1
|
Bispo DSC, Graça ICR, Jesus CSH, Rodrigues JE, Correia MC, Atella S, Duarte IF, Goodfellow BJ, Oliveira MB, Mano JF, Gil AM. Metabolic markers detect early ostedifferentiation of mesenchymal stem cells from multiple donors. Stem Cell Res Ther 2025; 16:294. [PMID: 40483499 PMCID: PMC12145603 DOI: 10.1186/s13287-025-04419-x] [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] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 05/23/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) are pivotal bioengineering tools, offering significant promise for applications in bone regeneration. However, their therapeutic potential is limited by inter-donor variability and experimental issues. This study aimed to identify robust metabolic markers of osteodifferentiation applicable across multiple donors, while providing insight into the metabolic pathways actively involved in the process. METHODS Untargeted nuclear magnetic resonance (NMR) metabolomics was applied to characterize the intra- and extracellular metabolic adaptations of human adipose-derived MSC (hAMSC) undergoing osteogenic differentiation, compared to proliferation alone. Multivariate and univariate statistical analysis was carried out on data from three independent donors, and cross-validation was employed to evaluate the predictive capacity of the proposed markers. RESULTS Variations in the levels of selected (nine) intracellular and (seventeen) extracellular metabolites detect osteodifferentiation by day 7 (out of 21), with nearly 100% accuracy. These signatures suggest a metabolic shift from glycolysis/OxPhos to lactic fermentation, fatty acid β-oxidation and phosphocreatine hydrolysis. Intracellular glucose, lactate, citrate and specific amino acids are redirected towards protein synthesis and glycosylation, with some of the secreted metabolites (e.g., citrate) seemingly involved in biomineralization and other extracellular roles. Membrane metabolism, antioxidant mechanisms and adenosine metabolism are also impacted by osteodifferentiation. CONCLUSIONS These findings reveal effective donor-independent markers of hAMSC osteodifferentiation, with a robust extracellular signature standing out for potential rapid and non-invasive detection of osteocommitted cells.
Collapse
Affiliation(s)
- Daniela S C Bispo
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Inês C R Graça
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Catarina S H Jesus
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - João E Rodrigues
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Marlene C Correia
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Sabrina Atella
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Iola F Duarte
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Brian J Goodfellow
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal.
| | - Ana M Gil
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro, 3810-193, Portugal.
| |
Collapse
|
2
|
Feng W, Zeng S, Liu D, Gong W, Hu J, Xu W, Ma Z, Fu S, Chen X. Single-cell dynamic RNA and glycosylation sequencing reveals the mechanism underlying the differentiation of pluripotent stem cells into hematopoietic stem cells. Hum Cell 2025; 38:110. [PMID: 40425941 PMCID: PMC12116958 DOI: 10.1007/s13577-025-01234-7] [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] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 05/09/2025] [Indexed: 05/29/2025]
Abstract
Studying the mechanism of hematopoietic stem cells' generation from induced pluripotent stem cells in vitro can be useful for understanding embryonic hematopoiesis, as well as for the application of related cell therapy. This study aimed to delineate the process of the differentiation of induced pluripotent stem cells into hematopoietic stem cells' models and provide a theoretical basis and clinical value for the production of hematopoietic stem cells in vitro. We analyzed the differentiation model by single-cell dynamic transcriptome and glycosylation sequencing, which was divided into three differentiation stages based on the new-to-total RNA ratio and glycosylation level. Two differentiation fates were found in the pseudo-time, including hematopoietic development and other tissue development. Precursor hematopoietic cells with a high glycosylation level greatly expressed hematopoietic regulation and vascular endothelial genes, suggesting that glycosylation is associated with angiogenesis and hematopoietic regulation. The multiple differentiation events in the in vitro model are similar to those in hematopoietic development in vivo, including yolk sac hematopoiesis, cellular communication between non-potential hematopoietic subsets and potential hematopoietic subsets, gene expression, and temporal deviations in hematopoietic fate. Our study has revealed the similar hematopoiesis process in the differentiation model via single-cell dynamic RNA and glycosylation sequencing, which provides an important theoretical basis for the study of hematopoietic stem cell development.
