Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Research Article

Efficacy and Mechanism of Highly Active Umbilical Cord Mesenchymal Stem Cells in the Treatment of Osteoporosis in Rats

Author(s): Chuan Tian, Guanke Lv, Li Ye, Xiaojuan Zhao, Mengdie Chen, Qianqian Ye, Qiang Li, Jing Zhao, Xiangqing Zhu and Xinghua Pan*

Volume 20, Issue 1, 2025

Published on: 14 February, 2024

Page: [91 - 102] Pages: 12

DOI: 10.2174/011574888X284911240131100909

Price: $65

Abstract

Background: Osteoporosis increases bone brittleness and the risk of fracture. Umbilical cord mesenchymal stem cell (UCMSC) treatment is effective, but how to improve the biological activity and clinical efficacy of UCMSCs has not been determined.

Methods: A rat model of osteoporosis was induced with dexamethasone sodium phosphate. Highly active umbilical cord mesenchymal stem cells (HA-UCMSCs) and UCMSCs were isolated, cultured, identified, and infused intravenously once at a dose of 2.29 × 106 cells/kg. In the 4th week of treatment, bone mineral density (BMD) was evaluated via cross-micro-CT, tibial structure was observed via HE staining, osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs) was examined via alizarin red staining, and carboxy-terminal cross-linked telopeptide (CTX), nuclear factor-κβ ligand (RANKL), procollagen type 1 N-terminal propeptide (PINP) and osteoprotegerin (OPG) levels were investigated via enzyme-linked immunosorbent assays (ELISAs). BMMSCs were treated with 10-6 mol/L dexamethasone and cocultured with HA-UCMSCs and UCMSCs in transwells. The osteogenic and adipogenic differentiation of BMMSCs was subsequently examined through directional induction culture. The protein expression levels of WNT, β-catenin, RUNX2, IFN-γ and IL-17 in the bone tissue were measured via Western blotting.

Results: The BMD in the healthy group was higher than that in the model group. Both UCMSCs and HA-UCMSCs exhibited a fusiform morphology; swirling growth; high expression of CD73, CD90 and CD105; and low expression of CD34 and CD45 and could differentiate into adipocytes, osteoblasts and chondrocytes, while HA-UCMSCs were smaller in size; had a higher nuclear percentage; and higher differentiation efficiency. Compared with those in the model group, the BMD increased, the bone structure improved, the trabecular area, number, and perimeter increased, the osteogenic differentiation of BMMSCs increased, RANKL expression decreased, and PINP expression increased after UCMSC and HA-UCMSC treatment for 4 weeks. Furthermore, the BMD, trabecular area, number and perimeter, calcareous nodule counts, and OPG/RANKL ratio were higher in the HA-UCMSC treatment group than in the UCMSC treatment group. The osteogenic and adipogenic differentiation of dexamethasone-treated BMMSCs was enhanced after the coculture of UCMSCs and HA-UCMSCs, and the HA-UCMSC group exhibited better effects than the UCMSC coculture group. The protein expression of WNT, β-catenin, and runx2 was upregulated, and IFN-γ and IL-17 expression was downregulated after UCMSC and HA-UCMSC treatment.

Conclusion: HA-UCMSCs have a stronger therapeutic effect on osteoporosis compared with that of UCMSCs. These effects include an improved bone structure, increased BMD, an increased number and perimeter of trabeculae, and enhanced osteogenic differentiation of BMMSCs via activation of the WNT/β-catenin pathway and inhibition of inflammation.

Keywords: Osteoporosis, umbilical cord mesenchymal stem cells, highly active umbilical cord mesenchymal stem cells, menopause, microenvironment, BMD.

