Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Mar 26, 2024; 16(3): 228-231
Published online Mar 26, 2024. doi: 10.4252/wjsc.v16.i3.228
O-linked β-N-acetylglucosaminylation may be a key regulatory factor in promoting osteogenic differentiation of bone marrow mesenchymal stromal cells
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
ORCID number: Xu-Chang Zhou (0000-0003-1390-7659); Guo-Xin Ni (0000-0001-9181-8155).
Author contributions: Zhou XC and Ni GX designed and coordinated the study; Zhou XC wrote the manuscript; and all authors approved the final version of the article.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Guo-Xin Ni, Doctor, MD, PhD, Chief Doctor, Chief Physician, Professor, Department of Rehabilitation Medicine, The First Affiliated Hospital of Xiamen University, No. 55 Zhenhai Road, Siming District, Xiamen 361003, Fujian Province, China. nigx@xmu.edu.cn
Received: December 7, 2023
Peer-review started: December 7, 2023
First decision: January 29, 2024
Revised: February 2, 2024
Accepted: February 29, 2024
Article in press: February 29, 2024
Published online: March 26, 2024
Processing time: 108 Days and 10.8 Hours

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.

Key Words: O-GlcNAcylation, Osteogenic differentiation, Bone marrow mesenchymal stromal cells, Osteoporosis

Core Tip: O-linked β-N-acetylglucosaminylation (O-GlcNAcylation), an important post-translational modification of proteins, widely involved in the regulation of biological processes such as signal transduction and proteasomal degradation, plays an essential role in the initiation and progression of various diseases such as bone metabolism. In this study, we emphasized that maintaining appropriate levels of O-GlcNAcylation is beneficial for the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Insufficient or excessive levels of O-GlcNAcylation are detrimental to BMSC osteogenic differentiation.



INTRODUCTION

Bone marrow mesenchymal stromal cells (BMSCs), important precursors of osteoblastic lineage cells, are pluripotent stem cells with self-renewal, immunomodulatory, and multidifferentiation potentials[1]. As the major source of osteoblasts, BMSCs are important contributors to the bone tissue repair process. The abnormal osteogenic differentiation of BMSCs is an important cause of bone metabolism-related diseases, including osteoporosis[2,3]. O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) is an important post-translational modification in which involves the attachment of a single O-linked N-acetylglucosamine (O-GlcNAc) moiety to Ser or Thr residues of cytoplasmic, nuclear, and mitochondrial proteins. O-GlcNAcylation can regulate fundamental cellular processes ranging from gene transcription and translation to protein localization, interaction, and degradation[4]. The donor for O-GlcNAcylation is a nucleoside sugar, such as uridine diphosphate GlcNAc (UDP-GlcNAc). UDP-GlcNAc, a key metabolite produced by the hexosamine biosynthetic pathway, is synthesized by consumption of uridine triphosphate, glucose, glutamine, and acetyl-CoA[5]. As a ubiquitous post-translational modification of proteins, O-GlcNAcylation is regulated by two conserved enzymes: O-GlcNAc transferase (OGT), which can add O-GlcNAc to proteins, and O-GlcNAc enzyme (OGA), which can remove O-GlcNAc from proteins. O-GlcNAcylation maintains optimal homeostatic balance through mutual regulation of OGT and OGA[4]. However, uncoupled OGT and OGA homeostasis have been shown to be associated with the pathogenesis of multiple human diseases, including bone metabolic diseases. Emerging evidence shows that O-GlcNAc modification is closely related to the osteogenic differentiation of BMSCs[6].