Collapse
Affiliation(s)
- Wanyi Feng
- School of Life and Health Sciences, Hainan University, 58th People's Avenue, Haikou, Hainan, China
- Affiliated Cancer Hospital of Hainan Medical University, 4th Changbin West Street, Haikou, 570100, Hainan, China
| | - Sheng Zeng
- Susheng Biotech (Hainan) Co., Ltd, 1st Medicine Valley Road, Haikou, Hainan, China
| | - Donghui Liu
- Susheng Biotech (Hainan) Co., Ltd, 1st Medicine Valley Road, Haikou, Hainan, China
| | - Wei Gong
- Affiliated Cancer Hospital of Hainan Medical University, 4th Changbin West Street, Haikou, 570100, Hainan, China
| | - Junjie Hu
- Affiliated Cancer Hospital of Hainan Medical University, 4th Changbin West Street, Haikou, 570100, Hainan, China
- Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Weihua Xu
- Affiliated Cancer Hospital of Hainan Medical University, 4th Changbin West Street, Haikou, 570100, Hainan, China
- Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Zhichao Ma
- Affiliated Cancer Hospital of Hainan Medical University, 4th Changbin West Street, Haikou, 570100, Hainan, China
- Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Shengmiao Fu
- Academician Innovation Platform of Hainan Province, Haikou, Hainan, China
| | - Xinping Chen
- School of Life and Health Sciences, Hainan University, 58th People's Avenue, Haikou, Hainan, China.
- Affiliated Cancer Hospital of Hainan Medical University, 4th Changbin West Street, Haikou, 570100, Hainan, China.
- Academician Innovation Platform of Hainan Province, Haikou, Hainan, China.
| |
Collapse
|
3
|
Liu M, Hu Y, You C, Xiong D, Ye L, Shi Y. O-GlcNAcylation in Gli1 + Mesenchymal Stem Cells Is Indispensable for Bone Formation and Fracture Healing. Int J Mol Sci 2025; 26:2712. [PMID: 40141354 PMCID: PMC11943158 DOI: 10.3390/ijms26062712] [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] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Adult mesenchymal stem cells (MSCs) play a crucial role in maintaining bone health and promoting regeneration. In our previous research, we identified Gli1+ MSCs as key contributors to the formation of most trabecular bone in adulthood and as essential for healing bicortical fractures. However, the mechanisms behind the maintenance and differentiation of Gli1+ MSCs are still not fully understood. O-linked N-acetylglucosamine modification (O-GlcNAcylation), mediated by O-GlcNAc glycosyltransferase (OGT), is involved in various biological processes and diseases. Our earlier work also demonstrated that O-GlcNAcylation is necessary for Wnt-stimulated bone formation. Nonetheless, the specific functions of O-GlcNAcylation in MSCs have not been completely elucidated. In this study, we found that the absence of OGT in Gli1+ MSCs led to a decrease in O-GlcNAcylation, which impaired both the bone formation and regeneration following fractures. Mechanistically, the Hedgehog signaling pathway induced O-GlcNAcylation through the insulin-like growth factor (Igf)-mTORC2 axis. This process stabilized the Gli2 protein at a specific site Ser355 and promoted osteogenesis in MSCs in vitro. Our findings reveal a significant mechanism by which O-GlcNAcylation regulates bone development and repair in mammals.
Collapse
Affiliation(s)
- Moyu Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (M.L.); (Y.H.); (C.Y.); (D.X.); (L.Y.)
| | - Yujie Hu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (M.L.); (Y.H.); (C.Y.); (D.X.); (L.Y.)
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chengjia You
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (M.L.); (Y.H.); (C.Y.); (D.X.); (L.Y.)
| | - Ding Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (M.L.); (Y.H.); (C.Y.); (D.X.); (L.Y.)
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (M.L.); (Y.H.); (C.Y.); (D.X.); (L.Y.)