Graphical Abstract
[1]
Liu P, Wang W, Li Z, et al. Ferroptosis: A new regulatory mechanism in osteoporosis. Oxid Med Cell Longev 2022; 2022: 1-10.
[http://dx.doi.org/10.1155/2022/2634431] [PMID: 35082963]
[2]
Gopinath V. Osteoporosis. Med Clin North Am 2023; 107: 213-25.
[http://dx.doi.org/10.1016/j.mcna.2022.10.013] [PMID: 36759092]
[3]
Williams C, Sapra A. Osteoporosis markers. StatPearls 2023.
[4]
Teng Z, Zhu Y, Lin D, et al. Deciphering the chromatin spatial organization landscapes during BMMSC differentiation. J Genet Genomics 2023; 50(4): 264-75.
[http://dx.doi.org/10.1016/j.jgg.2023.01.009] [PMID: 36720443]
[5]
Liu J, You Y, Sun Z, et al. WTAP-mediated m6A RNA methylation regulates the differentiation of bone marrow mesenchymal stem cells via the miR-29b-3p/HDAC4 axis. Stem Cells Transl Med 2023; 12(5): 307-21.
[http://dx.doi.org/10.1093/stcltm/szad020] [PMID: 37010483]
[6]
Li Y, Hu M, Xie J, Li S, Dai L. Dysregulation of histone modifications in bone marrow mesenchymal stem cells during skeletal ageing: Roles and therapeutic prospects. Stem Cell Res Ther 2023; 14(1): 166.
[http://dx.doi.org/10.1186/s13287-023-03393-6] [PMID: 37357311]
[7]
Zhang L, Zheng YL, Wang R, Wang XQ, Zhang H. Exercise for osteoporosis: A literature review of pathology and mechanism. Front Immunol 2022; 13: 1005665.
[http://dx.doi.org/10.3389/fimmu.2022.1005665] [PMID: 36164342]
[8]
Ma J, Chen P, Wang R. G-protein-coupled receptor 124 promotes osteogenic differentiation of BMSCs through the Wnt/β-catenin pathway. In Vitro Cell Dev Biol Anim 2022; 58(7): 529-38.
[http://dx.doi.org/10.1007/s11626-022-00684-9] [PMID: 35916978]
[9]
Mo C, Ke J, Zhao D, Zhang B. Role of the renin–angiotensin–aldosterone system in bone metabolism. J Bone Miner Metab 2020; 38(6): 772-9.
[http://dx.doi.org/10.1007/s00774-020-01132-y] [PMID: 32734523]
[10]
Biver E, Herrou J, Larid G, et al. Dietary recommendations in the prevention and treatment of osteoporosis. Joint Bone Spine 2023; 90(3): 105521.
[http://dx.doi.org/10.1016/j.jbspin.2022.105521] [PMID: 36566976]
[11]
Fleet JC. Vitamin D-mediated regulation of intestinal calcium absorption. Nutrients 2022; 14(16): 3351.
[http://dx.doi.org/10.3390/nu14163351] [PMID: 36014856]
[12]
Gosset A, Pouillès JM, Trémollieres F. Menopausal hormone therapy for the management of osteoporosis. Best Pract Res Clin Endocrinol Metab 2021; 35(6): 101551.
[http://dx.doi.org/10.1016/j.beem.2021.101551] [PMID: 34119418]
[13]
LeBoff MS, Greenspan SL, Insogna KL, et al. The clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int 2022; 33(10): 2049-102.
[http://dx.doi.org/10.1007/s00198-021-05900-y] [PMID: 35478046]
[14]
Suh B, Yu H, Kim H, et al. Interpretable deep-learning approaches for osteoporosis risk screening and individualized feature analysis using large population-based data: Model development and performance evaluation. J Med Internet Res 2023; 25: e40179.
[http://dx.doi.org/10.2196/40179] [PMID: 36482780]
[15]
Mei J, Hu H, Ding H, et al. Investigating the causal relationship between ankylosing spondylitis and osteoporosis in the European population: A bidirectional mendelian randomization study. Front Immunol 2023; 14: 1163258.
[http://dx.doi.org/10.3389/fimmu.2023.1163258] [PMID: 37359532]
[16]
Tian L, Luo C, Li YF, et al. Economic evaluation of four treatment strategies for postmenopausal patients with osteoporosis and a recent fracture in mainland China: A cost-effectiveness analysis. Arch Osteoporos 2023; 18(1): 100.
[http://dx.doi.org/10.1007/s11657-023-01309-8] [PMID: 37460858]
[17]
