnb4 were predicted, and the results showed that all three genes were possibly methylated by m6A, and more than 9 sites per gene were possibly methylated. Finally, we constructed the regulatory networks of Mettl3, Rbm15, Mettl1, and Wdr4 and 109 miRNAs in osteogenic differentiation, Fmr1 and 118 miRNAs in adipogenic differentiation. CONCLUSIONS Mettl3(m6A), Rbm15(m6A), Wdr4 and Mettl1(m7G) were differentially expressed in osteogenic differentiation, while Fmr1(m6A) was differentially expressed in adipogenic differentiation. These findings offered potential candidates for further research on the involvement of RNA methylation in the osteogenic and adipogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Li Wei
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Yuping Xie
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Peiyang Yu
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Qiang Zhu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Jingang Xiao
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
3
|
Huang Y, Zhang M, Zhang J, Liu S, Li D, Qiao Z, Yao H, Shi Q, Zhou X, Ma F. diABZI and poly(I:C) inhibit osteoclastic bone resorption by inducing IRF7 and IFIT3. J Bone Miner Res 2024; 39:1132-1146. [PMID: 38874138 PMCID: PMC11337579 DOI: 10.1093/jbmr/zjae093] [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: 02/19/2024] [Revised: 05/11/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
Type I interferons (IFN-I) are pleiotropic factors endowed with multiple activities that play important roles in innate and adaptive immunity. Although many studies indicate that IFN-I inducers exert favorable effects on broad-spectrum antivirus, immunomodulation, and anti-tumor activities by inducing endogenous IFN-I and IFN-stimulated genes, their function in bone homeostasis still needs further exploration. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. First, IFN-I inducers suppress the genes that control osteoclast (OC) differentiation and activity in vitro. Moreover, diABZI alleviates bone loss in Ti particle-induced osteolysis and ovariectomized -induced osteoporosis in vivo by inhibiting OC differentiation and function. In addition, the inhibitory effects of IFN-I inducers on OC differentiation are not observed in macrophages derived from Ifnar1-/-mice, which indicate that the suppressive effect of IFN-I inducers on OC is IFNAR-dependent. Mechanistically, RNAi-mediated silencing of IRF7 and IFIT3 in OC precursors impairs the suppressive effect of the IFN-I inducers on OC differentiation. Taken together, these results demonstrate that IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway.
Collapse
Affiliation(s)
- Yingkang Huang
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
| | - Mingchao Zhang
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, China
| | - Jun Zhang
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou 310014, Zhejiang, China
| | - Siying Liu
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, China
| | - Dapei Li
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
| | - Zigang Qiao
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
| | - Haiping Yao
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, China
| | - Feng Ma
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou Institute of Systems Medicine, Suzhou 215123, Jiangsu, China
| |
Collapse
|
4
|
Han Y, Dong Y, Jia B, Shi X, Zhao H, Li S, Wang H, Sun B, Yin L, Dai K. High-precision bioactive scaffold with dECM and extracellular vesicles targeting 4E-BP inhibition for cartilage injury repair. Mater Today Bio 2024; 27:101114. [PMID: 39211509 PMCID: PMC11360177 DOI: 10.1016/j.mtbio.2024.101114] [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: 01/29/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 09/04/2024] Open
Abstract
The restoration of cartilage injuries remains a formidable challenge in orthopedics, chiefly attributed to the absence of vascularization and innervation in cartilage. Decellularized extracellular matrix (dECM) derived from cartilage, following antigenic removal through decellularization processes, has exhibited remarkable biocompatibility and bioactivity, rendering it a viable candidate for cartilage repair. Additionally, extracellular vesicles (EVs) generated from cartilage have demonstrated a synergistic effect when combined with dECM, potentially mitigating the inhibitory impact on protein synthesis by phosphorylating 4ebp, thereby promoting the synthesis of cartilage-related proteins such as collagen. In pursuit of this objective, we have innovated a novel bioink and repair scaffold characterized by exceptional biocompatibility, bioactivity, and biodegradability, establishing a tissue-specific microenvironment conducive to chondrogenesis. Within rat osteochondral defects, the biologically active scaffold successfully prompted the formation of transparent cartilage, featuring adequate mechanical strength, favorable elasticity, and dECM deposition indicative of cartilage. In summary, this study has effectively engineered a hydrogel bioink tailored for cartilage repair and devised a bioactive cartilage repair scaffold proficient in instigating cell differentiation and fostering cartilage repair.