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (M.L.); (Y.H.); (C.Y.); (D.X.); (L.Y.)
| |
Collapse
|
4
|
Xiao Y, Yue Z, Zijing H, Yao Z, Sui M, Xuemin Z, Qiang Z, Xiao Y, Dapeng R. Mechanical compression induces chondrocyte hypertrophy by regulating Runx2 O-GlcNAcylation during temporomandibular joint condyle degeneration. Bone Joint Res 2025; 14:209-222. [PMID: 40058403 PMCID: PMC11890221 DOI: 10.1302/2046-3758.143.bjr-2024-0257.r1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Aims Excessive chondrocyte hypertrophy is a common feature in cartilage degeneration which is susceptible to joint overloading, but the relationship between mechanical overloading and chondrocyte hypertrophy still remains elusive. The aim of our study was to explore the mechanism of mechanical compression-induced chondrocyte hypertrophy. Methods In this study, the temporomandibular joint (TMJ) degeneration model was built through forced mandibular retrusion (FMR)-induced compression in TMJ. Chondrocytes were also mechanically compressed in vitro. The role of O-GlcNAcylation in mechanical compression-induced chondrocyte hypertrophy manifested through specific activator Thiamet G and inhibitor OSMI-1. Results Both in vivo and in vitro data revealed that chondrocyte hypertrophic differentiation is promoted by compression. Immunofluorescent and immunoblotting results showed that protein pan-O-GlcNAcylation levels were elevated in these hypertrophic chondrocytes. Pharmacologically inhibiting protein pan-O-GlcNAcylation by OSMI-1 partially mitigated the compression-induced hypertrophic differentiation of chondrocytes. Specifically, runt-related transcription factor 2 (Runx2) and SRY-Box 9 transcription factor (Sox9) were subjected to modification of O-GlcNAcylation under mechanical compression, and pharmacological activation or inhibition of O-GlcNAcylation affected the transcriptional activity of Runx2 but not Sox9. Furthermore, compression-induced protein pan-O-GlcNAcylation in chondrocytes was induced by enhanced expression of glucose transporter 1 (GLUT1), and depletion of GLUT1 by WZB117 dampened the effect of compression on chondrocyte hypertrophy. Conclusion Our study proposes a novel function of GLUT1-mediated protein O-GlcNAcylation in driving compression-induced hypertrophic differentiation of chondrocytes by O-GlcNAc modification of Runx2, which promoted its transcriptional activity and strengthened the expressions of downstream hypertrophic marker.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhang Yue
- Department of Orthodontics, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - He Zijing
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zheng Yao
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mao Sui
- College of Materials Science and Engineering, Qingdao University, Qingdao, China
| | - Zeng Xuemin
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhang Qiang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuan Xiao
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ren Dapeng
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
5
|
Du Y, Gao X, Chen J, Chen X, Liu H, He W, Liu L, Jiang Y, He B, Deng Z, Liang C, Guo F. OGT mediated HDAC5 O-GlcNAcylation promotes osteogenesis by regulating the homeostasis of epigenetic modifications and proteolysis. J Orthop Translat 2025; 50:14-29. [PMID: 39659899 PMCID: PMC11626777 DOI: 10.1016/j.jot.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024] Open
Abstract
Background O-GlcNAc transferase (OGT) is responsible for attaching O-linked N-acetylglucosamine (O-GlcNAc) to proteins, regulating diverse cellular processes ranging from transcription and translation to signaling and metabolism. This study focuses on the role and mechanisms of OGT in osteogenesis. Materials and methods We found that OGT is downregulated in osteoporosis by bioinformatics analysis, determined its role in osteogenic differentiation by using OGT inhibitors (or OGA inhibitors) as well as conditional knockout OGT mice in vitro and in vivo, and explored and specific mechanisms by quantitative proteomic analysis and RNA-seq, qRT-PCR, western blotting, immunofluorescence, H&E, ALP, ARS, Masson staining, IHC, micro CT, etc. Results we revealed that OGT positively influenced osteogenesis and osteoblast differentiation in vitro as well as ovariectomy (OVX) mice in vivo. Consistently, mice with conditionally depleted OGT exhibited a reduction in bone mass, while O-GlcNAcylation enhancer could partially recover bone mass in ovariectomy (OVX) mice. Mechanistically, quantitative proteomic analysis and high-throughput RNAseq further reveals that HDAC5 is one of the endogenous O-GlcNAcylation substrates, and O-GlcNAcylation of HDAC5 on Thr934 promotes its translocation to lysosomes and subsequent degradation, thus, elevating the O-GlcNAcylation level of HDAC5 leads to its cytoplasmic cleavage, consequently diminished its nuclear entry and enhanced DNA transcription. The OGT-mediated O-GlcNAcylation of HDAC5 modulates the balance between its cytoplasmic proteolysis and nuclear entry, thereby impacting the Notch signaling pathway and DNA epigenetic modifications then playing a role in osteogenesis. Conclusion OGT is a regulator that promotes osteoblast differentiation and bone regeneration. Additionally, it highlights the critical function of HDAC5 O-GlcNAcylation in controlling epigenetics. This study offers fresh perspectives on osteogenesis and O-GlcNAcylation, proposing that the OGT-mediated O-GlcNAcylation of HDAC5 could be a promising target for osteoporosis treatment. The translational potential of this article On one side, OGT might potentially be used as a new biomarker for clinical diagnosis of osteoporosis (OP) in the future. On the other side, small molecule inhibitors of HDAC5, a glycosylation substrate of OGT, or OGT agonists such as silymarin, could all potentially serve as therapeutic targets for the prevention or treatment of OP in the future.
Collapse
Affiliation(s)
- Yu Du
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xiang Gao
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jianqiang Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Hang Liu
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Wenge He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Lu Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yue Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Baicheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zhongliang Deng
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Fengjin Guo
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Zhang Z, Zhou C, Yu L. LEP O-GlcNAcylation inactivates NF-κB pathway by suppressing LEP protein level and thus mediates cellular senescence and osteogenic differentiation in mouse mesenchymal stem cells. BMC Mol Cell Biol 2024; 25:26. [PMID: 39695926 DOI: 10.1186/s12860-024-00523-7] [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: 05/15/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Cellular senescence is a key driver of decreased bone formation and osteoporosis. Leptin (LEP) has been implicated in cellular senescence and osteogenic differentiation. The aim of this study was to investigate the mechanisms by which LEP mediates cellular senescence and osteogenic differentiation. METHODS C3H10T1/2 cells were treated with etoposide to induce cellular senescence, which was assessed by β-galactosidase staining. Quantitative real-time PCR and western blotting were used to measure the levels of senescence markers p21 and p16, as well as osteogenic differentiation-related genes ALP, COL1A1, and RUNX2. Alkaline phosphatase (ALP) staining and alizarin red S staining were performed to evaluate osteogenic differentiation. The NF-κB pathway and O-GlcNAcylation were assessed by western blotting. RESULTS Etoposide treatment increased the number of senescent cells and the levels of p21 and p16, along with elevated LEP expression. These effects were reversed by LEP knockdown. Additionally, LEP knockdown increased ALP staining density and osteoblast mineralization nodules, as well as the mRNA and protein levels of ALP, COL1A1, and RUNX2, indicating that LEP knockdown promoted osteogenic differentiation in C3H10T1/2 cells. Mechanistically, LEP knockdown inactivated the NF-κB pathway by inhibiting the nuclear translocation of p65. Furthermore, OGT was found to promote O-GlcNAcylation of LEP at the S50 site. CONCLUSION Our findings demonstrated that O-GlcNAcylation of LEP inactivated the NF-κB pathway by reducing LEP protein levels, thereby inhibiting cellular senescence and promoting osteogenic differentiation in C3H10T1/2 cells. This study may provide a novel therapeutic target for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zhuang Zhang
- Macau University of Science and Technology, Faculty of Chinese Medicine, E205, Avenida Wai Long, Taipa, Macau, 999078, China
- The 2nd People's Hospital of Zhuhai, Zhuhai, China
| | - Chaoqing Zhou
- Department of Traumatology, The 2nd People's Hospital of Zhuhai, No.208 Yuehua Road, Zhuhai, Guangdong, 519020, China.