Yeh EJ, Gitlin M, Sorio F, McCloskey E. Estimating the future clinical and economic benefits of improving osteoporosis diagnosis and treatment among postmenopausal women across eight European countries. Arch Osteoporos 2023; 18(1): 68.
[http://dx.doi.org/10.1007/s11657-023-01230-0] [PMID: 37191892]
[18]
Hoang DM, Pham PT, Bach TQ, et al. Stem cell-based therapy for human diseases. Signal Transduct Target Ther 2022; 7(1): 272.
[http://dx.doi.org/10.1038/s41392-022-01134-4] [PMID: 35933430]
[19]
Dao TTT, Nguyen CTH, Vu NB, Le HTN, Nguyen PDN, Van Pham P. Evaluation of proliferation and osteogenic differentiation of human umbilical cord-derived mesenchymal stem cells in porous scaffolds. Adv Exp Med Biol 2019; 1084: 207-20.
[http://dx.doi.org/10.1007/5584_2019_343] [PMID: 31214911]
[20]
Lu Y, Zhang W, Tian Z, et al. The optimal transplantation strategy of umbilical cord mesenchymal stem cells in spinal cord injury: A systematic review and network meta-analysis based on animal studies. Stem Cell Res Ther 2022; 13(1): 441.
[http://dx.doi.org/10.1186/s13287-022-03103-8] [PMID: 36056386]
[21]
Zhao X, Fu L, Zou H, et al. Optogenetic engineered umbilical cord MSC-derived exosomes for remodeling of the immune microenvironment in diabetic wounds and the promotion of tissue repair. J Nanobiotechnology 2023; 21(1): 176.
[http://dx.doi.org/10.1186/s12951-023-01886-3] [PMID: 37269014]
[22]
Wang Y, Jing L, Lei X, et al. Umbilical cord mesenchymal stem cell-derived apoptotic extracellular vesicles ameliorate cutaneous wound healing in type 2 diabetic mice via macrophage pyroptosis inhibition. Stem Cell Res Ther 2023; 14(1): 257.
[http://dx.doi.org/10.1186/s13287-023-03490-6] [PMID: 37726853]
[23]
Zhang S, Ni W. High systemic immune-inflammation index is relevant to osteoporosis among middle-aged and older people: A cross-sectional study. Immun Inflamm Dis 2023; 11(8): e992.
[http://dx.doi.org/10.1002/iid3.992] [PMID: 37647432]
[24]
Jiang Y, Li S, Zhou Q, et al. PDCD4 negatively regulated osteogenic differentiation and bone defect repair of mesenchymal stem cells through GSK-3β/β-Catenin pathway. Stem Cells Dev 2021; 30(16): 806-15.
[http://dx.doi.org/10.1089/scd.2021.0041] [PMID: 34088227]
[25]
Liu A, Chen J, Zhang J, et al. Intra-articular injection of umbilical cord mesenchymal stem cells loaded with graphene oxide granular lubrication ameliorates inflammatory responses and osteoporosis of the subchondral bone in rabbits of modified papain-induced osteoarthritis. Front Endocrinol 2022; 12: 822294.
[http://dx.doi.org/10.3389/fendo.2021.822294] [PMID: 35095776]
[26]
Liu C, Zhang H, Tang X, et al. Mesenchymal stem cells promote the osteogenesis in collagen-induced arthritic mice through the inhibition of TNF- α. Stem Cells Int 2018; 2018: 1-10.
[http://dx.doi.org/10.1155/2018/4069032] [PMID: 29853911]
[27]
Xu Y, Jiang Y, Xia C, Wang Y, Zhao Z, Li T. Stem cell therapy for osteonecrosis of femoral head: Opportunities and challenges. Regen Ther 2020; 15: 295-304.
[http://dx.doi.org/10.1016/j.reth.2020.11.003] [PMID: 33426232]
[28]
Colicchia M, Jones DA, Beirne AM, et al. Umbilical cord–derived mesenchymal stromal cells in cardiovascular disease: Review of preclinical and clinical data. Cytotherapy 2019; 21(10): 1007-18.
[http://dx.doi.org/10.1016/j.jcyt.2019.04.056] [PMID: 31540804]
[29]
Zhang S, Wang JY, Li B, Yin F, Liu H. Single-cell transcriptome analysis of uncultured human umbilical cord mesenchymal stem cells. Stem Cell Res Ther 2021; 12(1): 25.
[http://dx.doi.org/10.1186/s13287-020-02055-1] [PMID: 33413643]
[30]
Zhang L, Sun Y, Zhang XX, et al. Comparison of CD146 +/− mesenchymal stem cells in improving premature ovarian failure. Stem Cell Res Ther 2022; 13(1): 267.