BMSCs have the potential to differentiate into osteoblasts, adipocytes, and chondrocytes[7,8]. A recent study showed that OGT knockout in mouse BMSCs inhibited bone formation while promoting bone marrow adipogenesis[9] , indicating that O-GlcNACylation may be a key regulatory factor affecting the differentiation fate of BMSCs. Runt-related transcription factor 2 (RUNX2) is a member of the polyomavirus enhancer-binding protein 2/core-binding factor superfamily[10,11]. The balance between osteogenesis and adipogenic differentiation in BMSC is coordinated regulated by transcription factors Runx2 and CCAAT/enhancer-binding protein beta (C/EBPβ) through O-GlcNAc post-translational modifications. The increased O-glycosylation of Runx2 is not only critical for osteogenic differentiation, but also promotes B lymphocytes by activating interleukin-7. Knockdown of OGT can activate the transcriptional activity of C/EBPβ to promote the adipogenic differentiation of BMSCs[12,13], and upregulate the expression of myelopoietic stem cell factor encoded by the Kitl gene, thereby increasing myopoiesis[14-17]. In addition, Kim et al[6] observed that elevated protein O-GlcNAc modification enhances the binding of Runx2 to Ose2 by promoting the transcriptional activity of Runx2 and inducing an increase in the expression of the osteoblast-specific marker osteocalcin (OCN)[18-21]. Another study reported that the osteogenic differentiation marker bone morphogenetic protein 2/7 reduced OGA activity[18]. During osteogenic differentiation process of BMSC, the overall level of O-GlcNACylation increases. Pharmacological inhibition of OGA promotes the expression of osteogenic differentiation makers, including alkaline phosphatase (ALP), OCN, and bone sialoprotein[6,18,22,23].

Hyperglycemia is reported to be closely related to bone formation inhibition and is a major factor in diabetic osteoporosis[24-27]. Previous studies have shown that high blood sugar levels increase the O-GlcNAcylation of proteins. Abnormal regulation of O-GlcNAcylation is closely associated with the pathogenesis of diabetes mellitus[28]. Therefore, hyperglycemia-induced excessive and abnormal O-GlcNAcylation may lead to reduced osteogenic differentiation and diabetic osteoporosis. Gu et al[29] demonstrated that excessive O-GlcNAcylation induced by high glucose, glucosamine, or GlcNAc treatment or OGT overexpression can reduce the expression levels of osteoblast markers, such as ALP, type I collagen, OCN, Runx2, and osterix, thereby inhibiting osteogenic differentiation. These results are consistent with the phenotypic reduction in bone formation observed in patients with type 2 diabetes. However, other studies have shown that the upregulation of O-GlcNAcylation through supplementation with OGA inhibitors promotes osteogenic differentiation and increases Runx2 transcriptional activity and matrix mineralization[6,18]. One explanation for aforementioned difference is that the effects of metabolic treatment (high concentration glucose treatment) and drug treatment (OGA inhibitors) may be different. Pharmacological inhibition of OGA increases the O-GlcNAcylation level by breaking the dynamic on/off cycle, whereas metabolic treatment or OGT overexpression increases the O-GlcNAcylation level by shifting the balance toward modification[30].

CONCLUSION

The osteogenic differentiation of BMSCs requires a moderate increase of O-GlcNAcylation, and an excessive increase in overall O-GlcNAcylation may inhibit the osteogenic differentiation of BMSCS. Therefore, the overall O-GlcNAcylation level should be maintained within an optimal range to protect normal cellular functions. The precise regulation of O-GlcNAcylation may be an effective strategy for promoting the osteogenic differentiation of BMSCs, correcting abnormal bone metabolism, and preventing bone-related diseases. Further elucidation of the potential regulatory mechanism between O-GlcNAcylation and the osteogenic differentiation of BMSCs will help to better understand the pathogenesis of bone metabolic diseases and provide novel ideas for the treatment and prevention of bone metabolic diseases.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Naik D, India S-Editor: Wang JJ L-Editor: A P-Editor: Yuan YY