| | - Lili Yu
- Macau University of Science and Technology, Faculty of Chinese Medicine, E205, Avenida Wai Long, Taipa, Macau, 999078, China.
| |
Collapse
|
7
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [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: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
8
|
You C, Shen F, Yang P, Cui J, Ren Q, Liu M, Hu Y, Li B, Ye L, Shi Y. O-GlcNAcylation mediates Wnt-stimulated bone formation by rewiring aerobic glycolysis. EMBO Rep 2024; 25:4465-4487. [PMID: 39256595 PMCID: PMC11467389 DOI: 10.1038/s44319-024-00237-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Wnt signaling is an important target for anabolic therapies in osteoporosis. A sclerostin-neutralizing antibody (Scl-Ab), that blocks the Wnt signaling inhibitor (sclerostin), has been shown to promote bone mass in animal models and clinical studies. However, the cellular mechanisms by which Wnt signaling promotes osteogenesis remain to be further investigated. O-GlcNAcylation, a dynamic post-translational modification of proteins, controls multiple critical biological processes including transcription, translation, and cell fate determination. Here, we report that Wnt3a either induces O-GlcNAcylation rapidly via the Ca2+-PKA-Gfat1 axis, or increases it in a Wnt-β-catenin-dependent manner following prolonged stimulation. Importantly, we find O-GlcNAcylation indispensable for osteoblastogenesis both in vivo and in vitro. Genetic ablation of O-GlcNAcylation in the osteoblast-lineage diminishes bone formation and delays bone fracture healing in response to Wnt stimulation in vivo. Mechanistically, Wnt3a induces O-GlcNAcylation at Serine 174 of PDK1 to stabilize the protein, resulting in increased glycolysis and osteogenesis. These findings highlight O-GlcNAcylation as an important mechanism regulating Wnt-induced glucose metabolism and bone anabolism.
Collapse
Affiliation(s)
- Chengjia You
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fangyuan Shen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Puying Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiaoyue Ren
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Moyu Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujie Hu
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boer Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yu Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Li S, Ren W, Zheng J, Li S, Zhi K, Gao L. Role of O-linked N-acetylglucosamine protein modification in oxidative stress-induced autophagy: a novel target for bone remodeling. Cell Commun Signal 2024; 22:358. [PMID: 38987770 PMCID: PMC11238385 DOI: 10.1186/s12964-024-01734-3] [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: 04/06/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024] Open
Abstract
O-linked N-acetylglucosamine protein modification (O-GlcNAcylation) is a dynamic post-translational modification (PTM) involving the covalent binding of serine and/or threonine residues, which regulates bone cell homeostasis. Reactive oxygen species (ROS) are increased due to oxidative stress in various pathological contexts related to bone remodeling, such as osteoporosis, arthritis, and bone fracture. Autophagy serves as a scavenger for ROS within bone marrow-derived mesenchymal stem cells, osteoclasts, and osteoblasts. However, oxidative stress-induced autophagy is affected by the metabolic status, leading to unfavorable clinical outcomes. O-GlcNAcylation can regulate the autophagy process both directly and indirectly through oxidative stress-related signaling pathways, ultimately improving bone remodeling. The present interventions for the bone remodeling process often focus on promoting osteogenesis or inhibiting osteoclast absorption, ignoring the effect of PTM on the overall process of bone remodeling. This review explores how O-GlcNAcylation synergizes with autophagy to exert multiple regulatory effects on bone remodeling under oxidative stress stimulation, indicating the application of O-GlcNAcylation as a new molecular target in the field of bone remodeling.
Collapse
Affiliation(s)
- Shengqian Li
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
| | - Jingjing Zheng
- Department of Endodontics, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Shaoming Li
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
- School of Stomatology, Qingdao University, Qingdao, 266003, China.