Collapse
Affiliation(s)
- Yu Han
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Orthopedic Surgery, Shanghai Key Laboratory of Orthopedic Implants,Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yixin Dong
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Bo Jia
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiangyu Shi
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongbo Zhao
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shushan Li
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Haitao Wang
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Li Yin
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kerong Dai
- Department of Orthopedic Surgery, Shanghai Key Laboratory of Orthopedic Implants,Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
5
|
Feng R, Cheng S, Zhang F, Xu K, Liu L, Yang M, Xu P. Evaluating the association between lifestyle factors and heel bone mineral density in different inflammatory states. Heliyon 2024; 10:e33435. [PMID: 39040264 PMCID: PMC11261795 DOI: 10.1016/j.heliyon.2024.e33435] [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: 09/11/2023] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Rationale It is unclear whether lifestyle factors affect bone mineral density (BMD) during different inflammatory states. Objective This study investigated the effects of coffee consumption, vitamin D (VD) intake, smoking, and alcohol consumption on heel BMD in adults with different inflammatory states. Methods The phenotypic data from 249,825 participants were analyzed using the UK Biobank cohort. The inflammatory status was evaluated using C-reactive protein (CRP) levels and the systemic immune-inflammation index. Linear regression analysis was used to examine the association between coffee consumption, VD, smoking, alcohol consumption, and heel BMD in adults with different inflammatory states. Linear regression models were used to analyze the interaction between inflammation and the four lifestyle factors with respect to their influence on heel BMD in adults. Results Our findings revealed that VD was positively associated with adult heel BMD (β = 2.41 × 10-2, SE = 5.14 × 10-3, P = 2.72 × 10-6), while alcohol consumption and smoking were negatively associated with adult heel BMD. Coffee was negatively associated with adult heel BMD in low inflammatory states (β = -1.27 × 10-2, SE = 4.79 × 10-3, P = 8.00 × 10-3), while there was no association between coffee and adult heel BMD in high inflammatory states. Overall, it was found that these four lifestyle factors interacted negatively with inflammatory states. Conclusion Our study suggests that VD is positively associated with adult heel BMD and that alcohol consumption and smoking are negatively associated with adult heel BMD. Coffee may reverse the adverse effects of inflammation on BMD when the patient is in a highly inflammatory state, thus acting as a protective agent against heel BMD in adults.
Collapse
Affiliation(s)
- Ruoyang Feng
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiao Tong University, Xi'an, Shanxi, 710054, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiao Tong University, Xi'an, Shanxi, 710054, China
| | - Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiao Tong University, Xi'an, Shanxi, 710054, China
| | - Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiao Tong University, Xi'an, Shanxi, 710054, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiao Tong University, Xi'an, Shanxi, 710054, China
| |
Collapse
|
6
|
Fu L, Zhang P, Wang Y, Liu X. Microbiota-bone axis in ageing-related bone diseases. Front Endocrinol (Lausanne) 2024; 15:1414350. [PMID: 39076510 PMCID: PMC11284018 DOI: 10.3389/fendo.2024.1414350] [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: 04/08/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Bone homeostasis in physiology depends on the balance between bone formation and resorption, and in pathology, this homeostasis is susceptible to disruption by different influences, especially under ageing condition. Gut microbiota has been recognized as a crucial factor in regulating host health. Numerous studies have demonstrated a significant association between gut microbiota and bone metabolism through host-microbiota crosstalk, and gut microbiota is even an important factor in the pathogenesis of bone metabolism-related diseases that cannot be ignored. This review explores the interplay between gut microbiota and bone metabolism, focusing on the roles of gut microbiota in bone ageing and aging-related bone diseases, including osteoporosis, fragility fracture repair, osteoarthritis, and spinal degeneration from different perspectives. The impact of gut microbiota on bone metabolism during aging through modification of endocrinology system, immune system and gut microbiota metabolites are summarized, facilitating a better grasp of the pathogenesis of aging-related bone metabolic diseases. This review offers innovative insights into targeting the gut microbiota for the treatment of bone ageing-related diseases as a clinical therapeutic strategy.