[http://dx.doi.org/10.1186/s13287-022-02916-x] [PMID: 35729643]
[31]
Zhang Y, Zhou L, Fu Q, Liu Z. ANKRD1 activates the Wnt signaling pathway by modulating CAV3 expression and thus promotes BMSC osteogenic differentiation and bone formation in ovariectomized mice. Biochim Biophys Acta Mol Basis Dis 2023; 1869(5): 166693.
[http://dx.doi.org/10.1016/j.bbadis.2023.166693] [PMID: 36958710]
[32]
Wang B, Xu N, Cao L, et al. miR-31 from mesenchymal stem cell-derived extracellular vesicles alleviates intervertebral disc degeneration by inhibiting NFAT5 and upregulating the Wnt/β-catenin pathway. Stem Cells Int 2022; 2022: 1-16.
[http://dx.doi.org/10.1155/2022/2164057] [PMID: 36311041]
[33]
Zuo R, Liu M, Wang Y, et al. BM-MSC-derived exosomes alleviate radiation-induced bone loss by restoring the function of recipient BM-MSCs and activating Wnt/β-catenin signaling. Stem Cell Res Ther 2019; 10(1): 30.
[http://dx.doi.org/10.1186/s13287-018-1121-9] [PMID: 30646958]
[34]
Huang X, Chen W, Gu C, et al. Melatonin suppresses bone marrow adiposity in ovariectomized rats by rescuing the imbalance between osteogenesis and adipogenesis through SIRT1 activation. J Orthop Translat 2023; 38: 84-97.
[http://dx.doi.org/10.1016/j.jot.2022.10.002] [PMID: 36381247]
[35]
Liu C, Liu AS, Zhong D, et al. Circular RNA AFF4 modulates osteogenic differentiation in BM-MSCs by activating SMAD1/5 pathway through miR-135a-5p/FNDC5/Irisin axis. Cell Death Dis 2021; 12(7): 631.
[http://dx.doi.org/10.1038/s41419-021-03877-4] [PMID: 34145212]
[36]
Guo DB, Zhu XQ, Li QQ, et al. Efficacy and mechanisms underlying the effects of allogeneic umbilical cord mesenchymal stem cell transplantation on acute radiation injury in tree shrews. Cytotechnology 2018; 70(5): 1447-68.
[http://dx.doi.org/10.1007/s10616-018-0239-z] [PMID: 30066056]
[37]
He J, Yao X, Mo P, et al. Lack of tumorigenesis and protumorigenic activity of human umbilical cord mesenchymal stem cells in NOD SCID mice. BMC Cancer 2022; 22(1): 307.
[http://dx.doi.org/10.1186/s12885-022-09431-5] [PMID: 35317758]
[38]
Ruan G, Yao X, Mo P, et al. Establishment of a systemic inflammatory response syndrome model and evaluation of the efficacy of umbilical cord mesenchymal stem cell transplantation. Cells Tissues Organs 2021; 210(2): 118-34.
[http://dx.doi.org/10.1159/000514619] [PMID: 34182545]
[39]
Li F, Zhou C, Xu L, Tao S, Zhao J, Gu Q. Effect of stem cell therapy on bone mineral density: A meta-analysis of preclinical studies in animal models of osteoporosis. PLoS One 2016; 11(2): e0149400.
[http://dx.doi.org/10.1371/journal.pone.0149400] [PMID: 26882451]
[40]
Kabat M, Bobkov I, Kumar S, Grumet M. Trends in mesenchymal stem cell clinical trials 2004-2018: Is efficacy optimal in a narrow dose range? Stem Cells Transl Med 2020; 9(1): 17-27.
[http://dx.doi.org/10.1002/sctm.19-0202] [PMID: 31804767]
[41]
Yang Y, Li Y, Wang Y, et al. The effects of BMMSC treatment on lung tissue degeneration in elderly macaques. Stem Cell Res Ther 2021; 12(1): 156.
[http://dx.doi.org/10.1186/s13287-021-02201-3] [PMID: 33648583]
[42]
Pan XH, Yang XY, Yao X, et al. Bone-marrow mesenchymal stem cell transplantation to treat diabetic nephropathy in tree shrews. Cell Biochem Funct 2014; 32(5): 453-63.
[http://dx.doi.org/10.1002/cbf.3037] [PMID: 24867093]
[43]
Dai X, Heng BC, Bai Y, et al. Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization. Bioact Mater 2021; 6(7): 2029-38.
[http://dx.doi.org/10.1016/j.bioactmat.2020.12.020] [PMID: 33474514]
[44]
Ekeuku SO, Mohd Ramli ES, Abdullah Sani N, Abd Ghafar N, Soelaiman IN, Chin KY. Tocotrienol as a protecting agent against glucocorticoid-induced osteoporosis: A mini review of potential mechanisms. Molecules 2022; 27(18): 5862.