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
| | - Ling Gao
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
10
|
Yang Y, Zhou X, Deng H, Chen L, Zhang X, Wu S, Song A, Liang F. The role of O-GlcNAcylation in bone metabolic diseases. Front Physiol 2024; 15:1416967. [PMID: 38915778 PMCID: PMC11194333 DOI: 10.3389/fphys.2024.1416967] [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: 04/13/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
O-GlcNAcylation, as a post-translational modification, can modulate cellular activities such as kinase activity, transcription-translation, protein degradation, and insulin signaling by affecting the function of the protein substrate, including cellular localization of proteins, protein stability, and protein/protein interactions. Accumulating evidence suggests that dysregulation of O-GlcNAcylation is associated with disease progression such as cancer, neurodegeneration, and diabetes. Recent studies suggest that O-GlcNAcylation is also involved in the regulation of osteoblast, osteoclast and chondrocyte differentiation, which is closely related to the initiation and development of bone metabolic diseases such as osteoporosis, arthritis and osteosarcoma. However, the potential mechanisms by which O-GlcNAcylation regulates bone metabolism are not fully understood. In this paper, the literature related to the regulation of bone metabolism by O-GlcNAcylation was summarized to provide new potential therapeutic strategies for the treatment of orthopedic diseases such as arthritis and osteoporosis.
Collapse
Affiliation(s)
- Yajing Yang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- School of Medicine, Xiamen University, Xiamen, China
| | - HuiLi Deng
- School of Medicine, Xiamen University, Xiamen, China
| | - Li Chen
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Xiaolin Zhang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Song Wu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Aiqun Song
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Fengxia Liang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| |
Collapse
|
11
|
Yan X, Zheng J, Ren W, Li S, Yang S, Zhi K, Gao L. O-GlcNAcylation: roles and potential therapeutic target for bone pathophysiology. Cell Commun Signal 2024; 22:279. [PMID: 38773637 PMCID: PMC11106977 DOI: 10.1186/s12964-024-01659-x] [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: 02/23/2024] [Accepted: 05/10/2024] [Indexed: 05/24/2024] Open
Abstract
O-linked N-acetylglucosamine (O-GlcNAc) protein modification (O-GlcNAcylation) is a critical post-translational modification (PTM) of cytoplasmic and nuclear proteins. O-GlcNAcylation levels are regulated by the activity of two enzymes, O-GlcNAc transferase (OGT) and O‑GlcNAcase (OGA). While OGT attaches O-GlcNAc to proteins, OGA removes O-GlcNAc from proteins. Since its discovery, researchers have demonstrated O-GlcNAcylation on thousands of proteins implicated in numerous different biological processes. Moreover, dysregulation of O-GlcNAcylation has been associated with several pathologies, including cancers, ischemia-reperfusion injury, and neurodegenerative diseases. In this review, we focus on progress in our understanding of the role of O-GlcNAcylation in bone pathophysiology, and we discuss the potential molecular mechanisms of O-GlcNAcylation modulation of bone-related diseases. In addition, we explore significant advances in the identification of O-GlcNAcylation-related regulators as potential therapeutic targets, providing novel therapeutic strategies for the treatment of bone-related disorders.
Collapse
Affiliation(s)
- Xiaohan Yan
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao District, Qingdao, 266555, Shandong, China
| | - Jingjing Zheng
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
- Department of Endodontics, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao District, Qingdao, 266555, Shandong, China
| | - Shaoming Li
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao District, Qingdao, 266555, Shandong, China
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Keqian Zhi
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
- School of Stomatology, Qingdao University, Qingdao, 266003, China.
- Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao District, Qingdao, 266555, Shandong, China.
| | - Ling Gao
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
- School of Stomatology, Qingdao University, Qingdao, 266003, China.
- Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao District, Qingdao, 266555, Shandong, China.
| |
Collapse
|
12
|
Xie A, Wang J, Liu Y, Li G, Yang N. Impacts of β-1, 3-N-acetylglucosaminyltransferases (B3GNTs) in human diseases. Mol Biol Rep 2024; 51:476. [PMID: 38553573 DOI: 10.1007/s11033-024-09405-9] [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: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 04/02/2024]
Abstract
Glycosylation modification of proteins is a common post-translational modification that exists in various organisms and has rich biological functions. It is usually catalyzed by multiple glycosyltransferases located in the Golgi apparatus. β-1,3-N-acetylglucosaminyltransferases (B3GNTs) are members of the glycosyltransferases and have been found to be involved in the occurrence and development of a variety of diseases including autoimmunity diseases, cancers, neurodevelopment, musculoskeletal system, and metabolic diseases. The functions of B3GNTs represent the glycosylation of proteins is a crucial and frequently life-threatening step in progression of most diseases. In this review, we give an overview about the roles of B3GNTs in tumor, nervous system, musculoskeletal and metabolic diseases, describing the recent results about B3GNTs, in order to provide a research direction and exploration value for the prevention, diagnosis and treatment of these diseases.
Collapse
Affiliation(s)
- Anna Xie
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jingjing Wang
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yi Liu
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Guoqing Li
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Nanyang Yang
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
13
|
Zhou XC, Ni GX. O-linked β-N-acetylglucosaminylation may be a key regulatory factor in promoting osteogenic differentiation of bone marrow mesenchymal stromal cells. World J Stem Cells 2024; 16:228-231. [PMID: 38577231 PMCID: PMC10989286 DOI: 10.4252/wjsc.v16.i3.228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/29/2024] [Indexed: 03/25/2024] Open
Abstract
Cumulative evidence suggests that O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) plays an important regulatory role in pathophysiological processes. Although the regulatory mechanisms of O-GlcNAcylation in tumors have been gradually elucidated, the potential mechanisms of O-GlcNAcylation in bone metabolism, particularly, in the osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs) remains unexplored. In this study, the literature related to O-GlcNAcylation and BMSC osteogenic differentiation was reviewed, assuming that it could trigger more scholars to focus on research related to O-GlcNAcylation and bone metabolism and provide insights into the development of novel therapeutic targets for bone metabolism disorders such as osteoporosis.
Collapse
Affiliation(s)
- Xu-Chang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Guo-Xin Ni
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, Fujian Province, China.
| |
Collapse
|
14
|
Shao Y, Xiong M, Liu J, Gu Z, Wu Z, Cao L. LOC646762 Is Involved in Adipogenic Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells. ACS OMEGA 2024; 9:8464-8470. [PMID: 38405496 PMCID: PMC10882647 DOI: 10.1021/acsomega.3c09684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/27/2024]
Abstract
Long noncoding RNA (lncRNA) has been shown to participate in adipogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). In this study, we aimed to investigate the role of lncRNA-LOC646762 in adipogenic differentiation of BMSCs. Transcriptome sequencing revealed a positive correlation between LOC646762 transcription and expression of adipogenic marker genes in adipogenic differentiation. Moreover, LOC646762 overexpression did not negatively impact the cell proliferation of BMSCs. Besides, LOC646762 plays a crucial role in adipogenic differentiation, as evidenced by its positive correlation with adipogenic marker gene expression. Its possible interaction with its proposed target C/EBPβ suggests its involvement in essential pathways governing adipogenesis. Collectively, our study outcomes provide valuable insights into the molecular mechanisms underlying the adipogenic differentiation of BMSCs and lay a strong foundation for further research in regenerative medicine.