Collapse
Affiliation(s)
| | | | | | - Xiaonan Liu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Dai K, Geng Z, Zhang W, Wei X, Wang J, Nie G, Liu C. Biomaterial design for regenerating aged bone: materiobiological advances and paradigmatic shifts. Natl Sci Rev 2024; 11:nwae076. [PMID: 38577669 PMCID: PMC10989671 DOI: 10.1093/nsr/nwae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/04/2024] [Accepted: 02/26/2024] [Indexed: 04/06/2024] Open
Abstract
China's aging demographic poses a challenge for treating prevalent bone diseases impacting life quality. As bone regeneration capacity diminishes with age due to cellular dysfunction and inflammation, advanced biomaterials-based approaches offer hope for aged bone regeneration. This review synthesizes materiobiology principles, focusing on biomaterials that target specific biological functions to restore tissue integrity. It covers strategies for stem cell manipulation, regulation of the inflammatory microenvironment, blood vessel regeneration, intervention in bone anabolism and catabolism, and nerve regulation. The review also explores molecular and cellular mechanisms underlying aged bone regeneration and proposes a database-driven design process for future biomaterial development. These insights may also guide therapies for other age-related conditions, contributing to the pursuit of 'healthy aging'.
Collapse
Affiliation(s)
- Kai Dai
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology; Shanghai 200237, China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Wenchao Zhang
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology; Shanghai 200237, China
| | - Xue Wei
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology; Shanghai 200237, China
| | - Jing Wang
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology; Shanghai 200237, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology; Shanghai 200237, China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
8
|
Xiao CL, Liu LL, Tang W, Liu WY, Wu LY, Zhao K. Reduction of the trans-cortical vessel was associated with bone loss, another underlying mechanism of osteoporosis. Microvasc Res 2024; 152:104650. [PMID: 38123064 DOI: 10.1016/j.mvr.2023.104650] [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: 09/15/2023] [Revised: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
RATIONALE Numerous studies have established a robust association between bone morrow microvascular diseases and osteoporosis. This study sought to investigate the relationship between alterations in trans-cortical vessel (TCVs) and the onset of osteoporosis in various mouse models. METHODS Aged mice, ovariectomized mice, and db/db mice, were utilized as osteoporosis models. TCVs in the tibia were detected using tissue clearing and light sheet fluorescence microscopy imaging. Femurs bone mass were analyzed using micro-CT scanning. Correlations between the number of TCVs and bone mass were analyzed using Pearson correlation analysis. RESULTS All osteoporosis mouse models showed a significant reduction in the number of TCVs compared to the control group. Correlation analysis revealed a positive association between the number of TCVs and bone mass. TCVs were also expressed high levels of CD31 and EMCN proteins as type H vessels. CONCLUSIONS This study underscores a consistent correlation between the number of TCVs and bone mass. Moreover, TCVs may serve as a potential biomarker for bone mass evaluation.
Collapse
Affiliation(s)
- Chun-Lin Xiao
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China
| | - Lu-Lin Liu
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China
| | - Wen Tang
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China
| | - Wu-Yang Liu
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China
| | - Long-Yan Wu
- Ganzhou People's Hospital, Ganzhou, PR China.
| | - Kai Zhao
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China; Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
9
|
Xiao W, Huang W, Zhou Y, Jin Z, Wei X, Li J. Ti 3C 2@UiO-TCPP Schottky junction photoelectrochemical sensor for detecting alkaline phosphatase through the steric hindrance effect of phosphopeptide. Anal Chim Acta 2024; 1289:342210. [PMID: 38245201 DOI: 10.1016/j.aca.2024.342210] [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: 10/20/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024]
Abstract
Alkaline phosphatase (ALP) is a major biomarker for clinical diagnosis, but detection methods of ALP are limited in sensitivity and selectivity. In this paper, a novel method for ALP determination is proposed. A photoelectrochemical (PEC) sensor was prepared by growing UiO-tetratopic tetrakis (4-carbox-yphenyl) porphyrin (TCPP) in situ between layered Ti3C2 through a one-pot hydrothermal method. The obtained Schottky heterojunction photoelectric material Ti3C2@UiO-TCPP not only has a large light absorption range but also greatly improves the efficiency of photogenerated electron hole separation and thereby enhances sensitivity for PEC detection. The phosphate group on the phosphorylated polypeptide was utilized to form a Zr-O-P bond with the zirconium ion on UiO-66, and then photocurrent decreases due to the steric hindrance effect of phosphorylated polypeptides, that is, the hindrance of electron transfer between the photoelectric material and a solution. The specific interaction between ALP and phosphorylated polypeptides shears the bond between phosphate and zirconium ion on UiO-66 in the peptides then weakens the hindrance effect and increases the photocurrent, thus realizing ALP detection. The linear range of ALP is 0.03-10,000 U·L-1, and the detection limit is 0.012 U·L-1. The method is highly sensitive and selective, and has been applied in detection of ALP in serum samples.
Collapse
Affiliation(s)
- Wei Xiao
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi, 541004, China
| | - WanJin Huang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi, 541004, China
| | - Yu Zhou
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi, 541004, China
| | - Zhenhuan Jin
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi, 541004, China
| | - Xiaoping Wei
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi, 541004, China.
| | - Jianping Li
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi, 541004, China.
| |
Collapse
|
10
|
Li T, Li W, Guo X, Tan T, Xiang C, Ouyang Z. Unraveling the potential mechanisms of the anti-osteoporotic effects of the Achyranthes bidentata-Dipsacus asper herb pair: a network pharmacology and experimental study. Front Pharmacol 2023; 14:1242194. [PMID: 37849727 PMCID: PMC10577322 DOI: 10.3389/fphar.2023.1242194] [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: 06/18/2023] [Accepted: 09/07/2023] [Indexed: 10/19/2023] Open
Abstract
Background: Osteoporosis is a prevalent bone metabolism disease characterized by a reduction in bone density, leading to several complications that significantly affect patients' quality of life. The Achyranthes bidentata-Dipsacus asper (AB-DA) herb pair is commonly used in Traditional Chinese Medicine (TCM) to treat osteoporosis. This study aimed to investigate the therapeutic compounds and potential mechanisms of AB-DA using network pharmacology, molecular docking, molecular dynamics simulation, and experimental verification. Methods: Identified compounds of AB-DA were collected from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), Traditional Chinese Medicine Information Database (TCM-ID), TCM@Taiwan Database, BATMAN-TCM, and relevant literature. The main bioactive ingredients were screened based on the criteria of "OB (oral bioavailability) ≥ 30, DL (drug-likeness) ≥ 0.18." Potential targets were predicted using the PharmMapper and SwissTargetPrediction websites, while disease (osteoporosis)-related targets were obtained from the GeneCards, DisGeNET, and OMIM databases. The PPI network and KEGG/GO enrichment analysis were utilized for core targets and pathway screening in the STRING and Metascape databases, respectively. A drug-compound-target-pathway-disease network was constructed using Cytoscape software to display core regulatory mechanisms. Molecular docking and dynamics simulation techniques explored the binding reliability and stability between core compounds and targets. In vitro and in vivo validation experiments were utilized to explore the anti-osteoporosis efficiency and mechanism of sitogluside. Results: A total of 31 compounds with 83 potential targets for AB-DA against osteoporosis were obtained. The PPI analysis revealed several hub targets, including AKT1, CASP3, EGFR, IGF1, MAPK1, MAPK8, and MAPK14. GO/KEGG analysis indicated that the MAPK cascade (ERK/JNK/p38) is the main pathway involved in treating osteoporosis. The D-C-T-P-T network demonstrated therapeutic compounds that mainly consisted of iridoids, steroids, and flavonoids, such as sitogluside, loganic acid, and β-ecdysterone. Molecular docking and dynamics simulation analyses confirmed strong binding affinity and stability between core compounds and targets. Additionally, the validation experiments showed preliminary evidence of antiosteoporosis effects. Conclusion: This study identified iridoids, steroids, and flavonoids as the main therapeutic compounds of AB-DA in treating osteoporosis. The underlying mechanisms may involve targeting core MAPK cascade (ERK/JNK/p38) targets, such as MAPK1, MAPK8, and MAPK14. In vivo experiments preliminarily validated the anti-osteoporosis effect of sitogluside. Further in-depth experimental studies are required to validate the therapeutic value of AB-DA for treating osteoporosis in clinical practice.
Collapse
Affiliation(s)
- Tao Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenzhao Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoning Guo
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Tan
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Cheng Xiang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Lee YCJ, Javdan B, Cowan A, Smith K. More than skin deep: cyclic peptides as wound healing and cytoprotective compounds. Front Cell Dev Biol 2023; 11:1195600. [PMID: 37325572 PMCID: PMC10267460 DOI: 10.3389/fcell.2023.1195600] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
The prevalence and cost of wounds pose a challenge to patients as well as the healthcare system. Wounds can involve multiple tissue types and, in some cases, become chronic and difficult to treat. Comorbidities may also decrease the rate of tissue regeneration and complicate healing. Currently, treatment relies on optimizing healing factors rather than administering effective targeted therapies. Owing to their enormous diversity in structure and function, peptides are among the most prevalent and biologically important class of compounds and have been investigated for their wound healing bioactivities. A class of these peptides, called cyclic peptides, confer stability and improved pharmacokinetics, and are an ideal source of wound healing therapeutics. This review provides an overview of cyclic peptides that have been shown to promote wound healing in various tissues and in model organisms. In addition, we describe cytoprotective cyclic peptides that mitigate ischemic reperfusion injuries. Advantages and challenges in harnessing the healing potential for cyclic peptides from a clinical perspective are also discussed. Cyclic peptides are a potentially attractive category of wound healing compounds and more research in this field could not only rely on design as mimetics but also encompass de novo approaches as well.
Collapse
Affiliation(s)
- Ying-Chiang J. Lee
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Bahar Javdan
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Alexis Cowan
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Keith Smith
- Merck & Co., Inc., Kenilworth, NJ, United States
| |
Collapse
|
12
|
Wang X, Zhu X, Wang D, Li X, Wang J, Yin G, Huang Z, Pu X. Identification of a Specific Phage as Growth Factor Alternative Promoting the Recruitment and Differentiation of MSCs in Bone Tissue Regeneration. ACS Biomater Sci Eng 2023; 9:2426-2437. [PMID: 37023478 DOI: 10.1021/acsbiomaterials.2c01538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Inefficient use and loss of exogenously implanted mesenchymal stem cells (MSCs) are major concerns in MSCs-based bone tissue engineering. It is a promising approach to overcome the above issues by recruiting and regulation of endogenous MSCs. However, there are few substances that can recruit MSCs effectively and specifically to the site of bone injury. In this study, we identified a phage clone (termed P11) with specific affinity for MSCs through phage display biopanning, and further investigated the effects of P11 on the cytological behavior of MSCs and macrophages. The results showed that P11 could bind MSCs specifically and promote the proliferation and migration of MSCs. Meanwhile, P11 could polarize macrophages to the M1 phenotype and significantly changed their morphology, which further enhanced the chemotaxis of MSCs. Additionally, RNA-seq results revealed that P11 could promote the secretion of osteogenesis-related markers in MSCs through the TPL2-MEK-ERK signaling pathway. Altogether, P11 has great potential to be used as growth factor alternatives in bone tissue engineering, with the advantages of cheaper and stable activity. Our study also advances the understanding of the effects of phages on macrophages and MSCs, and provides a new idea for the development in the field of phage-based tissue engineering.
Collapse
Affiliation(s)
- Xingming Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiupeng Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Danni Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
13
|
García-García RD, Garay-Pacheco E, Marín-Llera JC, Chimal-Monroy J. Recombinant Limb Assay as in Vivo Organoid Model. Front Cell Dev Biol 2022; 10:863140. [PMID: 35557939 PMCID: PMC9086426 DOI: 10.3389/fcell.2022.863140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Organ formation initiates once cells become committed to one of the three embryonic germ layers. In the early stages of embryogenesis, different gene transcription networks regulate cell fate after each germ layer is established, thereby directing the formation of complex tissues and functional organs. These events can be modeled in vitro by creating organoids from induced pluripotent, embryonic, or adult stem cells to study organ formation. Under these conditions, the induced cells are guided down the developmental pathways as in embryonic development, resulting in an organ of a smaller size that possesses the essential functions of the organ of interest. Although organoids are widely studied, the formation of skeletal elements in an organoid model has not yet been possible. Therefore, we suggest that the formation of skeletal elements using the recombinant limb (RL) assay system can serve as an in vivo organoid model. RLs are formed from undissociated or dissociated-reaggregated undifferentiated mesodermal cells introduced into an ectodermal cover obtained from an early limb bud. Next, this filled ectoderm is grafted into the back of a donor chick embryo. Under these conditions, the cells can receive the nascent embryonic signals and develop complex skeletal elements. We propose that the formation of skeletal elements induced through the RL system may occur from stem cells or other types of progenitors, thus enabling the study of morphogenetic properties in vivo from these cells for the first time.
Collapse
Affiliation(s)
| | | | | | - Jesús Chimal-Monroy
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| |
Collapse
|
14
|
Natesan V, Kim SJ. Metabolic Bone Diseases and New Drug Developments. Biomol Ther (Seoul) 2022; 30:309-319. [PMID: 35342038 PMCID: PMC9252877 DOI: 10.4062/biomolther.2022.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/03/2022] [Accepted: 03/05/2022] [Indexed: 11/05/2022] Open
Abstract
Metabolic bone diseases are serious health issues worldwide, since several million individuals over the age of 50 are at risk of bone damage and should be worried about their bone health. One in every two women and one in every four men will break a bone during their lifetime due to a metabolic bone disease. Early detection, raising bone health awareness, and maintaining a balanced healthy diet may reduce the risk of skeletal fractures caused by metabolic bone diseases. This review compiles information on the most common metabolic bone diseases (osteoporosis, primary hyperparathyroidism, osteomalacia, and fluorosis disease) seen in the global population, including their symptoms, mechanisms, and causes, as well as discussing their prevention and the development of new drugs for treatment. A large amount of research literature suggests that balanced nutrition and balanced periodic supplementation of calcium, phosphate, and vitamin D can improve re-absorption and the regrowth of bones, and inhibit the formation of skeletal fractures, except in the case of hereditary bone diseases. Meanwhile, new and improved drug formulations, such as raloxifene, teriparatide, sclerostin, denosumab, and abaloparatide, have been successfully developed and administered as treatments for metabolic bone diseases, while others (romososumab and odanacatib) are in various stages of clinical trials.
Collapse
Affiliation(s)
- Vijayakumar Natesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - Sung-Jin Kim
- Department of Pharmacology and Toxicology, Metabolic Diseases Research Laboratory, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
Generation of functional oligopeptides that promote osteogenesis based on unsupervised deep learning of protein IDRs. Bone Res 2022; 10:23. [PMID: 35228528 PMCID: PMC8885677 DOI: 10.1038/s41413-022-00193-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/21/2021] [Indexed: 12/30/2022] Open
Abstract
Deep learning (DL) is currently revolutionizing peptide drug development due to both computational advances and the substantial recent expansion of digitized biological data. However, progress in oligopeptide drug development has been limited, likely due to the lack of suitable datasets and difficulty in identifying informative features to use as inputs for DL models. Here, we utilized an unsupervised deep learning model to learn a semantic pattern based on the intrinsically disordered regions of ~171 known osteogenic proteins. Subsequently, oligopeptides were generated from this semantic pattern based on Monte Carlo simulation, followed by in vivo functional characterization. A five amino acid oligopeptide (AIB5P) had strong bone-formation-promoting effects, as determined in multiple mouse models (e.g., osteoporosis, fracture, and osseointegration of implants). Mechanistically, we showed that AIB5P promotes osteogenesis by binding to the integrin α5 subunit and thereby activating FAK signaling. In summary, we successfully established an oligopeptide discovery strategy based on a DL model and demonstrated its utility from cytological screening to animal experimental verification.
Collapse
|
16
|
Liang N, Ren N, Feng Z, Sun Z, Dong M, Wang W, Liu F, Sun C, Zhou W, Xing Z, Wang J, Liu C, Liu H. Biomimetic Metal-Organic Frameworks as Targeted Vehicles to Enhance Osteogenesis. Adv Healthc Mater 2022; 11:e2102821. [PMID: 35182414 DOI: 10.1002/adhm.202102821] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Indexed: 12/22/2022]
Abstract
Although engineered nanoparticles loaded with specific growth factors are used to regulate differentiation of stem cells, the low loading efficiency and biocompatibility are still great challenges in tissue repair. A nature-inspired biomimetic delivery system with targeted functions is attractive for enhancing cell activity and controlling cell fate. Herein, a stem cell membrane (SCM)-wrapped dexamethasone (DEX)-loaded zeolitic imidazolate framework-8 (ZIF-8) is constructed, which integrates the synthetic nanomaterials with native plasma membrane, to achieve efficient DEX delivery and DEX-mediated bone repair. The DEX@ZIF-8-SCM enables high DEX loading capacity, modulates the sustained release, and facilitates the specific uptake of mesenchymal stem cells (MSCs), owing to the porous property of ZIF-8 and the innate targeting capability of SCM. The endocytosed DEX@ZIF-8-SCM shows high cytocompatibility and greatly enhances the osteogenic differentiation of MSCs. Furthermore, RNA-sequencing data reveal that the phosphoinositide 3-kinase (PI3K)-Akt signaling pathways are activated and dominantly involved in the accelerated osteogenesis. In the bone defect model, the administrated DEX@ZIF-8-SCM exerts excellent biocompatibility and effectively promotes bone regeneration. Overall, the SCM-derived biomimetic nanoplatform achieves targeted delivery, excellent biosafety, and enhanced osteogenic differentiation and bone repair, which provides a new and valid strategy for treating various tissue injuries.
Collapse
Affiliation(s)
- Na Liang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Na Ren
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Zhichao Feng
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Zhaoyang Sun
- Department of Oral and Maxillofacial Surgery Qilu Hospital of Shandong University Institute of Stomatology Shandong University Jinan 250012 P. R. China
| | - Mengwei Dong
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Wenhan Wang
- State Key Laboratory of Crystal Materials Shandong University Jinan 250100 P. R. China
| | - Feng Liu
- State Key Laboratory of Crystal Materials Shandong University Jinan 250100 P. R. China
| | - Chunhui Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Weijia Zhou
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Zhiqing Xing
- Ji'nan Pantheum Biological Technology Limited Company Jinan 250100 P. R. China
| | - Jingang Wang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Chao Liu
- Department of Oral and Maxillofacial Surgery Qilu Hospital of Shandong University Institute of Stomatology Shandong University Jinan 250012 P. R. China
| | - Hong Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
- State Key Laboratory of Crystal Materials Shandong University Jinan 250100 P. R. China
| |
Collapse
|
17
|
Cao X, He W, Rong K, Xu S, Chen Z, Liang Y, Han S, Zhou Y, Yang X, Ma H, Qin A, Zhao J. DZNep promotes mouse bone defect healing via enhancing both osteogenesis and osteoclastogenesis. Stem Cell Res Ther 2021; 12:605. [PMID: 34930462 PMCID: PMC8686256 DOI: 10.1186/s13287-021-02670-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022] Open
Abstract
Background Enhancer of zeste homolog 2 (EZH2) is a novel oncogene that can specifically trimethylate the histone H3 lysine 27 (H3K27me3) to transcriptionally inhibit the expression of downstream tumor-suppressing genes. As a small molecular inhibitor of EZH2, 3-Deazaneplanocin (DZNep) has been widely studied due to the role of tumor suppression. With the roles of epigenetic regulation of bone cells emerged in past decades, the property and molecular mechanism of DZNep on enhancing osteogenesis had been reported and attracted a great deal of attention recently. This study aims to elucidate the role of DZNep on EZH2-H3K27me3 axis and downstream factors during both osteoclasts and osteoblasts formation and the therapeutic possibility of DZNep on bone defect healing. Methods Bone marrow-derived macrophages (BMMs) cells were cultured, and their responsiveness to DZNep was evaluated by cell counting kit-8, TRAP staining assay, bone resorption assay, podosome actin belt. Bone marrow-derived mesenchymal stem cells (BMSC) were cultured and their responsiveness to DZNep was evaluated by cell counting kit-8, ALP and AR staining assay. The expression of nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), Wnt signaling pathway was determined by qPCR and western blotting. Mouse bone defect models were created, rescued by DZNep injection, and the effectiveness was evaluated by X-ray and micro-CT and histological staining. Results Consistent with the previous study that DZNep enhances osteogenesis via Wnt family member 1(Wnt1), Wnt6, and Wnt10a, our results showed that DZNep also promotes osteoblasts differentiation and mineralization through the EZH2-H3K27me3-Wnt4 axis. Furthermore, we identified that DZNep promoted the receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast formation via facilitating the phosphorylation of IKKα/β, IκB, and subsequently NF-κB nuclear translocation, which credit to the EZH2-H3K27me3-Foxc1 axis. More importantly, the enhanced osteogenesis and osteoclastogenesis result in accelerated mice bone defect healing in vivo. Conclusion DZNep targeting EZH2-H3K27me3 axis facilitated the healing of mice bone defect via simultaneously enhancing osteoclastic bone resorption and promoting osteoblastic bone formation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02670-6.
Collapse
Affiliation(s)
- Xiankun Cao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Wenxin He
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Shenggui Xu
- Department of Orthopaedics, Mindong Hospital Affiliated to Fujian Medical University, Fuan, 355000, Fujian Province, People's Republic of China
| | - Zhiqian Chen
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yuwei Liang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Shuai Han
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Collaborative Innovation Center for Biomedicine, GuangxiASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Yifan Zhou
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Xiao Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Hui Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
18
|
Park SH, Ju HJ, Ji YB, Shah M, Min BH, Choi HS, Choi S, Kim MS. Endogenous Stem Cell-Based In Situ Tissue Regeneration Using Electrostatically Interactive Hydrogel with a Newly Discovered Substance P Analog and VEGF-Mimicking Peptide. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103244. [PMID: 34480409 DOI: 10.1002/smll.202103244] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/22/2021] [Indexed: 06/13/2023]
Abstract
The use of chemoattractants to promote endogenous stem cell-based in situ tissue regeneration has recently garnered much attention. This study is the first to assess the endogenous stem cell migration using a newly discovered substance P (SP) analog (SP1) by molecular dynamics simulations as an efficient chemoattractant. Further, a novel strategy based on electrostatic interaction using cationic chitosan (Ch) and anionic hyaluronic acid (HA) to prepare an SP1-loaded injectable C/H formulation without SP1 loss is developed. The formulation quickly forms an SP1-loaded C/H hydrogel in situ through in vivo injection. The newly discovered SP1 is found to possess human mesenchymal stromal cells (hMSCs) migration-inducing ability that is approximately two to three times higher than that of the existing SP. The designed VEGF-mimicking peptide (VP) chemically reacts with the hydrogel (C/H-VP) to sustain the release of VP, thus inducing vasculogenic differentiation of the hMSCs that migrate toward the C/H-VP hydrogel. Similarly, in animal experiments, SP1 attracts a large number of hMSCs toward the C/H-VP hydrogel, after which VP induces vasculogenic differentiation. Collectively, these findings indicate that SP1-loaded C/H-VP hydrogels are a promising strategy to facilitate endogenous stem cell-based in situ tissue regeneration.
Collapse
Affiliation(s)
- Seung Hun Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Hyeon Jin Ju
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Yun Bae Ji
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Byoung Hyun Min
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
- Medipolymers, Research Institute, Woncheon Dong 332-2, Suwon, 16522, Korea
| |
Collapse
|
19
|
Li S, Li J, Geng B, Yang X, Song Z, Li Z, Ding B, Zhang J, Lin W, Yan M. TPE based electrochemiluminescence for ALP selective rapid one-step detection applied in vitro. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|