[http://dx.doi.org/10.3390/molecules27185862] [PMID: 36144598]
[45]
Jiang Y, Zhang P, Zhang X, Lv L, Zhou Y. Advances in mesenchymal stem cell transplantation for the treatment of osteoporosis. Cell Prolif 2021; 54(1): e12956.
[http://dx.doi.org/10.1111/cpr.12956] [PMID: 33210341]
[46]
Yahao G, Xinjia W. The role and mechanism of exosomes from umbilical cord mesenchymal stem cells in inducing osteogenesis and preventing osteoporosis. Cell Transplant 2021; 30: 9636897211057465.
[http://dx.doi.org/10.1177/09636897211057465] [PMID: 34814742]
[47]
Li M, Yang N, Hao L, et al. Melatonin inhibits the ferroptosis pathway in rat bone marrow mesenchymal stem cells by activating the PI3K/AKT/mTOR signaling axis to attenuate steroid-induced osteoporosis. Oxid Med Cell Longev 2022; 2022: 1-22.
[http://dx.doi.org/10.1155/2022/8223737] [PMID: 36035224]
[48]
Liu Y, Chen Y, Li XH, et al. Dissection of cellular communication between human primary osteoblasts and bone marrow mesenchymal stem cells in osteoarthritis at single-cell resolution. Int J Stem Cells 2023; 16(3): 342-55.
[http://dx.doi.org/10.15283/ijsc22101] [PMID: 37105556]
[49]
Gholami Farashah MS, Javadi M, Mohammadi A, Soleimani Rad J, Shakouri SK, Roshangar L. Bone marrow mesenchymal stem cell’s exosomes as key nanoparticles in osteogenesis and bone regeneration: Specific capacity based on cell type. Mol Biol Rep 2022; 49(12): 12203-18.
[http://dx.doi.org/10.1007/s11033-022-07807-1] [PMID: 36224447]
[50]
Wang Z, Li X, Yang J, et al. Single-cell RNA sequencing deconvolutes the in vivo heterogeneity of human bone marrow-derived mesenchymal stem cells. Int J Biol Sci 2021; 17(15): 4192-206.
[http://dx.doi.org/10.7150/ijbs.61950] [PMID: 34803492]
[51]
McDaniels-Davidson CR, Kritz-Silverstein D, Huang MH, et al. The association between bone turnover markers and kyphosis in community-dwelling older adults. Bone Rep 2016; 5: 57-61.
[http://dx.doi.org/10.1016/j.bonr.2016.04.001] [PMID: 27868084]
[52]
Sølling AS, Harsløf T, Jørgensen NR, Langdahl B. Changes in RANKL and TRAcP 5b after discontinuation of denosumab suggest RANKL mediated formation of osteoclasts results in the increased bone resorption. Osteoporos Int 2023; 34(3): 599-605.
[http://dx.doi.org/10.1007/s00198-022-06651-0] [PMID: 36543965]
[53]
Eastell R, Szulc P. Use of bone turnover markers in postmenopausal osteoporosis. Lancet Diabetes Endocrinol 2017; 5(11): 908-23.
[http://dx.doi.org/10.1016/S2213-8587(17)30184-5] [PMID: 28689768]
[54]
Diemar SS, Dahl SS, West AS, Simonsen SA, Iversen HK, Jørgensen NR. A systematic review of the circadian rhythm of bone markers in blood. Calcif Tissue Int 2022; 112(2): 126-47.
[http://dx.doi.org/10.1007/s00223-022-00965-1] [PMID: 35305134]
[55]
Jura-Półtorak A, Szeremeta A, Olczyk K, Zoń-Giebel A, Komosińska-Vassev K. Bone metabolism and RANKL/OPG ratio in rheumatoid arthritis women treated with TNF-α inhibitors. J Clin Med 2021; 10(13): 2905.
[http://dx.doi.org/10.3390/jcm10132905] [PMID: 34209821]
[56]
Nair S, Hatkar S, Patil A, et al. Age-related changes and reference intervals of RANKL, OPG, and bone turnover markers in Indian women. Arch Osteoporos 2021; 16(1): 146.
[http://dx.doi.org/10.1007/s11657-021-01014-4] [PMID: 34606009]
[57]
Gifre L, Ruiz-Gaspà S, Carrasco JL, et al. Effect of recent spinal cord injury on the OPG/RANKL system and its relationship with bone loss and the response to denosumab therapy. Osteoporos Int 2017; 28(9): 2707-15.
[http://dx.doi.org/10.1007/s00198-017-4090-4] [PMID: 28580511]
[58]