Collapse
Affiliation(s)
- Yifan Shao
- Jiujiang
City Key Laboratory of Cell Therapy, The
First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Minqi Xiong
- The
Chinese University of Hong Kong, Shenzhen 518000, China
| | - Jianyun Liu
- Key
Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang 332000, China
| | - Zhiping Gu
- Jiujiang
City Key Laboratory of Cell Therapy, The
First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Zhaoping Wu
- Jiujiang
City Key Laboratory of Cell Therapy, The
First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Lingling Cao
- Jiujiang
City Key Laboratory of Cell Therapy, The
First Hospital of Jiujiang City, Jiujiang 332000, China
| |
Collapse
|
15
|
Yu B, Li Q, Sun X, Yin Y. O-GlcNAcylation of TLR4 inhibits osteogenic differentiation of periodontal ligament stem cells. J Periodontal Res 2024; 59:119-127. [PMID: 37817319 DOI: 10.1111/jre.13193] [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: 07/11/2023] [Revised: 08/28/2023] [Accepted: 09/21/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Toll-like receptor 4 (TLR4)-mediated inflammatory responses are associated with diabetes and periodontitis, which are dysregulated by O-GlcNAcylation. OBJECTIVE This study aimed to investigate the effects of O-GlcNAc transferase (OGT)-mediated TLR4 O-GlcNAcylation on the osteogenesis of periodontal ligament stem cells (PDLCs). METHODS PDLCs were treated with high glucose (HG) to establish a cell model. Osteogenic differentiation was evaluated using western blotting, an alkaline phosphatase activity assay, and an alizarin red S staining assay. The regulation of OGT on the O-GlcNAcylation of TLR4 was analyzed using co-immunoprecipitation, immunoprecipitation, western blotting, and immunofluorescence staining. RESULTS The results showed that HG inhibited osteogenic differentiation and promoted inflammatory response. Knockdown of OGT promoted osteogenic differentiation of HG-treated PDLCs. OGT interacted with TLR4 and increased the O-GlcNAcylation and protein levels of TLR4 in the cytomembrane of PDLCs. Moreover, silenced TLR4 reversed the effects on osteogenic differentiation induced by OGT in HG-treated PDLCs. CONCLUSION O-GlcNAcylation of TLR4 induced by OGT suppresses osteogenic differentiation of PDLCs after HG stimulation. The findings suggest a promising strategy for treating DP.
Collapse
Affiliation(s)
- Bohan Yu
- Department of Periodontics, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, ShangHai, China
| | - Qin Li
- Department of Periodontics, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, ShangHai, China
| | - Xi Sun
- Department of Periodontics, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, ShangHai, China
| | - Yue Yin
- Department of Periodontics, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, ShangHai, China
| |
Collapse
|
16
|
Ye L, Ding W, Xiao D, Jia Y, Zhao Z, Ao X, Wang J. O-GlcNAcylation: cellular physiology and therapeutic target for human diseases. MedComm (Beijing) 2023; 4:e456. [PMID: 38116061 PMCID: PMC10728774 DOI: 10.1002/mco2.456] [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: 07/02/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
O-linked-β-N-acetylglucosamine (O-GlcNAcylation) is a distinctive posttranslational protein modification involving the coordinated action of O-GlcNAc transferase and O-GlcNAcase, primarily targeting serine or threonine residues in various proteins. This modification impacts protein functionality, influencing stability, protein-protein interactions, and localization. Its interaction with other modifications such as phosphorylation and ubiquitination is becoming increasingly evident. Dysregulation of O-GlcNAcylation is associated with numerous human diseases, including diabetes, nervous system degeneration, and cancers. This review extensively explores the regulatory mechanisms of O-GlcNAcylation, its effects on cellular physiology, and its role in the pathogenesis of diseases. It examines the implications of aberrant O-GlcNAcylation in diabetes and tumorigenesis, highlighting novel insights into its potential role in cardiovascular diseases. The review also discusses the interplay of O-GlcNAcylation with other protein modifications and its impact on cell growth and metabolism. By synthesizing current research, this review elucidates the multifaceted roles of O-GlcNAcylation, providing a comprehensive reference for future studies. It underscores the potential of targeting the O-GlcNAcylation cycle in developing novel therapeutic strategies for various pathologies.
Collapse
Affiliation(s)
- Lin Ye
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Wei Ding
- The Affiliated Hospital of Qingdao UniversityQingdao Medical CollegeQingdao UniversityQingdaoChina
| | - Dandan Xiao
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Yi Jia
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Zhonghao Zhao
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Xiang Ao
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Jianxun Wang
- School of Basic MedicineQingdao UniversityQingdaoChina
| |
Collapse
|