References
1.  Zhang X, Cao D, Xu L, Xu Y, Gao Z, Pan Y, Jiang M, Wei Y, Wang L, Liao Y, Wang Q, Yang L, Xu X, Gao Y, Gao S, Wang J, Yue R. Harnessing matrix stiffness to engineer a bone marrow niche for hematopoietic stem cell rejuvenation. Cell Stem Cell. 2023;30:378-395.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 14]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
2.  Jensen PR, Andersen TL, Chavassieux P, Roux JP, Delaisse JM. Bisphosphonates impair the onset of bone formation at remodeling sites. Bone. 2021;145:115850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
3.  Zhang Y, Ma L, Lu E, Huang W. Atorvastatin Upregulates microRNA-186 and Inhibits the TLR4-Mediated MAPKs/NF-κB Pathway to Relieve Steroid-Induced Avascular Necrosis of the Femoral Head. Front Pharmacol. 2021;12:583975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
4.  Yang Y, Yan Y, Yin J, Tang N, Wang K, Huang L, Hu J, Feng Z, Gao Q, Huang A. O-GlcNAcylation of YTHDF2 promotes HBV-related hepatocellular carcinoma progression in an N(6)-methyladenosine-dependent manner. Signal Transduct Target Ther. 2023;8:63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 31]  [Reference Citation Analysis (0)]
5.  Paneque A, Fortus H, Zheng J, Werlen G, Jacinto E. The Hexosamine Biosynthesis Pathway: Regulation and Function. Genes (Basel). 2023;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 22]  [Reference Citation Analysis (0)]
6.  Kim SH, Kim YH, Song M, An SH, Byun HY, Heo K, Lim S, Oh YS, Ryu SH, Suh PG. O-GlcNAc modification modulates the expression of osteocalcin via OSE2 and Runx2. Biochem Biophys Res Commun. 2007;362:325-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
7.  Huang M, Xu S, Liu L, Zhang M, Guo J, Yuan Y, Xu J, Chen X, Zou J. m6A Methylation Regulates Osteoblastic Differentiation and Bone Remodeling. Front Cell Dev Biol. 2021;9:783322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 39]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
8.  Uzieliene I, Bernotiene E, Rakauskiene G, Denkovskij J, Bagdonas E, Mackiewicz Z, Porvaneckas N, Kvederas G, Mobasheri A. The Antihypertensive Drug Nifedipine Modulates the Metabolism of Chondrocytes and Human Bone Marrow-Derived Mesenchymal Stem Cells. Front Endocrinol (Lausanne). 2019;10:756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
9.  Zhang Z, Huang Z, Awad M, Elsalanty M, Cray J, Ball LE, Maynard JC, Burlingame AL, Zeng H, Mansky KC, Ruan HB. O-GlcNAc glycosylation orchestrates fate decision and niche function of bone marrow stromal progenitors. Elife. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
10.  Kim WJ, Shin HL, Kim BS, Kim HJ, Ryoo HM. RUNX2-modifying enzymes: therapeutic targets for bone diseases. Exp Mol Med. 2020;52:1178-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
11.  Chen Y, Zhao X, Wu H. Transcriptional Programming in Arteriosclerotic Disease: A Multifaceted Function of the Runx2 (Runt-Related Transcription Factor 2). Arterioscler Thromb Vasc Biol. 2021;41:20-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
12.  Qian K, Wang S, Fu M, Zhou J, Singh JP, Li MD, Yang Y, Zhang K, Wu J, Nie Y, Ruan HB, Yang X. Transcriptional regulation of O-GlcNAc homeostasis is disrupted in pancreatic cancer. J Biol Chem. 2018;293:13989-14000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 55]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
13.  Li X, Molina H, Huang H, Zhang YY, Liu M, Qian SW, Slawson C, Dias WB, Pandey A, Hart GW, Lane MD, Tang QQ. O-linked N-acetylglucosamine modification on CCAAT enhancer-binding protein beta: role during adipocyte differentiation. J Biol Chem. 2009;284:19248-19254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
14.  Zhang Z, Huang Z, Ong B, Sahu C, Zeng H, Ruan HB. Bone marrow adipose tissue-derived stem cell factor mediates metabolic regulation of hematopoiesis. Haematologica. 2019;104:1731-1743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
15.  Fistonich C, Zehentmeier S, Bednarski JJ, Miao R, Schjerven H, Sleckman BP, Pereira JP. Cell circuits between B cell progenitors and IL-7(+) mesenchymal progenitor cells control B cell development. J Exp Med. 2018;215:2586-2599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
16.  Asada N, Kunisaki Y, Pierce H, Wang Z, Fernandez NF, Birbrair A, Ma'ayan A, Frenette PS. Differential cytokine contributions of perivascular haematopoietic stem cell niches. Nat Cell Biol. 2017;19:214-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 317]  [Cited by in F6Publishing: 297]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
17.  Cordeiro Gomes A, Hara T, Lim VY, Herndler-Brandstetter D, Nevius E, Sugiyama T, Tani-Ichi S, Schlenner S, Richie E, Rodewald HR, Flavell RA, Nagasawa T, Ikuta K, Pereira JP. Hematopoietic Stem Cell Niches Produce Lineage-Instructive Signals to Control Multipotent Progenitor Differentiation. Immunity. 2016;45:1219-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 219]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
18.  Nagel AK, Ball LE. O-GlcNAc modification of the runt-related transcription factor 2 (Runx2) links osteogenesis and nutrient metabolism in bone marrow mesenchymal stem cells. Mol Cell Proteomics. 2014;13:3381-3395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
19.  Sun C, Lan W, Li B, Zuo R, Xing H, Liu M, Li J, Yao Y, Wu J, Tang Y, Liu H, Zhou Y. Glucose regulates tissue-specific chondro-osteogenic differentiation of human cartilage endplate stem cells via O-GlcNAcylation of Sox9 and Runx2. Stem Cell Res Ther. 2019;10:357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
20.  Komori T. Mechanism of transcriptional regulation by Runx2 in osteoblasts. Clin Calcium. 2006;16:801-807.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Shui C, Spelsberg TC, Riggs BL, Khosla S. Changes in Runx2/Cbfa1 expression and activity during osteoblastic differentiation of human bone marrow stromal cells. J Bone Miner Res. 2003;18:213-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 179]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
22.  Koyama T, Kamemura K. Global increase in O-linked N-acetylglucosamine modification promotes osteoblast differentiation. Exp Cell Res. 2015;338:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
23.  Weng Y, Wang Z, Fukuhara Y, Tanai A, Ikegame M, Yamada D, Takarada T, Izawa T, Hayano S, Yoshida K, Kamioka H, Okamura H. O-GlcNAcylation drives calcium signaling toward osteoblast differentiation: A bioinformatics-oriented study. Biofactors. 2021;47:992-1015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
24.  Botolin S, McCabe LR. Chronic hyperglycemia modulates osteoblast gene expression through osmotic and non-osmotic pathways. J Cell Biochem. 2006;99:411-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 179]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
25.  Schwartz AV. Diabetes Mellitus: Does it Affect Bone? Calcif Tissue Int. 2003;73:515-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 223]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
26.  Strotmeyer ES, Cauley JA, Orchard TJ, Steenkiste AR, Dorman JS. Middle-aged premenopausal women with type 1 diabetes have lower bone mineral density and calcaneal quantitative ultrasound than nondiabetic women. Diabetes Care. 2006;29:306-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 79]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
27.  Botolin S, McCabe LR. Bone loss and increased bone adiposity in spontaneous and pharmacologically induced diabetic mice. Endocrinology. 2007;148:198-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 199]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
28.  Bolanle IO, Palmer TM. Targeting Protein O-GlcNAcylation, a Link between Type 2 Diabetes Mellitus and Inflammatory Disease. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
29.  Gu H, Song M, Boonanantanasarn K, Baek K, Woo KM, Ryoo HM, Baek JH. Conditions Inducing Excessive O-GlcNAcylation Inhibit BMP2-Induced Osteogenic Differentiation of C2C12 Cells. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
30.  Vaidyanathan K, Wells L. Multiple tissue-specific roles for the O-GlcNAc post-translational modification in the induction of and complications arising from type II diabetes. J Biol Chem. 2014;289:34466-34471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 75]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]