Brown JP, Don-Wauchope A, Douville P, Albert C, Vasikaran SD. Current use of bone turnover markers in the management of osteoporosis. Clin Biochem 2022; 109-110: 1-10.
[http://dx.doi.org/10.1016/j.clinbiochem.2022.09.002] [PMID: 36096182]
[59]
Ma D, Wu Z, Zhao X, et al. Immunomodulatory effects of umbilical mesenchymal stem cell-derived exosomes on CD4+ T cells in patients with primary Sjögren’s syndrome. Inflammopharmacology 2023; 31(4): 1823-38.
[http://dx.doi.org/10.1007/s10787-023-01189-x] [PMID: 37012581]
[60]
Xu C, Feng C, Huang P, et al. TNFα and IFNγ rapidly activate PI3K-AKT signaling to drive glycolysis that confers mesenchymal stem cells enhanced anti-inflammatory property. Stem Cell Res Ther 2022; 13(1): 491.
[http://dx.doi.org/10.1186/s13287-022-03178-3] [PMID: 36195887]
[61]
Wang Z, Hu Y, Wang X, et al. Comparative analysis of the therapeutic effects of fresh and cryopreserved human umbilical cord derived mesenchymal stem cells in the treatment of psoriasis. Stem Cell Rev Rep 2023; 19(6): 1922-36.
[http://dx.doi.org/10.1007/s12015-023-10556-8] [PMID: 37199874]
[62]
Chen H, Luo Y, Zhu Y, et al. Enhanced secretion of hepatocyte growth factor in human umbilical cord mesenchymal stem cells ameliorates pulmonary fibrosis induced by bleomycin in rats. Front Pharmacol 2023; 13: 1070736.
[http://dx.doi.org/10.3389/fphar.2022.1070736] [PMID: 36726784]
[63]
Xiang E, Han B, Zhang Q, et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Res Ther 2020; 11(1): 336.
[http://dx.doi.org/10.1186/s13287-020-01852-y] [PMID: 32746936]
[64]
Eldaly AS, Mashaly SM, Fouda E, et al. Systemic anti-inflammatory effects of mesenchymal stem cells in burn: A systematic review of animal studies. J Clin Transl Res 2022; 8(4): 276-91.
[PMID: 35991083]
[65]
Shen D, Wang Z, Wang H, et al. Evaluation of preclinical efficacy of human umbilical cord mesenchymal stem cells in ankylosing spondylitis. Front Immunol 2023; 14: 1153927.
[http://dx.doi.org/10.3389/fimmu.2023.1153927] [PMID: 37063838]
[66]
Wu L, Wei Q, Lv Y, et al. Wnt/β-catenin pathway is involved in cadmium-induced inhibition of osteoblast differentiation of bone marrow mesenchymal stem cells. Int J Mol Sci 2019; 20(6): 1519.
[http://dx.doi.org/10.3390/ijms20061519] [PMID: 30917596]
[67]
Wang E, Zhang Y, Ding R, Wang X, Zhang S, Li X. miR-30a-5p induces the adipogenic differentiation of bone marrow mesenchymal stem cells by targeting FAM13A/Wnt/β-catenin signaling in aplastic anemia. Mol Med Rep 2022; 25(1): 25.
[PMID: 34821370]
[68]
Jing H, Su X, Gao B, et al. Correction: Epigenetic inhibition of Wnt pathway suppresses osteogenic differentiation of BMSCs during osteoporosis. Cell Death Dis 2022; 13(2): 194.
[http://dx.doi.org/10.1038/s41419-022-04616-z] [PMID: 35228514]
[69]
Xie L, Zou L, Chen J, Liu Y. All-trans retinoic acid inhibits bone marrow mesenchymal stem cell commitment to adipocytes via upregulating FRA1 signaling. Int J Endocrinol 2020; 2020: 1-11.
[http://dx.doi.org/10.1155/2020/6525787] [PMID: 32089684]
[70]
Zhao Y, Kang Z, Mao Y, Luo F, Huo Y. GLIS family zinc finger protein 2 (GLIS2) negatively regulates the Wnt/β-catenin pathway to inhibit the osteogenic differentiation of human bone marrow mesenchymal stem cells. Xibao Yu Fenzi Mianyixue Zazhi 2023; 39(2): 159-64.
[PMID: 36872435]
[71]
Sahebdel F, Parvaneh Tafreshi A, Arefian E, Roussa E, Nadri S, Zeynali B. Wnt/β-catenin signaling pathway is involved in early dopaminergic differentiation of trabecular meshwork-derived mesenchymal stem cells. J Cell Biochem 2022; 123(6): 1120-9.
[http://dx.doi.org/10.1002/jcb.30269] [PMID: 35533251]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy