1
|
Jung SI, Choi SH, Kim JW, Lim J, Rim YA, Ju JH. The Effect of Nerve Growth Factor on Cartilage Fibrosis and Hypertrophy during In Vitro Chondrogenesis Using Induced Pluripotent Stem Cells. Int J Stem Cells 2025; 18:59-71. [PMID: 39734065 PMCID: PMC11867901 DOI: 10.15283/ijsc24097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 12/31/2024] Open
Abstract
Nerve growth factor (NGF) is a neurotrophic factor usually involved in the survival, differentiation, and growth of sensory neurons and nociceptive function. Yet, it has been suggested to play a role in the pathogenesis of osteoarthritis (OA). Previous studies suggested a possible relationship between NGF and OA; however, the underlying mechanisms remain unknown. Therefore, we investigated the impact of NGF in chondrogenesis using human induced pluripotent stem cells (hiPSCs)-derived chondrogenic pellets. To investigate how NGF affects the cartilage tissue, hiPSC-derived chondrogenic pellets were treated with NGF on day 3 of differentiation, expression of chondrogenic, hypertrophic, and fibrotic markers was confirmed. Also, inflammatory cytokine arrays were performed using the culture medium of the NGF treated chondrogenic pellets. As a result, NGF treatment decreased the expression of pro-chondrogenic markers by approximately 2~4 times, and hypertrophic (pro-osteogenic) markers and fibrotic markers were increased by approximately 3-fold or more in the NGF-treated cartilaginous pellets. In addition, angiogenesis was upregulated by approximately 4-fold or more, bone formation by more than 2-fold, and matrix metalloproteinase induction by more than 2-fold. These inflammatory cytokine array were using the NGF-treated chondrogenic pellet cultured medium. Furthermore, it was confirmed by Western blot to be related to the induction of the glycogen synthase kinase-3 beta (GSK3β) pathway by NGF. In Conclusions, these findings provide valuable insights into the multifaceted role of NGF in cartilage hypertrophy and fibrosis, which might play a critical role in OA progression.
Collapse
Affiliation(s)
- Se In Jung
- Catholic iPSCs Research Center, CiSTEM Laboratory, Department of Medical Sciences, Graduate School The Catholic University of Korea, Seoul, Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Korea
| | - Si Hwa Choi
- Catholic iPSCs Research Center, CiSTEM Laboratory, Department of Medical Sciences, Graduate School The Catholic University of Korea, Seoul, Korea
| | - Jang-Woon Kim
- Catholic iPSCs Research Center, CiSTEM Laboratory, Department of Medical Sciences, Graduate School The Catholic University of Korea, Seoul, Korea
| | - Jooyoung Lim
- Catholic iPSCs Research Center, CiSTEM Laboratory, Department of Medical Sciences, Graduate School The Catholic University of Korea, Seoul, Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Korea
| | - Yeri Alice Rim
- Catholic iPSCs Research Center, CiSTEM Laboratory, Department of Medical Sciences, Graduate School The Catholic University of Korea, Seoul, Korea
| | - Ji Hyeon Ju
- Catholic iPSCs Research Center, CiSTEM Laboratory, Department of Medical Sciences, Graduate School The Catholic University of Korea, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
2
|
Cohen DJ, Berger MB, Deng J, Jacobs TW, Boyan BD, Schwartz Z. Non-Canonical Wnt16 and microRNA-145 Mediate the Response of Human Bone Marrow Stromal Cells to Additively Manufactured Porous 3-Dimensional Biomimetic Titanium-Aluminum-Vanadium Constructs. Cells 2025; 14:211. [PMID: 39937002 PMCID: PMC11816670 DOI: 10.3390/cells14030211] [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: 12/03/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Metal 3D printing is increasingly being used to manufacture titanium-aluminum-vanadium (Ti6Al4V) implants. In vitro studies using 2D substrates demonstrate that the osteoblastic differentiation of bone marrow stromal cells (MSCs) on Ti6Al4V surfaces, with a microscale/nanoscale surface topography that mimics an osteoclast resorption pit, involves non-canonical Wnt signaling; Wnt3a is downregulated and Wnt5a is upregulated, leading to the local production of BMP2 and semaphorin 3A (sema3A). In this study, it was examined whether the regulation of MSCs in a 3D environment occurs by a similar mechanism. Human MSCs from two different donors were cultured for 7, 14, or 21 days on porous (3D) or solid (2D) constructs fabricated by powder-bed laser fusion. mRNA and secretion of osteoblast markers, as well as factors that enhance peri-implant osteogenesis, were analyzed, with a primary focus on the Wnt family, sema3A, and microRNA-145 (miR-145) signaling pathways. MSCs exhibited greater production of osteocalcin, latent and active TGFβ1, sema3A, and Wnt16 on the 3D constructs compared to 2D, both of which had similar microscale/nanoscale surface modifications. Wnt3a was reduced on 2D constructs as a function of time; Wnt11 and Wnt5a remained elevated in the 3D and 2D cultures. To better understand the role of Wnt16, cultures were treated with rhWnt16; endogenous Wnt16 was blocked using an antibody. Wnt16 promoted proliferation and inhibited osteoblast differentiation, potentially by reducing production of BMP2 and BMP4. Wnt16 expression was reduced by exogenous Wnt16 in 3D cells. Addition of the anti-Wnt16 antibody to the cultures reversed the effects of exogenous Wnt16, indicating an autocrine mechanism. Wnt16 increased miR-145-5p, suggesting a potential feedback mechanism. The miR-145-5p mimic increased Wnt16 production and inhibited sema3A in a 3D porous substrate-specific manner. Wnt16 did not affect sema3A production, but it was reduced by miR-145-5p mimic on the 3D constructs and stimulated by miR-145-5p inhibitor. Media from 7-, 14-, and 21-day cultures of MSCs grown on 3D constructs inhibited osteoclast activity to a greater extent than media from the 2D cultures. The findings present a significant step towards understanding the complex molecular interplay that occurs in 3D Ti6Al4V constructs fabricated by additive manufacturing. In addition to enhancing osteogenesis, the 3D porous biomimetic structure inhibits osteoclast activities, indicating its role in modulating bone remodeling processes. Our data suggest that the pathway mediated by sema3A/Wnt16/miR145-5p was enhanced by the 3D surface and contributes to bone regeneration in the 3D implants. This comprehensive exploration contributes valuable insights to guide future strategies in implant design, customization, and ultimately aims at improving clinical outcomes and successful osseointegration.
Collapse
Affiliation(s)
- David. J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (M.B.B.); (J.D.); (T.W.J.); (B.D.B.)
| | - Michael B. Berger
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (M.B.B.); (J.D.); (T.W.J.); (B.D.B.)
| | - Jingyao Deng
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (M.B.B.); (J.D.); (T.W.J.); (B.D.B.)
| | - Thomas W. Jacobs
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (M.B.B.); (J.D.); (T.W.J.); (B.D.B.)
| | - Barbara D. Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (M.B.B.); (J.D.); (T.W.J.); (B.D.B.)
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (M.B.B.); (J.D.); (T.W.J.); (B.D.B.)
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
3
|
Tahoori M, Tafreshi AP, Naghshnejad F, Zeynali B. Transforming Growth Factor-β Signaling Inhibits the Osteogenic Differentiation of Mesenchymal Stem Cells via Activation of Wnt/β-Catenin Pathway. J Bone Metab 2025; 32:11-20. [PMID: 40098425 DOI: 10.11005/jbm.24.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Due to the contradictory and temporally variable effects of transforming growth factor-β (TGF-β) and the Wnt/β-catenin pathways on osteogenic differentiation in different stem cell types, we sought to examine the activity of these pathways as well as their interaction during the osteogenic differentiation of the osteo-induced adiposederived mesenchymal stem cells (AD-MSCs). METHODS The osteo-induced AD-MSCs were treated with TGF-β1 (1 ng/mL) either alone or together with its antagonist SB- 431542 (10 μM) or that of the Wnt antagonist, inhibitor of Wnt production 2 (IWP2) (3 μM), every 3 days for 21 days. Cells were then analyzed for calcium deposit, bone matrix production, and the osteogenic markers gene expression. RESULTS Our results showed firstly that, either of the pathways is active since the mRNA expressions of their respective target genes, PAI-1 and Cyclin D1 were detectable although the latter was at a very low level. Secondly that, treatment with TGF-β1 decreased levels of calcium deposit, bone matrix production and the osteogenic markers gene expression. Accordingly, osteogenesis was induced in those treated with SB either alone or together with the TGF-β1, pointing to inhibitory effect of TGF-β pathway on osteogenic differentiation. Thirdly that following treatment with IWP2 and TGF-β1, the inhibitory effect of TGF-β1 on bone matrix production was reversed. Fourthly, there was constantly low expression of Wnt3amRNA but progressively increasing that of its endogenous antagonist Dkk-1mRNA throughout. CONCLUSIONS Together these results suggest that TGF-β1 requires the active Wnt/β-catenin signaling pathway to exert its inhibitory effects on osteogenic differentiation of AD-MSCs.
Collapse
Affiliation(s)
- Mahsa Tahoori
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Azita Parvaneh Tafreshi
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Department of Molecular Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fatemeh Naghshnejad
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Bahman Zeynali
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
4
|
Chen M, Huang B, Su X. Mesenchymal stem cell-derived extracellular vesicles in periodontal bone repair. J Mol Med (Berl) 2025; 103:137-156. [PMID: 39821702 DOI: 10.1007/s00109-025-02513-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
Periodontitis is a chronic inflammatory disease that destroys tooth-supporting structures and poses significant public health challenges due to its high prevalence and links to systemic health conditions. Traditional treatments are effective in reducing the inflammatory response and improving the clinical symptoms of periodontitis. However, these methods are challenging to achieve an ideal treatment effect in alveolar bone repair. Mesenchymal stem cells (MSCs) represent a potential alternative for the treatment of periodontal bone defects due to their self-renewal and differentiation capabilities. Recent research indicates that MSCs exert their effects primarily through paracrine mechanisms. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) serve as pivotal mediators in intercellular communication, transferring microRNAs (miRNAs), messenger RNAs (mRNAs), proteins, and cytokines to recipient cells, thereby emulating the therapeutic effects of MSCs. In periodontitis, MSC-EVs play a pivotal role in immunomodulation and bone remodeling, thereby facilitating periodontal bone repair. As a cell-free therapy, MSC-EVs demonstrate considerable clinical potential due to their specialized membrane structure, minimal immunogenicity, low toxicity, high biocompatibility, and nanoscale size. This review indicates that MSC-EVs represent a promising approach for periodontitis treatment, with the potential to overcome the limitations of traditional therapies and offer a more effective solution for bone repair in periodontal disease. In this review, we introduce MSC-EVs, emphasizing their mechanisms and clinical applications in periodontal bone repair. It synthesizes recent advances, existing challenges, and future prospects to present up-to-date information and novel techniques for periodontal regeneration and to guide the improvement of MSC-EV therapy in clinical practice.
Collapse
Affiliation(s)
- Mengbing Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bo Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
5
|
Cohen DJ, Van Duyn CM, Deng J, Lodi MK, Gallagher MB, Sugar JT, Rawlinson JJ, Ghosh P, Boyan BD, Schwartz Z. Bone Marrow Stromal Cells Generate a Pro-Healing Inflammasome When Cultured on Titanium-Aluminum-Vanadium Surfaces with Microscale/Nanoscale Structural Features. Biomimetics (Basel) 2025; 10:66. [PMID: 39851782 PMCID: PMC11759188 DOI: 10.3390/biomimetics10010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The surface topography and chemistry of titanium-aluminum-vanadium (Ti6Al4V) implants play critical roles in the osteoblast differentiation of human bone marrow stromal cells (MSCs) and the creation of an osteogenic microenvironment. To assess the effects of a microscale/nanoscale (MN) topography, this study compared the effects of MN-modified, anodized, and smooth Ti6Al4V surfaces on MSC response, and for the first time, directly contrasted MN-induced osteoblast differentiation with culture on tissue culture polystyrene (TCPS) in osteogenic medium (OM). Surface characterization revealed distinct differences in microroughness, composition, and topography among the Ti6Al4V substrates. MSCs on MN surfaces exhibited enhanced osteoblastic differentiation, evidenced by increased expression of RUNX2, SP7, BGLAP, BMP2, and BMPR1A (fold increases: 3.2, 1.8, 1.4, 1.3, and 1.2). The MN surface also induced a pro-healing inflammasome with upregulation of anti-inflammatory mediators (170-200% increase) and downregulation of pro-inflammatory factors (40-82% reduction). Integrin expression shifted towards osteoblast-associated integrins on MN surfaces. RNA-seq analysis revealed distinct gene expression profiles between MSCs on MN surfaces and those in OM, with only 199 shared genes out of over 1000 differentially expressed genes. Pathway analysis showed that MN surfaces promoted bone formation, maturation, and remodeling through non-canonical Wnt signaling, while OM stimulated endochondral bone development and mineralization via canonical Wnt3a signaling. These findings highlight the importance of Ti6Al4V surface properties in directing MSC differentiation and indicate that MN-modified surfaces act via signaling pathways that differ from OM culture methods, more accurately mimicking peri-implant osteogenesis in vivo.
Collapse
Affiliation(s)
- David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Christine M. Van Duyn
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Jingyao Deng
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Musaddiq K. Lodi
- Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | | | - James T. Sugar
- Medtronic Spine, Memphis, TN 38132, USA; (M.B.G.); (J.T.S.); (J.J.R.)
| | | | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
- Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
6
|
Zheng Z, Zhou H, Zhang W, Wang T, Swamiappan S, Peng X, Zhou Y. Effects of advanced glycation end products on stem cell. Front Cell Dev Biol 2024; 12:1532614. [PMID: 39777263 PMCID: PMC11703976 DOI: 10.3389/fcell.2024.1532614] [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: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
In recent years, stem cell therapy has become a pivotal component of regenerative medicine. Stem cells, characterized by their self-renewal capacity and multidirectional differentiation potential, can be isolated from a variety of biological tissues, including adipose tissue, bone marrow, the umbilical cord, and the placenta. The classic applications of stem cells include human pluripotent stem cells (hPSCs) and mesenchymal stem cells (MSCs). However, numerous factors can influence the normal physiological function of stem cells. For instance, in diabetes mellitus, advanced glycation end products (AGEs) accumulate in the extracellular matrix (ECM), impairing the physiological function of stem cells. These substances are closely associated with aging and the progression of numerous degenerative diseases. AGEs can create an environment that is detrimental to the normal physiological functions of stem cells. By binding to the primary cellular receptor for advanced glycation end products (RAGE), AGEs disrupt the physiological activities of stem cells. The binding of RAGE to various ligands triggers the activation of downstream signaling pathways, contributing to the pathophysiological development of diabetes, aging, neurodegenerative diseases, and cancer. Therefore, there is an urgent need for comprehensive research on the impact of AGEs on stem cells, which could provide new insights into the therapeutic application of stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Zetai Zheng
- Department of Pathophysiology, Guangdong Medical University, Dongguan, China
| | - Hui Zhou
- School of Pharmaceutical Sciences, Guangdong Medical University, Dongguan, China
| | - Wenwen Zhang
- Department of Pathophysiology, Guangdong Medical University, Dongguan, China
| | - Tingyu Wang
- Department of Pathophysiology, Guangdong Medical University, Dongguan, China
| | | | - Xinsheng Peng
- School of Pharmaceutical Sciences, Guangdong Medical University, Dongguan, China
| | - Yanfang Zhou
- Department of Pathophysiology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
7
|
Liu S, Ren J, Hu Y, Zhou F, Zhang L. TGFβ family signaling in human stem cell self-renewal and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:26. [PMID: 39604763 PMCID: PMC11602941 DOI: 10.1186/s13619-024-00207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Human stem cells are undifferentiated cells with the capacity for self-renewal and differentiation into distinct cell lineages, playing important role in the development and maintenance of diverse tissues and organs. The microenvironment of stem cell provides crucial factors and components that exert significant influence over the determination of cell fate. Among these factors, cytokines from the transforming growth factor β (TGFβ) superfamily, including TGFβ, bone morphogenic protein (BMP), Activin and Nodal, have been identified as important regulators governing stem cell maintenance and differentiation. In this review, we present a comprehensive overview of the pivotal roles played by TGFβ superfamily signaling in governing human embryonic stem cells, somatic stem cells, induced pluripotent stem cells, and cancer stem cells. Furthermore, we summarize the latest research and advancements of TGFβ family in various cancer stem cells and stem cell-based therapy, discussing their potential clinical applications in cancer therapy and regeneration medicine.
Collapse
Affiliation(s)
- Sijia Liu
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Ren
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanmei Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Long Zhang
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Chen Y, Petho A, Ganapathy A, George A. DPP an extracellular matrix molecule induces Wnt5a mediated signaling to promote the differentiation of adult stem cells into odontogenic lineage. Sci Rep 2024; 14:26187. [PMID: 39478025 PMCID: PMC11525562 DOI: 10.1038/s41598-024-76069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Dentin phosphophoryn (DPP) an extracellular matrix protein activates Wnt signaling in DPSCs (dental pulp stem cells). Wnt/β catenin signaling is essential for tooth development but the role of DPP-mediated Wnt5a signaling in odontogenesis is not well understood. Wnt5a is typically considered as a non-canonical Wnt ligand that elicits intracellular signals through association with a specific cohort of receptors and co-receptors in a cell and context-dependent manner. In this study, DPP facilitated the interaction of Wnt5a with Frizzled 5 and LRP6 to induce nuclear translocation of β-catenin. β-catenin has several nuclear binding partners that promote the activation of Wnt target genes responsible for odontogenic differentiation. Interestingly, steady increase in the expression of Vangl2 receptor suggest planar cell polarity signaling during odontogenic differentiation. In vitro observations were further strengthened by the low expression levels of Wnt5a and β-catenin in the teeth of DSPP KO mice which exhibit impaired odontoblast differentiation and defective dentin mineralization. Together, this study suggests that the DPP-mediated Wnt5a signaling could be exploited as a therapeutic approach for the differentiation of dental pulp stem cells into functional odontoblasts and dentin regeneration.
Collapse
Affiliation(s)
- Yinghua Chen
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Adrienn Petho
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Amudha Ganapathy
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Anne George
- Department of Oral Biology, University of Illinois Chicago, Chicago, IL, 60612, USA.
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina St, Chicago, IL, 60612, USA.
| |
Collapse
|
9
|
Sharma G, Abdullah KM, Qais FA, Khan P, Cox JL, Sarwar T, Nasser MW, Batra SK, Siddiqui JA. Clofazimine inhibits small-cell lung cancer progression by modulating the kynurenine/aryl hydrocarbon receptor axis. Int J Biol Macromol 2024; 282:136921. [PMID: 39490481 DOI: 10.1016/j.ijbiomac.2024.136921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Small cell lung cancer (SCLC) is one of the highly metastatic malignancies that contributes to ~15 % of all lung cancers. Most SCLC patients (50-60 %) develop osteolytic bone metastases, significantly affecting their quality of life. Among several factors, environmental pollutant 2,3,7,8-Tetrachlorodibenzodioxin (TCDD) and kynurenine (Kyn), an endogenous ligand derived from tryptophan (Trp) metabolism, activate the aryl hydrocarbon receptor (AhR) and are responsible for SCLC progression and metastasis. Further, elevated AhR expression in bone cells intensifies bone resorption, making the Kyn/AhR axis a potential target for the bone metastatic propensity of SCLC. We first assessed the expression profile of AhR in human SCLC cell lines and found a significantly increased expression compared to normal lung cells. Additionally, we also evaluated the clinical significance of AhR expression in the patient samples of SCLC along with the relevance of the same in the Rb1fl/fl; Trp53fl/fl; MycLSL/LSL (RPM) mouse model using immunohistochemistry and found the higher AhR expression in the patient samples and RPM mouse tumor tissues. Using computational simulations, we found that clofazimine (CLF) binds at the activator (Kyn) binding site by forming a stable complex with AhR. The CLF binding with AhR was favored by Van der Waals and hydrophobic forces, and the proteins retained their secondary structure. Furthermore, we found that Kyn treatment potentiates the migration and clonogenic ability of SCLC cell lines by activating Erk/Akt oncogenic signaling. Blocking AhR with CLF reduces AhR expression, inhibits Kyn-mediated proliferation of SCLC cells, and induces apoptosis and cell cycle arrest in the G2/M phase; further, our examination indicates that Kyn treatment also promotes osteoblast-mediated osteoclast differentiation through RANKL. The treatment with CLF impedes RANKL expression and osteoclastogenesis, suggesting that CLF has the potential for developing SCLC therapies that have efficacies against bone metastasis.
Collapse
Affiliation(s)
- Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson MS-68198, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson MS-68198, USA
| | - K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson MS-68198, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson MS-68198, USA
| | - Faizan Abul Qais
- Department of Agricultural Microbiology, Faculty of Agricultural Sciences, Aligarh Muslim University, Aligarh, UP 202002, India
| | - Parvez Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE-68198, USA
| | - Jesse L Cox
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha NE-68198, USA
| | - Tarique Sarwar
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE-68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha NE-68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha NE-68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha NE-68198, USA
| | - Jawed A Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson MS-68198, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson MS-68198, USA.
| |
Collapse
|
10
|
Chen W, Bedar M, Zhou Q, Ren X, Kang Y, Huang KX, Rubino G, Kolliopoulos V, Moghadam S, Cascavita CT, Taylor JM, Chevalier JM, Harley BA, Lee JC. Correlating Material Properties to Osteoprotegerin Expression on Nanoparticulate Mineralized Collagen Glycosaminoglycan Scaffolds. Adv Healthc Mater 2024; 13:e2401037. [PMID: 38885525 PMCID: PMC11489015 DOI: 10.1002/adhm.202401037] [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: 04/02/2024] [Revised: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Precision material design directed by cell biological processes represents a frontier in developing clinically translatable regenerative technologies. While understanding cell-material interactions on multipotent progenitor cells yields insights on target tissue differentiation, equally if not more important is the quantification of indirect multicellular interactions. In this work, the relationship of two material properties, phosphate content and stiffness, of a nanoparticulate mineralized collagen glycosaminoglycan scaffold (MC-GAG) in the expression of an endogenous anti-osteoclastogenic secreted protein, osteoprotegerin (OPG) by primary human mesenchymal stem cells (hMSCs) is evaluated. The phosphate content of MC-GAG requires the type III sodium phosphate symporter PiT-1/SLC20A1 for OPG expression, correlating with β-catenin downregulation, but is independent of the effects of phosphate ion on osteogenic differentiation. Using three stiffness MC-GAG variants that do not differ significantly by osteogenic differentiation, it is observed that the softest material elicited ≈1.6-2 times higher OPG expression than the stiffer materials. Knockdown of the mechanosensitive signaling axis of YAP, TAZ, β-catenin and combinations thereof in hMSCs on MC-GAG demonstrates that β-catenin downregulation increases OPG expression by 1.5-fold. Taken together, these data constitute a roadmap for material properties that can used to suppress osteoclast activation via osteoprotegerin expression separately from the anabolic processes of osteogenesis.
Collapse
Affiliation(s)
- Wei Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Meiwand Bedar
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Qi Zhou
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Xiaoyan Ren
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Youngnam Kang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Kelly X. Huang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Grace Rubino
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Vasiliki Kolliopoulos
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Shahrzad Moghadam
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Catherine T. Cascavita
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Jeremiah M. Taylor
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Jose M. Chevalier
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Department of Orthopaedic Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Surgery and Perioperative Care, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| |
Collapse
|
11
|
Zhang D, Jin X, Ma X, Qiu Y, Ma W, Dai X, Zhang Z. Tumour necrosis factor α regulates the miR-27a-3p-Sfrp1 axis in a mouse model of osteoporosis. Exp Physiol 2024; 109:1109-1123. [PMID: 38748896 PMCID: PMC11215474 DOI: 10.1113/ep090311] [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: 01/05/2022] [Accepted: 03/01/2024] [Indexed: 07/02/2024]
Abstract
Osteoporosis is a metabolic bone disease that involves gradual loss of bone density and mass, thus resulting in increased fragility and risk of fracture. Inflammatory cytokines, such as tumour necrosis factor α (TNF-α), inhibit osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), and several microRNAs are implicated in osteoporosis development. This study aimed to explore the correlation between TNF-α treatment and miR-27a-3p expression in BMSC osteogenesis and further understand their roles in osteoporosis. An osteoporosis animal model was established using ovariectomized (OVX) mice. Compared with Sham mice, the OVX mice had a significantly elevated level of serum TNF-α and decreased level of bone miR-27a-3p, and in vitro TNF-α treatment inhibited miR-27a-3p expression in BMSCs. In addition, miR-27a-3p promoted osteogenic differentiation of mouse BMSCs in vitro, as evidenced by alkaline phosphatase staining and Alizarin Red-S staining, as well as enhanced expression of the osteogenic markers Runx2 and Osterix. Subsequent bioinformatics analysis combined with experimental validation identified secreted frizzled-related protein 1 (Sfrp1) as a downstream target of miR-27a-3p. Sfrp1 overexpression significantly inhibited the osteogenic differentiation of BMSCs in vitro and additional TNF-α treatment augmented this inhibition. Moreover, Sfrp1 overexpression abrogated the promotive effect of miR-27a-3p on the osteogenic differentiation of BMSCs. Furthermore, the miR-27a-3p-Sfrp1 axis was found to exert its regulatory function in BMSC osteogenic differentiation via regulating Wnt3a-β-catenin signalling. In summary, this study revealed that TNF-α regulated a novel miR-27a-3p-Sfrp1 axis in osteogenic differentiation of BMSCs. The data provide new insights into the development of novel therapeutic strategies for osteoporosis.
Collapse
Affiliation(s)
- Dang‐Feng Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Xiao‐Na Jin
- Department of NursingXi'an International UniversityXi'anShaanxiChina
| | - Xing Ma
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Yu‐Sheng Qiu
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Wei Ma
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Xing Dai
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Zhi Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
12
|
Hrubi E, Imre L, Hegedüs C. Effects of EZH2 inhibitor, trichostatin A, and 5-azacytidine combinatorial treatment on osteogenic differentiation of dental pulp stem cells. Heliyon 2024; 10:e32553. [PMID: 39183840 PMCID: PMC11341346 DOI: 10.1016/j.heliyon.2024.e32553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 08/27/2024] Open
Abstract
Objective Epigenetic mechanisms play regulatory roles in dental pulp stem cell (DPSC) differentiation. The molecules that modulate these mechanisms can be used to enhance DPSC differentiation in experimental studies and clinical applications. We investigated the combined effects of an epigenetic modulator enhancer of zeste homologue 2 inhibitor (EZH2i), trichostatin A (TSA), and 5-azacytidine (5-AZA) on the osteogenic differentiation of DPSCs. Results To assess osteogenic differentiation, we measured alkaline phosphatase activity, calcium deposition, and expression of osteogenic differentiation marker genes (RUNX2, BMP2, and ALPL) after 7 or 21 days of combinatorial drug treatment in normal cell culture medium or osteo-inductive medium (OIM). No synergistic effects were observed for any possible combination of EZH2i, TSA, or 5-AZA. However, the effects of these drugs and their combinations depend on the time and culture conditions. Discussion We confirmed that EZH2i and TSA have positive effects on the osteogenic differentiation of DPSCs. EZH2i activates the expression of key regulatory genes (RUNX2, BMP2, and ALPL) directly, whereas TSA interacts with signalling pathways induced by supplements in OIM to activate these genes.
Collapse
Affiliation(s)
- Edit Hrubi
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, Debrecen, H-4032 Hungary
| | - László Imre
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Csaba Hegedüs
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, Debrecen, H-4032 Hungary
| |
Collapse
|
13
|
Berger MB, Bosh K, Deng J, Jacobs TW, Cohen DJ, Boyan BD, Schwartz Z. Wnt16 Increases Bone-to-Implant Contact in an Osteopenic Rat Model by Increasing Proliferation and Regulating the Differentiation of Bone Marrow Stromal Cells. Ann Biomed Eng 2024; 52:1744-1762. [PMID: 38517621 PMCID: PMC11082046 DOI: 10.1007/s10439-024-03488-y] [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: 04/24/2023] [Accepted: 03/07/2024] [Indexed: 03/24/2024]
Abstract
Osseointegration is a complex biological cascade that regulates bone regeneration after implant placement. Implants possessing complex multiscale surface topographies augment this regenerative process through the regulation of bone marrow stromal cells (MSCs) that are in contact with the implant surface. One pathway regulating osteoblastic differentiation is Wnt signaling, and upregulation of non-canonical Wnts increases differentiation of MSCs on these titanium substrates. Wnt16 is a non-canonical Wnt shown to regulate bone morphology in mouse models. This study evaluated the role of Wnt16 during surface-mediated osteoblastic differentiation of MSCs in vitro and osseointegration in vivo. MSCs were cultured on Ti substrates with different surface properties and non-canonical Wnt expression was determined. Subsequently, MSCs were cultured on Ti substrates +/-Wnt16 (100 ng/mL) and anti-Wnt16 antibodies (2 μg/mL). Wnt16 expression was increased in cells grown on microrough surfaces that were processed to be hydrophilic and have nanoscale roughness. However, treatment MSCs on these surfaces with exogenous rhWnt16b increased total DNA content and osteoprotegerin production, but reduced osteoblastic differentiation and production of local factors necessary for osteogenesis. Addition of anti-Wnt16 antibodies blocked the inhibitor effects of Wnt16. The response to Wnt16 was likely independent of other osteogenic pathways like Wnt11-Wnt5a signaling and semaphorin 3a signaling. We used an established rat model of cortical and trabecular femoral bone impairment following botox injections (2 injections of 8 units/leg each, starting and maintenance doses) to assess Wnt16 effects on whole bone morphology and implant osseointegration. Wnt16 injections did not alter whole bone morphology significantly (BV/TV, cortical thickness, restoration of trabecular bone) but were effective at increasing cortical bone-to-implant contact during impaired osseointegration in the botox model. The mechanical quality of the increased bone was not sufficient to rescue the deleterious effects of botox. Clinically, these results are important to understand the interaction of cortical and trabecular bone during implant integration. They suggest a role for Wnt16 in modulating bone remodeling by reducing osteoclastic activity. Targeted strategies to temporally regulate Wnt16 after implant placement could be used to improve osseointegration by increasing the net pool of osteoprogenitor cells.
Collapse
Affiliation(s)
- Michael B Berger
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Kyla Bosh
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Jingyao Deng
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Thomas W Jacobs
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - D Joshua Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA.
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA, 23284, USA
- Department of Periodontology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| |
Collapse
|
14
|
Li Z, Wang D, Li J, Liu H, Nie L, Li C. Bone Regeneration Facilitated by Autologous Bioscaffold Material: Liquid Phase of Concentrated Growth Factor with Dental Follicle Stem Cell Loading. ACS Biomater Sci Eng 2024; 10:3173-3187. [PMID: 38605468 DOI: 10.1021/acsbiomaterials.3c01981] [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] [Indexed: 04/13/2024]
Abstract
The application of bioengineering techniques for achieving bone regeneration in the oral environment is an increasingly prominent field. However, the clinical use of synthetic materials carries certain risks. The liquid phase of concentrated growth factor (LPCGF), as a biologically derived material, exhibits superior biocompatibility. In this study, LPCGF was employed as a tissue engineering scaffold, hosting dental follicle cells (DFCs) to facilitate bone regeneration. Both in vivo and in vitro experimental results demonstrate that this platform significantly enhances the expression of osteogenic markers in DFCs, such as alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and type I collagen (Col1a1). Simultaneously, it reduces the expression of inflammation-related genes, particularly interleukin-6 (IL-6) and interleukin-8 (IL-8), thereby alleviating the negative impact of the inflammatory microenvironment on DFCs. Further investigation into potential mechanisms reveals that this process is regulated over time by the WNT pathway. Our research results demonstrate that LPCGF, with its favorable physical characteristics, holds great potential as a scaffold. It can effectively carry DFCs, thereby providing an optimal initial environment for bone regeneration. Furthermore, LPCGF endeavors to closely mimic the mechanisms of bone healing post-trauma to facilitate bone formation. This offers new perspectives and insights into bone regeneration engineering.
Collapse
Affiliation(s)
- Zhentao Li
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Di Wang
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Hao Liu
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Li Nie
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| | - Conghua Li
- Stomatological Hospital of Chongqing Medical University, No. 426 Songshi North Road, Yubei District, Chongqing 401147, China
| |
Collapse
|
15
|
Olali AZ, Wallace J, Gonzalez H, Carpenter KA, Patel N, Winchester LC, Podany AT, Venkatesh I, Narasipura SD, Al-Harthi L, Ross RD. The anti-HIV drug abacavir stimulates β-catenin activity in osteoblast lineage cells. JBMR Plus 2024; 8:ziae037. [PMID: 38590756 PMCID: PMC11001392 DOI: 10.1093/jbmrpl/ziae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/28/2024] [Accepted: 03/14/2024] [Indexed: 04/10/2024] Open
Abstract
Bone mineral density (BMD) loss in people living with HIV occurs with the initiation of combined antiretroviral therapy (cART), particularly with tenofovir disoproxil fumarate (TDF) containing cART. Switching from TDF to abacavir (ABC) or dolutegravir (DTG) leads to increased BMD. Whether BMD gains are due to cessation of TDF or anabolic effects of ABC or DTG is unclear. We investigated the effects of ABC and DTG on osteoblast lineage cells in vitro and in vivo. Primary human osteoblasts and male C57BL/6 mice were treated with individual antiretrovirals (ARVs) or a combination of ABC/DTG/lamivudine (3TC). Nearly all ARVs and cART inhibited osteogenic activity in vitro. Due to the importance of Wnt/β-catenin in bone formation, we further investigated ARV effects on the Wnt/β-catenin pathway. ABC, alone and as part of ABC/DTG/3TC, increased osteoblastic β-catenin activity as indicated by increased TOPFlash activity, hypo-phosphorylated (active) β-catenin staining, and β-catenin targeted gene expression. Mice treated with TDF had decreased lumbar spine BMD and trabecular connectivity density in the vertebrae, while those treated with ABC/DTG/3TC reduced cortical area and thickness in the femur. Mice treated with ABC alone had no bone structural changes, increased circulating levels of the bone formation marker, P1NP, and elevated expression of the Wnt/β-catenin target gene, Lef1, in osteocyte enriched samples. Further, bones from ARV-treated mice were isolated to evaluate ARV distribution. All ARVs were detected in the bone tissue, which was inclusive of bone marrow, but when bone marrow was removed, only TDF, ABC, and DTG were detected at ~0.1% of the circulating levels. Overall, our findings demonstrate that ABC activates Wnt/β-catenin signaling, but whether this leads to increased bone formation requires further study. Assessing the impact of ARVs on bone is critical to informing ARV selection and/or discovery of regimens that do not negatively impact the skeleton.
Collapse
Affiliation(s)
- Arnold Z Olali
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Jennillee Wallace
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Hemil Gonzalez
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
- Division of Infectious Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States
| | - Kelsey A Carpenter
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Niyati Patel
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Lee C Winchester
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Anthony T Podany
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Ishwarya Venkatesh
- Department of Internal Medicine, Drug Discovery Center, Rush University Medical Center, Chicago, IL 60612, United States
| | - Srinivas D Narasipura
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Ryan D Ross
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, United States
| |
Collapse
|
16
|
Petersen SI, Okolicsanyi RK, Haupt LM. Exploring Heparan Sulfate Proteoglycans as Mediators of Human Mesenchymal Stem Cell Neurogenesis. Cell Mol Neurobiol 2024; 44:30. [PMID: 38546765 PMCID: PMC10978659 DOI: 10.1007/s10571-024-01463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/19/2024] [Indexed: 04/01/2024]
Abstract
Alzheimer's disease (AD) and traumatic brain injury (TBI) are major public health issues worldwide, with over 38 million people living with AD and approximately 48 million people (27-69 million) experiencing TBI annually. Neurodegenerative conditions are characterised by the accumulation of neurotoxic amyloid beta (Aβ) and microtubule-associated protein Tau (Tau) with current treatments focused on managing symptoms rather than addressing the underlying cause. Heparan sulfate proteoglycans (HSPGs) are a diverse family of macromolecules that interact with various proteins and ligands and promote neurogenesis, a process where new neural cells are formed from stem cells. The syndecan (SDC) and glypican (GPC) HSPGs have been implicated in AD pathogenesis, acting as drivers of disease, as well as potential therapeutic targets. Human mesenchymal stem cells (hMSCs) provide an attractive therapeutic option for studying and potentially treating neurodegenerative diseases due to their relative ease of isolation and subsequent extensive in vitro expansive potential. Understanding how HSPGs regulate protein aggregation, a key feature of neurodegenerative disorders, is essential to unravelling the underlying disease processes of AD and TBI, as well as any link between these two neurological disorders. Further research may validate HSPG, specifically SDCs or GPCs, use as neurodegenerative disease targets, either via driving hMSC stem cell therapy or direct targeting.
Collapse
Affiliation(s)
- Sofia I Petersen
- Stem Cell and Neurogenesis Group, School of Biomedical Sciences, Genomics Research Centre, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Rachel K Okolicsanyi
- Stem Cell and Neurogenesis Group, School of Biomedical Sciences, Genomics Research Centre, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices, Kelvin Grove, Australia
| | - Larisa M Haupt
- Stem Cell and Neurogenesis Group, School of Biomedical Sciences, Genomics Research Centre, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia.
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Kelvin Grove, Australia.
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices, Kelvin Grove, Australia.
| |
Collapse
|
17
|
Knill C, Henderson EJ, Johnson C, Wah VY, Cheng K, Forster AJ, Itasaki N. Defects of the spliceosomal gene SNRPB affect osteo- and chondro-differentiation. FEBS J 2024; 291:272-291. [PMID: 37584444 DOI: 10.1111/febs.16934] [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: 03/06/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023]
Abstract
Although gene splicing occurs throughout the body, the phenotype of spliceosomal defects is largely limited to specific tissues. Cerebro-costo-mandibular syndrome (CCMS) is one such spliceosomal disease, which presents as congenital skeletal dysmorphism and is caused by mutations of SNRPB gene encoding Small Nuclear Ribonucleoprotein Polypeptides B/B' (SmB/B'). This study employed in vitro cell cultures to monitor osteo- and chondro-differentiation and examined the role of SmB/B' in the differentiation process. We found that low levels of SmB/B' by knockdown or mutations of SNRPB led to suppressed osteodifferentiation in Saos-2 osteoprogenitor-like cells, which was accompanied by affected splicing of Dlx5. On the other hand, low SmB/B' led to promoted chondrogenesis in HEPM mesenchymal stem cells. Consistent with other reports, osteogenesis was promoted by the Wnt/β-catenin pathway activator and suppressed by Wnt and BMP blockers, whereas chondrogenesis was promoted by Wnt inhibitors. Suppressed osteogenic markers by SNRPB knockdown were partly rescued by Wnt/β-catenin pathway activation. Reporter analysis revealed that suppression of SNRPB results in attenuated Wnt pathway and/or enhanced BMP pathway activities. SNRPB knockdown altered splicing of TCF7L2 which impacts Wnt/β-catenin pathway activities. This work helps unravel the mechanism underlying CCMS whereby reduced expression of spliceosomal proteins causes skeletal phenotypes.
Collapse
Affiliation(s)
- Chris Knill
- Faculty of Life Sciences, University of Bristol, UK
| | | | - Craig Johnson
- Faculty of Health Sciences, University of Bristol, UK
| | - Vun Yee Wah
- Faculty of Life Sciences, University of Bristol, UK
| | - Kevin Cheng
- Faculty of Life Sciences, University of Bristol, UK
| | | | - Nobue Itasaki
- Faculty of Health Sciences, University of Bristol, UK
| |
Collapse
|
18
|
Zhang X, Lin H, Zheng DL, Lu YG, Zou Y, Su B. Exploring the Role of Wnt Ligands in Osteogenic Differentiation of Human Periodontal Ligament Stem Cells. Clin Oral Investig 2023; 28:64. [PMID: 38158464 DOI: 10.1007/s00784-023-05449-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVES This study aimed to investigate the functions of 19 types of Wnt ligands during the process of osteogenic differentiation in human periodontal ligament stem cells (hPDLSCs), with particular attention to WNT3A and WNT4. MATERIALS AND METHODS The expression levels of 19 types of Wnt ligands were examined using real-time quantitative polymerase chain reaction (real-time qPCR) during hPDLSCs osteogenic differentiation at 7, 10, and 14 days. Knockdown of WNT3A and WNT4 expression was achieved using adenovirus vectors, and conditioned medium derived from WNT3A and WNT4 overexpression plasmids was employed to investigate their roles in hPDLSCs osteogenesis. Osteogenic-specific genes were analyzed using real-time qPCR. Alkaline phosphatase (ALP) and alizarin red S activities and staining were employed to assess hPDLSCs' osteogenic differentiation ability. RESULTS During hPDLSCs osteogenic differentiation, the expression of 19 types of Wnt ligands varied, with WNT3A and WNT4 showing significant upregulation. Inhibiting WNT3A and WNT4 expression hindered hPDLSCs' osteogenic capacity. Conditioned medium of WNT3A promoted early osteogenic differentiation, while WNT4 facilitated late osteogenesis slightly. CONCLUSION Wnt ligands, particularly WNT3A and WNT4, play an important role in hPDLSCs' osteogenic differentiation, highlighting their potential as promoters of osteogenesis. CLINICAL RELEVANCE Given the challenging nature of alveolar bone regeneration, therapeutic strategies that target WNT3A and WNT4 signaling pathways offer promising opportunities. Additionally, innovative gene therapy approaches aimed at regulating of WNT3A and WNT4 expression hold potential for improving alveolar bone regeneration outcomes.
Collapse
Affiliation(s)
- Xiao Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
| | - Hanrui Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
| | - Da-Li Zheng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
| | - You-Guang Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China
| | - Yuchun Zou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China.
- Department of Orthodontics, Fujian Key Laboratory of Oral Diseases, School of Stomatology, Fujian Medical University, Fuzhou, 350001, China.
| | - Bohua Su
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China.
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350001, China.
| |
Collapse
|
19
|
Huang Y, Peng Y, Li H, Li C, Wu Y, Wang X, Chang J, Miao C. Wilforine inhibits rheumatoid arthritis pathology through the Wnt11/β-catenin signaling pathway axis. Arthritis Res Ther 2023; 25:243. [PMID: 38098062 PMCID: PMC10720104 DOI: 10.1186/s13075-023-03224-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Wilforine (WFR) is a monomeric compound of the anti-RA plant Tripterygium wilfordii Hook. f. (TwHF). Whether WFR has anti-RA effect, its molecular mechanism has not been elucidated. AIM OF THE STUDY Our study aims to clarify how WFR inhibits fibroblast-like synovial cells (FLS) activation and improves RA through Wnt11 action on the Wnt11/β-catenin signaling pathway. METHODS The therapeutic effect of WFR on collagen-induced arthritis (CIA) rats was evaluated using methods such as rat arthritis score. The inhibitory effects and signaling pathways of WFR on the proliferation and inflammatory response of CIA FLS and RA FLS were studied using ELISA, CCK-8, RT-qPCR, Western blot, and immunofluorescence methods. RESULTS WFR could effectively alleviate the arthritis symptoms of CIA rats; reduce the levels of IL-6, IL-1β, and TNF-α in the peripheral blood of CIA rats; and inhibit the expression of MMP3 and fibronectin. The data showed that WFR has a significant inhibitory effect on FLS proliferation. Furthermore, WFR inhibited the activation of Wnt/β-catenin signaling pathway and decreased the expression of Wnt11, β-catenin, CCND1, GSK-3β, and c-Myc, while the effects of WFR were reversed after overexpression of Wnt11. CONCLUSIONS WFR improves RA by inhibiting the Wnt11/β-catenin signaling pathway, and Wnt11 is the direct target of WFR. This study provides a new molecular mechanism for WFR to improve RA and contributes to the clinical promotion of WFR.
Collapse
Affiliation(s)
- Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Yanhui Peng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Chen Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China
| | - Xiaomei Wang
- Department of Humanistic Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Jun Chang
- Department of Orthopaedics, the First Affiliated Hospital, Anhui Medical University, Hefei, 230032, China.
- Anhui Public Health Clinical Center, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui Province, China.
| |
Collapse
|
20
|
Alhattab DM, Isaioglou I, Alshehri S, Khan ZN, Susapto HH, Li Y, Marghani Y, Alghuneim AA, Díaz-Rúa R, Abdelrahman S, Al-Bihani S, Ahmed F, Felimban RI, Alkhatabi H, Alserihi R, Abedalthagafi M, AlFadel A, Awidi A, Chaudhary AG, Merzaban J, Hauser CAE. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomater Res 2023; 27:111. [PMID: 37932837 PMCID: PMC10626721 DOI: 10.1186/s40824-023-00457-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy that remains a therapeutic challenge due to the high incidence of disease relapse. To better understand resistance mechanisms and identify novel therapies, robust preclinical models mimicking the bone marrow (BM) microenvironment are needed. This study aimed to achieve an automated fabrication process of a three-dimensional (3D) AML disease model that recapitulates the 3D spatial structure of the BM microenvironment and applies to drug screening and investigational studies. METHODS To build this model, we investigated a unique class of tetramer peptides with an innate ability to self-assemble into stable hydrogel. An automated robotic bioprinting process was established to fabricate a 3D BM (niche-like) multicellular AML disease model comprised of leukemia cells and the BM's stromal and endothelial cellular fractions. In addition, monoculture and dual-culture models were also fabricated. Leukemia cell compatibility, functionalities (in vitro and in vivo), and drug assessment studies using our model were performed. In addition, RNAseq and gene expression analysis using TaqMan arrays were also performed on 3D cultured stromal cells and primary leukemia cells. RESULTS The selected peptide hydrogel formed a highly porous network of nanofibers with mechanical properties similar to the BM extracellular matrix. The robotic bioprinter and the novel quadruple coaxial nozzle enabled the automated fabrication of a 3D BM niche-like AML disease model with controlled deposition of multiple cell types into the model. This model supported the viability and growth of primary leukemic, endothelial, and stromal cells and recapitulated cell-cell and cell-ECM interactions. In addition, AML cells in our model possessed quiescent characteristics with improved chemoresistance attributes, resembling more the native conditions as indicated by our in vivo results. Moreover, the whole transcriptome data demonstrated the effect of 3D culture on enhancing BM niche cell characteristics. We identified molecular pathways upregulated in AML cells in our 3D model that might contribute to AML drug resistance and disease relapse. CONCLUSIONS Our results demonstrate the importance of developing 3D biomimicry models that closely recapitulate the in vivo conditions to gain deeper insights into drug resistance mechanisms and novel therapy development. These models can also improve personalized medicine by testing patient-specific treatments.
Collapse
Affiliation(s)
- Dana M Alhattab
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Ioannis Isaioglou
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Salwa Alshehri
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Zainab N Khan
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Yanyan Li
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yara Marghani
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Arwa A Alghuneim
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Rubén Díaz-Rúa
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Shuroug Al-Bihani
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Farid Ahmed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed I Felimban
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Heba Alkhatabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed Alserihi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, USA
| | - AlShaibani AlFadel
- Division of Hematology, Stem Cell Transplantation & Cellular Therapy, Oncology Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Medical School, The University of Jordan, Amman, Jordan
- Jordan University Hospital, Amman, Jordan
| | - Adeel Gulzar Chaudhary
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Jasmeen Merzaban
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
21
|
Kim HY, Shim JH, Heo CY. A Rare Skeletal Disorder, Fibrous Dysplasia: A Review of Its Pathogenesis and Therapeutic Prospects. Int J Mol Sci 2023; 24:15591. [PMID: 37958575 PMCID: PMC10650015 DOI: 10.3390/ijms242115591] [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: 09/16/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Fibrous dysplasia (FD) is a rare, non-hereditary skeletal disorder characterized by its chronic course of non-neoplastic fibrous tissue buildup in place of healthy bone. A myriad of factors have been associated with its onset and progression. Perturbation of cell-cell signaling networks and response outputs leading to disrupted building blocks, incoherent multi-level organization, and loss of rigid structural motifs in mineralized tissues are factors that have been identified to participate in FD induction. In more recent years, novel insights into the unique biology of FD are transforming our understandings of its pathology, natural discourse of the disease, and treatment prospects. Herein, we built upon existing knowledge with recent findings to review clinical, etiologic, and histological features of FD and discussed known and potential mechanisms underlying FD manifestations. Subsequently, we ended on a note of optimism by highlighting emerging therapeutic approaches aimed at either halting or ameliorating disease progression.
Collapse
Affiliation(s)
- Ha-Young Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Jung-Hee Shim
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
- Department of Research Administration Team, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Chan-Yeong Heo
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| |
Collapse
|
22
|
Hammouda DA, Mansour AM, Saeed MA, Zaher AR, Grawish ME. Stem cell-derived exosomes for dentin-pulp complex regeneration: a mini-review. Restor Dent Endod 2023; 48:e20. [PMID: 37284341 PMCID: PMC10240090 DOI: 10.5395/rde.2023.48.e20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/12/2023] [Accepted: 03/22/2023] [Indexed: 06/08/2023] Open
Abstract
This mini-review was conducted to present an overview of the use of exosomes in regenerating the dentin-pulp complex (DPC). The PubMed and Scopus databases were searched for relevant articles published between January 1, 2013 and January 1, 2023. The findings of basic in vitro studies indicated that exosomes enhance the proliferation and migration of mesenchymal cells, as human dental pulp stem cells, via mitogen-activated protein kinases and Wingless-Int signaling pathways. In addition, they possess proangiogenic potential and contribute to neovascularization and capillary tube formation by promoting endothelial cell proliferation and migration of human umbilical vein endothelial cells. Likewise, they regulate the migration and differentiation of Schwann cells, facilitate the conversion of M1 pro-inflammatory macrophages to M2 anti-inflammatory phenotypes, and mediate immune suppression as they promote regulatory T cell conversion. Basic in vivo studies have indicated that exosomes triggered the regeneration of dentin-pulp-like tissue, and exosomes isolated under odontogenic circumstances are particularly strong inducers of tissue regeneration and stem cell differentiation. Exosomes are a promising regenerative tool for DPC in cases of small pulp exposure or for whole-pulp tissue regeneration.
Collapse
Affiliation(s)
- Dina A. Hammouda
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Alaa M Mansour
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Mahmoud A. Saeed
- Department of Oral Biology, Faculty of Dentistry, Menoufia University, Shibin el Kom, Egypt
| | - Ahmed R. Zaher
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Mohammed E. Grawish
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
- Department of Oral Biology, Faculty of Oral and Dental Medicine, Delta University for Science and Technology, Dakahlia, Egypt
| |
Collapse
|
23
|
Shen L, He Y, Chen S, He L, Zhang Y. PTHrP Modulates the Proliferation and Osteogenic Differentiation of Craniofacial Fibrous Dysplasia-Derived BMSCs. Int J Mol Sci 2023; 24:ijms24087616. [PMID: 37108778 PMCID: PMC10146947 DOI: 10.3390/ijms24087616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Fibrous dysplasia (FD) is a skeletal stem cell disease caused by mutations in the guanine nucleotide-binding protein, alpha-stimulating activity polypeptide (GNAS) gene, which results in the abnormal accumulation of cyclic adenosine monophosphate (cAMP) and hyperactivation of downstream signaling pathways. Parathyroid hormone-related protein (PTHrP) is secreted by the osteoblast lineage and is involved in various physiological and pathological activities of bone. However, the association between the abnormal expression of PTHrP and FD, as well as its underlying mechanism, remains unclear. In this study, we discovered that FD patient-derived bone marrow stromal cells (FD BMSCs) expressed significantly higher levels of PTHrP during osteogenic differentiation and exhibited greater proliferation capacity but impaired osteogenic ability compared to normal control patient-derived BMSCs (NC BMSCs). Continuous exogenous PTHrP exposure on the NC BMSCs promoted the FD phenotype in both in vitro and in vivo experiments. Through the PTHrP/cAMP/PKA axis, PTHrP could partially influence the proliferation and osteogenesis capacity of FD BMSCs via the overactivation of the Wnt/β-Catenin signaling pathway. Furthermore, PTHrP not only directly modulated cAMP/PKA/CREB transduction but was also demonstrated as a transcriptional target of CREB. This study provides novel insight into the possible pathogenesis involved in the FD phenotype and enhances the understanding of its molecular signaling pathways, offering theoretical evidence for the feasibility of potential therapeutic targets for FD.
Collapse
Affiliation(s)
- Lihang Shen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Shuo Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Linhai He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
- First Clinical Division, Peking University School and Hospital of Stomatology, Beijing 100034, China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
24
|
Xu K, Zhang L, Yu N, Ren Z, Wang T, Zhang Y, Zhao X, Yu T. Effects of advanced glycation end products (AGEs) on the differentiation potential of primary stem cells: a systematic review. Stem Cell Res Ther 2023; 14:74. [PMID: 37038234 PMCID: PMC10088298 DOI: 10.1186/s13287-023-03324-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/27/2023] [Indexed: 04/12/2023] Open
Abstract
The formation and accumulation of advanced glycation end products (AGEs) have been associated with aging and the development, or worsening, of many degenerative diseases, such as atherosclerosis, chronic kidney disease, and diabetes. AGEs can accumulate in a variety of cells and tissues, and organs in the body, which in turn induces oxidative stress and inflammatory responses and adversely affects human health. In addition, under abnormal pathological conditions, AGEs create conditions that are not conducive to stem cell differentiation. Moreover, an accumulation of AGEs can affect the differentiation of stem cells. This, in turn, leads to impaired tissue repair and further aggravation of diabetic complications. Therefore, this systematic review clearly outlines the effects of AGEs on cell differentiation of various types of primary isolated stem cells and summarizes the possible regulatory mechanisms and interventions. Our study is expected to reveal the mechanism of tissue damage caused by the diabetic microenvironment from a cellular and molecular point of view and provide new ideas for treating complications caused by diabetes.
Collapse
Affiliation(s)
- Kuishuai Xu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Liang Zhang
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Ning Yu
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Zhongkai Ren
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Tianrui Wang
- Department of Traumatology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Yingze Zhang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xia Zhao
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
25
|
Administration of stem cells against cardiovascular diseases with a focus on molecular mechanisms: Current knowledge and prospects. Tissue Cell 2023; 81:102030. [PMID: 36709696 DOI: 10.1016/j.tice.2023.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
Cardiovascular diseases (CVDs) are a serious global concern for public and human health. Despite the emergence of significant therapeutic advances, it is still the leading cause of death and disability worldwide. As a result, extensive efforts are underway to develop practical therapeutic approaches. Stem cell-based therapies could be considered a promising strategy for the treatment of CVDs. The efficacy of stem cell-based therapeutic approaches is demonstrated through recent laboratory and clinical studies due to their inherent regenerative properties, proliferative nature, and their capacity to differentiate into different cells such as cardiomyocytes. These properties could improve cardiovascular functioning leading to heart regeneration. The two most common types of stem cells with the potential to cure heart diseases are induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs). Several studies have demonstrated the use, efficacy, and safety of MSC and iPSCs-based therapies for the treatment of CVDs. In this study, we explain the application of stem cells, especially iPSCs and MSCs, in the treatment of CVDs with a focus on cellular and molecular mechanisms and then discuss the advantages, disadvantages, and perspectives of using this technology in the treatment of these diseases.
Collapse
|
26
|
Sukarawan W, Rattanawarawipa P, Yaemkleebbua K, Nowwarote N, Pavasant P, Limjeerajarus CN, Osathanon T. Wnt3a promotes odonto/osteogenic differentiation in vitro and tertiary dentin formation in a rat model. Int Endod J 2023; 56:514-529. [PMID: 36633501 DOI: 10.1111/iej.13888] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023]
Abstract
AIM To investigate the effect of Wnt3a on odonto/osteogenic differentiation of stem cells isolated from human exfoliated deciduous teeth (SHEDs) and reparative dentine formation in a rat model. METHODOLOGY Stem cells isolated from human exfoliated deciduous teeth were cultured in media with Wnt3a (50-200 ng/ml). Wnt activation was confirmed by β-catenin immunocytochemistry. Colony-forming unit assay (normalized percentage area), osteogenic gene expression analysis by real-time polymerase chain reaction and mineralization assays measured by the absorption at 540 nm were performed. Tertiary dentine formation in vivo was evaluated using 8-week-old, male Wistar rats. Cavities with pinpoint pulp exposure by a sharp instrument were prepared at the mesial surface of the first molars. Teeth were divided into (n = 6): (1) distilled water (negative control), (2) phosphate-buffered saline (PBS), (3) lithium chloride in DI (20 μM), and (4) Wnt3a in PBS (200 ng/ml). Collagen sponge was used as a scaffold. The cavity was sealed with glass ionomer restoration. Four weeks later, animals were euthanized by sodium pentobarbital (120 mg/kg body weight). Hard tissue formation was evaluated using micro-computerized tomography. Sixty consecutive slides from the initial plane were analysed and calculated as bone/dentine volume per total tissue volume. Paraffin sections (2 μm) were stained with haematoxylin and eosin and Masson's trichrome for morphological evaluation. Data are presented as the mean ± standard error. Mann-Whitney U test was used for two-group comparison. Kruskal Wallis followed by pairwise comparison was employed for three or more group comparisons. Statistical analysis was performed using GraphPad Prism 7. Differences were considered significant at p < .05. RESULTS Wnt3a decreased SHEDs colony formation and increased OSX, BMP2, and DMP1 expression, corresponding to an increase in mineralization. Additionally, a significant increase in dentine/bone volume per total tissue volume was observed in Wnt3a treated defects. Dentine bridge formation at the exposure sites treated with Wnt3a demonstrated, while fibrous tissues were observed in the control. CONCLUSIONS Wnt3a suppressed proliferation, increased osteogenic differentiation of SHEDs and promotes tertiary dentine formation. Wnt3a could be utilized as biological molecule for vital pulp therapy.
Collapse
Affiliation(s)
- Waleerat Sukarawan
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Panarat Rattanawarawipa
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Kamonwan Yaemkleebbua
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Nunthawan Nowwarote
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM UMRS 1138, Molecular Oral Pathophysiology, Paris, France.,Dental Faculty Garancière, Oral Biology Department, Université de Paris, Paris, France
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chalida Nakalekha Limjeerajarus
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
27
|
Nellinger S, Kluger PJ. How Mechanical and Physicochemical Material Characteristics Influence Adipose-Derived Stem Cell Fate. Int J Mol Sci 2023; 24:ijms24043551. [PMID: 36834966 PMCID: PMC9961531 DOI: 10.3390/ijms24043551] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are a subpopulation of mesenchymal stem cells. Compared to bone marrow-derived stem cells, they can be harvested with minimal invasiveness. ASCs can be easily expanded and were shown to be able to differentiate into several clinically relevant cell types. Therefore, this cell type represents a promising component in various tissue engineering and medical approaches (e.g., cell therapy). In vivo cells are surrounded by the extracellular matrix (ECM) that provides a wide range of tissue-specific physical and chemical cues, such as stiffness, topography, and chemical composition. Cells can sense the characteristics of their ECM and respond to them in a specific cellular behavior (e.g., proliferation or differentiation). Thus, in vitro biomaterial properties represent an important tool to control ASCs behavior. In this review, we give an overview of the current research in the mechanosensing of ASCs and current studies investigating the impact of material stiffens, topography, and chemical modification on ASC behavior. Additionally, we outline the use of natural ECM as a biomaterial and its interaction with ASCs regarding cellular behavior.
Collapse
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra Juliane Kluger
- School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-07121-271-2061
| |
Collapse
|
28
|
Marini F, Giusti F, Palmini G, Brandi ML. Role of Wnt signaling and sclerostin in bone and as therapeutic targets in skeletal disorders. Osteoporos Int 2023; 34:213-238. [PMID: 35982318 DOI: 10.1007/s00198-022-06523-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/01/2022] [Indexed: 01/24/2023]
Abstract
UNLABELLED Wnt signaling and its bone tissue-specific inhibitor sclerostin are key regulators of bone homeostasis. The therapeutic potential of anti-sclerostin antibodies (Scl-Abs), for bone mass recovery and fragility fracture prevention in low bone mass phenotypes, has been supported by animal studies. The Scl-Ab romosozumab is currently used for osteoporosis treatment. INTRODUCTION Wnt signaling is a key regulator of skeletal development and homeostasis; germinal mutations affecting genes encoding components, inhibitors, and enhancers of the Wnt pathways were shown to be responsible for the development of rare congenital metabolic bone disorders. Sclerostin is a bone tissue-specific inhibitor of the Wnt/β-catenin pathway, secreted by osteocytes, negatively regulating osteogenic differentiation and bone formation, and promoting osteoclastogenesis and bone resorption. PURPOSE AND METHODS Here, we reviewed current knowledge on the role of sclerostin and Wnt pathways in bone metabolism and skeletal disorders, and on the state of the art of therapy with sclerostin-neutralizing antibodies in low-bone-mass diseases. RESULTS Various in vivo studies on animal models of human low-bone-mass diseases showed that targeting sclerostin to recover bone mass, restore bone strength, and prevent fragility fracture was safe and effective in osteoporosis, osteogenesis imperfecta, and osteoporosis pseudoglioma. Currently, only treatment with romosozumab, a humanized monoclonal anti-sclerostin antibody, has been approved in human clinical practice for the treatment of osteoporosis, showing a valuable capability to increase BMD at various skeletal sites and reduce the occurrence of new vertebral, non-vertebral, and hip fragility fractures in treated male and female osteoporotic patients. CONCLUSIONS Preclinical studies demonstrated safety and efficacy of therapy with anti-sclerostin monoclonal antibodies in the preservation/restoration of bone mass and prevention of fragility fractures in low-bone-mass clinical phenotypes, other than osteoporosis, to be validated by clinical studies for their approved translation into prevalent clinical practice.
Collapse
Affiliation(s)
- Francesca Marini
- Fondazione FIRMO Onlus, Italian Foundation for the Research on Bone Diseases, Via San Gallo 123, 50129, Florence, Italy
| | - Francesca Giusti
- Donatello Bone Clinic, Villa Donatello Hospital, Sesto Fiorentino, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Fondazione FIRMO Onlus, Italian Foundation for the Research on Bone Diseases, Via San Gallo 123, 50129, Florence, Italy.
- Donatello Bone Clinic, Villa Donatello Hospital, Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
29
|
Bulte JWM, Wang C, Shakeri-Zadeh A. In Vivo Cellular Magnetic Imaging: Labeled vs. Unlabeled Cells. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2207626. [PMID: 36589903 PMCID: PMC9798832 DOI: 10.1002/adfm.202207626] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Indexed: 06/17/2023]
Abstract
Superparamagnetic iron oxide (SPIO)-labeling of cells has been applied for magnetic resonance imaging (MRI) cell tracking for over 30 years, having resulted in a dozen or so clinical trials. SPIO nanoparticles are biodegradable and can be broken down into elemental iron, and hence the tolerance of cells to magnetic labeling has been overall high. Over the years, however, single reports have accumulated demonstrating that the proliferation, migration, adhesion and differentiation of magnetically labeled cells may differ from unlabeled cells, with inhibition of chondrocytic differentiation of labeled human mesenchymal stem cells (hMSCs) as a notable example. This historical perspective provides an overview of some of the drawbacks that can be encountered with magnetic labeling. Now that magnetic particle imaging (MPI) cell tracking is emerging as a new in vivo cellular imaging modality, there has been a renaissance in the formulation of SPIO nanoparticles this time optimized for MPI. Lessons learned from the occasional past pitfalls encountered with SPIO-labeling of cells for MRI may expedite possible future clinical translation of (combined) MRI/MPI cell tracking.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chao Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ali Shakeri-Zadeh
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
30
|
Huang C, Dai R, Meng G, Dingkao R, Wang X, Ren W, Ma X, Wu X, Chu M, La Y, Bao P, Guo X, Pei J, Yan P, Liang C. Transcriptome-Wide Study of mRNAs and lncRNAs Modified by m 6A RNA Methylation in the Longissimus Dorsi Muscle Development of Cattle-Yak. Cells 2022; 11:cells11223654. [PMID: 36429081 PMCID: PMC9688506 DOI: 10.3390/cells11223654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/19/2022] Open
Abstract
Cattle-yak is a hybrid F1 generation of cattle and yak, which has a history of more than 3000 years and has shown better production performance and higher economic benefits than those of yaks. However, up to now, there has been no study on the transcriptome-wide m6A methylation profile of bovine skeletal muscle and its potential biological function during muscle development. Here, we observed significant changes in the expression levels of muscle-related marker genes and methylation-related enzymes during the development of cattle-yak, and the overall m6A content in the Longissimus dorsi muscle of 18-month-old cattle-yak decreased significantly. A total of 36,602 peaks, 11,223 genes and 8388 lncRNAs were identified in the two groups, including 2989 differential peaks (427 up-regulated peaks and 2562 down-regulated peaks), 1457 differentially expressed genes (833 up-regulated genes and 624 down-regulated genes) and 857 differentially expressed lncRNAs (293 up-regulated lncRNAs and 564 down-regulated lncRNAs). GO and KEGG analysis revealed that they were significantly enriched in some muscle-related pathways (Wnt signaling pathway and MAPK signaling pathway) and high-altitude adaptation-related pathway (HIF-1 signaling pathway). Moreover, m6A abundance was positively correlated with gene expression levels, while it was negatively correlated with lncRNA expression levels. This indicates that m6A modification played an important role in the Longissimus dorsi muscle development of cattle-yak; however, the regulation mechanism of m6A-modified mRNA and lncRNA may be different. This study was the first report of transcriptome-wide m6A-modified mRNAs and lncRNAs atlas in the Longissimus dorsi muscle development of cattle-yak, one which will provide new perspectives for genetic improvement in bovines.
Collapse
Affiliation(s)
- Chun Huang
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Rongfeng Dai
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Guangyao Meng
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Renqing Dingkao
- Animal Husbandry Station of Gannan Tibetan Autonomous Prefecture, Gannan 747000, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Wenwen Ren
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xiaoming Ma
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xiaoyun Wu
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Min Chu
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Yongfu La
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Pengjia Bao
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xian Guo
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Jie Pei
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Correspondence: (P.Y.); (C.L.)
| | - Chunnian Liang
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Correspondence: (P.Y.); (C.L.)
| |
Collapse
|
31
|
Zhao X, Tang L, Le TP, Nguyen BH, Chen W, Zheng M, Yamaguchi H, Dawson B, You S, Martinez-Traverso IM, Erhardt S, Wang J, Li M, Martin JF, Lee BH, Komatsu Y, Wang J. Yap and Taz promote osteogenesis and prevent chondrogenesis in neural crest cells in vitro and in vivo. Sci Signal 2022; 15:eabn9009. [PMID: 36282910 PMCID: PMC9938793 DOI: 10.1126/scisignal.abn9009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Neural crest cells (NCCs) are multipotent stem cells that can differentiate into multiple cell types, including the osteoblasts and chondrocytes, and constitute most of the craniofacial skeleton. Here, we show through in vitro and in vivo studies that the transcriptional regulators Yap and Taz have redundant functions as key determinants of the specification and differentiation of NCCs into osteoblasts or chondrocytes. Primary and cultured NCCs deficient in Yap and Taz switched from osteogenesis to chondrogenesis, and NCC-specific deficiency for Yap and Taz resulted in bone loss and ectopic cartilage in mice. Yap bound to the regulatory elements of key genes that govern osteogenesis and chondrogenesis in NCCs and directly regulated the expression of these genes, some of which also contained binding sites for the TCF/LEF transcription factors that interact with the Wnt effector β-catenin. During differentiation of NCCs in vitro and NCC-derived osteogenesis in vivo, Yap and Taz promoted the expression of osteogenic genes such as Runx2 and Sp7 but repressed the expression of chondrogenic genes such as Sox9 and Col2a1. Furthermore, Yap and Taz interacted with β-catenin in NCCs to coordinately promote osteoblast differentiation and repress chondrogenesis. Together, our data indicate that Yap and Taz promote osteogenesis in NCCs and prevent chondrogenesis, partly through interactions with the Wnt-β-catenin pathway.
Collapse
Affiliation(s)
- Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Li Tang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Tram P. Le
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Bao H. Nguyen
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, US
| | - Wen Chen
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Brian Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Shuangjie You
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, Texas 77030, USA
| | - Idaliz M. Martinez-Traverso
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, US
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, Texas 77030, USA
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - James F. Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, US
- Texas Heart Institute, Houston, Texas 77030, USA
| | - Brendan H. Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, Texas 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, Texas 77030, USA
| |
Collapse
|
32
|
Dienelt A, Keller KC, zur Nieden NI. High glucose impairs osteogenic differentiation of embryonic stem cells via early diversion of beta-catenin from Forkhead box O to T cell factor interaction. Birth Defects Res 2022; 114:1056-1074. [PMID: 36164276 PMCID: PMC9708100 DOI: 10.1002/bdr2.2085] [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: 02/01/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Diabetes, which is characterized by an increase in blood glucose concentration, is accompanied by low bone turnover, increased fracture risk, and the formation of embryonic skeletal malformations. Yet, there are few studies elucidating the underlying alterations in signaling pathways leading to these osteogenic defects. We hypothesized here that bone formation deficiencies in a high glucose environment result from altered activity of beta-catenin (CTNNB1), a key contributor to osteogenic differentiation, dysregulation of which has also been implicated in the development of diabetes. METHODS To test this hypothesis, we used a previously established embryonic stem cell (ESC) model of differentiation that mimics the diabetic environment of the developing embryo. We differentiated murine ESCs within osteogenic-inducing media containing either high (diabetic) or low (physiological) levels of D-glucose and performed time course analyses to study the influence of high glucose on early and late bone cell differentiation. RESULTS Endpoint measures for osteogenic differentiation were reduced in a glucose-dependent manner and expression of precursor-specific markers altered at multiple time points. Furthermore, transcriptional activity of the lymphoid enhancer factor (LEF)/T cell factor (TCF) transcription factors during precursor formation stages was significantly elevated while levels of CTNNB1 complexed with Forkhead box O 3a (FOXO3a) declined. Modulation of AKT, a known upstream regulator of both LEF/TCF and FOXO3a, as well as CTNNB1 rescued some of the reductions in osteogenic output seen in the high glucose condition. CONCLUSIONS Within our in vitro model, we found a clear involvement of LEF/TCF and FOXO3a signaling pathways in the regulation of osteogenic differentiation, which may account for the skeletal deficiencies found in newborns of diabetic mothers.
Collapse
Affiliation(s)
- Anke Dienelt
- Department of Cell Therapy, Applied Stem Cell Technologies Unit, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Kevin C. Keller
- Department of Molecular, Cell and Systems Biology & Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| | - Nicole I. zur Nieden
- Department of Cell Therapy, Applied Stem Cell Technologies Unit, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Department of Molecular, Cell and Systems Biology & Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
33
|
Aru B, Gürel G, Yanikkaya Demirel G. Mesenchymal Stem Cells: History, Characteristics and an Overview of Their Therapeutic Administration. TURKISH JOURNAL OF IMMUNOLOGY 2022. [DOI: 10.4274/tji.galenos.2022.18209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
34
|
Xue X, Li X, Yao J, Zhang X, Ren X, Xu S. Transient and Prolonged Activation of Wnt Signaling Contribute Oppositely to the Pathogenesis of Asherman's Syndrome. Int J Mol Sci 2022; 23:ijms23158808. [PMID: 35955940 PMCID: PMC9368949 DOI: 10.3390/ijms23158808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 11/23/2022] Open
Abstract
Asherman’s Syndrome (AS) is caused by dysfunction of endometrial regenerative ability, which is controlled by adult stem cells and their niche. The Wnt signaling pathway has been demonstrated to be implicated in this process. This study aimed to clarify the relationship between the Wnt signaling pathway and the progression of AS after initial endometrial damage. Endometria with and without adhesion as well as from the intrauterine devices three months after the surgery were collected to compare the area of fibrosis. The area% of fibrosis did not vary significantly. Significantly higher expression of non-phosphorylated β-catenin, Wnt5a and Wnt7a was identified in the endometria with adhesion. The CD140b+CD146+ endometrial stem-like cells were present in the endometria with adhesion. Both Wnt5a and Wnt7a promoted stem cell proliferation. However, only Wnt7a preserved stem cell population by stimulating self-renewal. A rat endometrial injury model was established to investigate the effect of the activated Wnt/β-catenin signaling pathway on endometrial healing. We found that a transient activation of the Wnt/β-catenin signaling pathway promoted angiogenesis and increased the number of glands. In conclusion, transient activation of the Wnt/β-catenin signaling pathway during the acute endometrial damage may help the tissue regeneration, while prolonged activation may correlate to fibrosis formation.
Collapse
Affiliation(s)
- Xiang Xue
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Xiaoli Li
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Jinmeng Yao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Xue Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Xu Ren
- Core Research Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Shan Xu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Correspondence:
| |
Collapse
|
35
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
36
|
Endometrial stem/progenitor cells: Properties, origins, and functions. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
37
|
Liang C, Liang Q, Xu X, Liu X, Gao X, Li M, Yang J, Xing X, Huang H, Tang Q, Liao L, Tian W. Bone morphogenetic protein 7 mediates stem cells migration and angiogenesis: therapeutic potential for endogenous pulp regeneration. Int J Oral Sci 2022; 14:38. [PMID: 35858911 PMCID: PMC9300630 DOI: 10.1038/s41368-022-00188-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 02/05/2023] Open
Abstract
Pulp loss is accompanied by the functional impairment of defense, sensory, and nutrition supply. The approach based on endogenous stem cells is a potential strategy for pulp regeneration. However, endogenous stem cell sources, exogenous regenerative signals, and neovascularization are major difficulties for pulp regeneration based on endogenous stem cells. Therefore, the purpose of our research is to seek an effective cytokines delivery strategy and bioactive materials to reestablish an ideal regenerative microenvironment for pulp regeneration. In in vitro study, we investigated the effects of Wnt3a, transforming growth factor-beta 1, and bone morphogenetic protein 7 (BMP7) on human dental pulp stem cells (h-DPSCs) and human umbilical vein endothelial cells. 2D and 3D culture systems based on collagen gel, matrigel, and gelatin methacryloyl were fabricated to evaluate the morphology and viability of h-DPSCs. In in vivo study, an ectopic nude mouse model and an in situ beagle dog model were established to investigate the possibility of pulp regeneration by implanting collagen gel loading BMP7. We concluded that BMP7 promoted the migration and odontogenic differentiation of h-DPSCs and vessel formation. Collagen gel maintained the cell adhesion, cell spreading, and cell viability of h-DPSCs in 2D or 3D culture. The transplantation of collagen gel loading BMP7 induced vascularized pulp-like tissue regeneration in vivo. The injectable approach based on collagen gel loading BMP7 might exert promising therapeutic application in endogenous pulp regeneration. BMP7 as a regenerative signaling molecule mediates stem cell migration and odontoblastic differentiation (a) and as a pro-angiogenic factor promotes revascularization of endothelial cells (b). Collagen gel supports cell adhesion, spreading, and viability (c). ![]()
Collapse
Affiliation(s)
- Cheng Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qingqing Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xun Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Gao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Maojiao Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaotao Xing
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haisen Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
38
|
Jing Z, Liang Z, Yang L, Du W, Yu T, Tang H, Li C, Wei W. Bone formation and bone repair: The roles and crosstalk of osteoinductive signaling pathways. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Zhang Y, Ling L, Ajay D/O Ajayakumar A, Eio YM, van Wijnen AJ, Nurcombe V, Cool SM. FGFR2 accommodates osteogenic cell fate determination in human mesenchymal stem cells. Gene 2022; 818:146199. [PMID: 35093449 PMCID: PMC9256080 DOI: 10.1016/j.gene.2022.146199] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 12/09/2021] [Accepted: 01/13/2022] [Indexed: 01/14/2023]
Abstract
The multilineage differentiation potential of human mesenchymal stem cells (hMSCs) underpins their clinical utility for tissue regeneration. Control of such cell-fate decisions is tightly regulated by different growth factors/cytokines and their cognate receptors. Fibroblast growth factors (FGFs) are among such factors critical for osteogenesis. However, how FGF receptors (FGFRs) help to orchestrate osteogenic progression remains to be fully elucidated. Here, we studied the protein levels of FGFRs during osteogenesis in human adult bone marrow-derived MSCs and discovered a positive correlation between FGFR2 expression and alkaline phosphatase (ALP) activity, an early marker of osteogenesis. Through RNA interference studies, we confirmed the role of FGFR2 in promoting the osteogenic differentiation of hMSCs. Knockdown of FGFR2 resulted in downregulation of pro-osteogenic genes and upregulation of pro-adipogenic genes and adipogenic commitment. Moreover, under osteogenic induction, FGFR2 knockdown resulted in upregulation of Enhancer of Zeste Homolog 2 (EZH2), an epigenetic enzyme that regulates MSC lineage commitment and suppresses osteogenesis. Lastly, we show that serial-passaged hMSCs have reduced FGFR2 expression and impaired osteogenic potential. Our study suggests that FGFR2 is critical for mediating osteogenic fate by regulating the balance of osteo-adipogenic lineage commitment. Therefore, examining FGFR2 levels during serial-passaging of hMSCs may prove useful for monitoring their multipotency.
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Ling Ling
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Arya Ajay D/O Ajayakumar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Yating Michelle Eio
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, 636921, Singapore
| | - Simon M Cool
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore.
| |
Collapse
|
40
|
Lei F, Li M, Lin T, Zhou H, Wang F, Su X. Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater 2022; 141:333-343. [PMID: 34979326 DOI: 10.1016/j.actbio.2021.12.035] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/08/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023]
Abstract
Periodontitis is the primary cause of tooth loss, but there is no effective treatment to repair inflammatory bone loss in periodontitis. Exosomes emerge as essential paracrine factors of mesenchymal stem cells (MSCs) that mediated tissue regeneration. Here, we investigated the potential of exosomes secreted by periodontal ligament stem cells (PDLSCs) as therapeutics for the bone defect in periodontitis. Exosomes secreted from PDLSCs derived from healthy periodontal ligaments (h-PDLSCs) and their function were evaluated on PDLSCs isolated from the inflammatory periodontal ligament of periodontitis patients (i-PDLSCs). Treatment of exosomes of h-PDLSCs led to an increase in the formation of mineralized nodules and the expressions of osteogenic genes and proteins in i-PDLSCs. Mechanistically, h-PDLSCs-exosomes suppressed the over-activation of canonical Wnt signaling to recover the osteogenic differentiation capacity of i-PDLSCs. To evaluate the therapeutic of exosomes on inflammatory bone loss, h-PDLSCs-exosomes loaded with Matrigel or β-TCP were employed to repair bone defects in rat models of periodontitis. Compared to the vehicle-treated control group, h-PDLSCs-exosomes-treated rats resulted in more bone formation in the defect of alveolar bone. In conclusion, these results demonstrated that exosomes derived from healthy PDLSCs could rescue the osteogenesis capacity of endogenous stem cells under an inflammatory environment and promote regeneration of alveolar bone. Our findings suggest that MSCs-derived exosome is an effective and practical cell-free MSC therapeutic for the treatment of periodontitis. STATEMENT OF SIGNIFICANCE: There is no effective treatment to repair inflammatory bone loss in periodontitis. As essential paracrine factors of PDLSCs, exosomes might mediate tissue regeneration during stem cell therapy. Here, we reported that exosomes secreted from healthy PDLSCs promoted the osteogenic differentiation of PDLSCs derived from periodontitis tissue. Healthy PDLSCs-exosomes treatment resulted in accelerated bone formation in the defect of alveolar bone in rat models of periodontitis. Mechanistically, h-PDLSCs-exosomes suppressed the over-activation of canonical Wnt signaling to recover the osteogenic differentiation capacity of inflammatory PDLSCs. These findings suggest that MSCs-derived exosome is an effective and practical cell-free MSC therapeutic for the treatment of periodontitis.
Collapse
|
41
|
Feng Y, Luo J, Cheng J, Xu A, Qiu D, He S, Zheng D, Jia C, Zhang Q, Lin N. A Small-Molecule Cocktails-Based Strategy in Culture of Mesenchymal Stem Cells. Front Bioeng Biotechnol 2022; 10:819148. [PMID: 35360405 PMCID: PMC8963903 DOI: 10.3389/fbioe.2022.819148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/11/2022] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a variety of unique properties, such as stem cell multipotency and immune regulation, making them attractive for use in cell therapy. Before infusion therapy, MSCs are required to undergo tissue separation, purification, and expansion in vitro for a certain duration. During the process of in vitro expansion of MSCs, the influence of culture time and environment can lead to cell senescence, increased heterogeneity, and function attenuation, which limits their clinical applications. We used a cocktail of three small-molecule compounds, ACY (A-83–01, CHIR99021, and Y-27632), to increase the proliferation activity of MSCs in vitro and reduce cell senescence. ACY inhibited the increase in heterogeneity of MSCs and conserved their differentiation potential. Additionally, ACY maintained the phenotype of MSCs and upregulated the expression of immunomodulatory factors. These results suggest that ACY can effectively improve the quantity and quality of MSCs.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jintao Cheng
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Aimin Xu
- The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Dongbo Qiu
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Sixiao He
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dayong Zheng
- The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Changchang Jia
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| | - Qi Zhang
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| |
Collapse
|
42
|
Bin-Bin Z, Da-Wa ZX, Chao L, Lan-Tao Z, Tao W, Chuan L, Chao-Zheng L, De-Chun L, Chang F, Shu-Qing W, Zu-Nan D, Xian-Wei P, Zhang ZX, Ke-Wen L. M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells. J Orthop Surg Res 2022; 17:137. [PMID: 35246197 PMCID: PMC8895825 DOI: 10.1186/s13018-022-03029-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/18/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells have always been a heated research topic in bone tissue regeneration and repair because of their self-renewal and multi-differentiation potential. A large number of studies have been focused on finding the inducing factors that will promote the osteogenic differentiation of bone marrow mesenchymal stem cells. Previous studies have shown that macrophage exosomes or miRNA-26a-5p can make it work, but the function of this kind of substance on cell osteogenic differentiation has not been public. METHODS M2 macrophages are obtained from IL-4 polarized bone marrow-derived macrophages. Exosomes were isolated from the supernatant of M2 macrophages and identified via transmission electron microscopy (TEM), western blotting, and DLS. Chondrogenic differentiation potential was detected by Alcian blue staining. Oil red O staining was used to detect the potential for lipogenic differentiation. And MTT would detect the proliferative capacity of cells. Western blot was performed to detect differential expression of osteogenic differentiation-related proteins. RESULTS The results showed that M2 macrophage exosomes will promote bone differentiation and at the same time inhibit lipid differentiation. In addition, M2 macrophage-derived exosomes have the function of promoting the expression of SOX and Aggrecan suppressing the level of MMP13. The exosome inhibitor GW4689 suppresses miRNA-26a-5p in M2 macrophage exosomes, and the treated exosomes do not play an important role in promoting bone differentiation. Moreover, miRNA-26a-5p can enable to promote bone differentiation and inhibit lipid differentiation. miRNA-26a-5p can promote the expression of ALP (alkaline phosphatase), RUNX-2 (Runt-related transcription factor 2), OPN(osteopontin), and Col-2(collagen type II). Therefore, it is speculated that exosomal miRNA-26a-5p is indispensable in osteogenic differentiation. CONCLUSIONS The present study indicated that M2 macrophage exosomes carrying miRNA-26a-5p can induce osteogenic differentiation of bone marrow-derived stem cells to inhibit lipogenic differentiation, and miRNA-26a-5p will also promote the expression of osteogenic differentiation-related proteins ALP, RUNX-2, OPN, and Col-2.
Collapse
Affiliation(s)
- Zhang Bin-Bin
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zha Xi Da-Wa
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Li Chao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zhang Lan-Tao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Wu Tao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Lu Chuan
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Liu Chao-Zheng
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Li De-Chun
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Feng Chang
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Wei Shu-Qing
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Dong Zu-Nan
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Pei Xian-Wei
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zhi-Xia Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Out-Patient, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Li Ke-Wen
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China.
| |
Collapse
|
43
|
Calabrese EJ. Hormesis and dental apical papilla stem cells. Chem Biol Interact 2022; 357:109887. [DOI: 10.1016/j.cbi.2022.109887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/23/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
|
44
|
Zhu Y, Ye L, Cai X, Li Z, Fan Y, Yang F. Icariin-Loaded Hydrogel Regulates Bone Marrow Mesenchymal Stem Cell Chondrogenic Differentiation and Promotes Cartilage Repair in Osteoarthritis. Front Bioeng Biotechnol 2022; 10:755260. [PMID: 35223781 PMCID: PMC8864219 DOI: 10.3389/fbioe.2022.755260] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Intra-articular injection of mesenchymal stem cells is a potential therapeutic strategy for cartilage protection and symptom relief for osteoarthritis (OA). However, controlling chondrogenesis of the implanted cells in the articular cavity remains a challenge. In this study, hydrogels containing different concentrations of icariin were prepared by in situ crosslinking of hyaluronic acid and Poloxamer 407. This injectable and thermoresponsive hydrogel, as a 3D cell culture system, showed good biocompatibility with chondrocytes and bone marrow mesenchymal stem cells (BMSCs), as well as promoted proliferation and chondrogenesis of BMSCs through the Wnt/β-catenin signaling pathway. Intra-articular injection of this kind of BMSC-loaded composite hydrogel can significantly prevent cartilage destruction by inducing chondrogenic differentiation of BMSCs, and relieve pain through regulating the expression of inflammatory cytokines (e.g., IL-10 and MMP-13) in the OA model. Incorporating BMSCs into this novel icariin-loaded hydrogel indicates a more superior efficacy than the single BMSC injection, which suggests a great potential for its application in OA.
Collapse
Affiliation(s)
- Yuefeng Zhu
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Le Ye
- Department of Pain, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoxi Cai
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zuhao Li
- Department of Pain, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongqian Fan
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Fengjian Yang
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
45
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
46
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
47
|
Widholz B, Westhauser F. Biomaterials for angiogenesis applications in an orthopedic context. BIOMATERIALS FOR VASCULOGENESIS AND ANGIOGENESIS 2022:415-438. [DOI: 10.1016/b978-0-12-821867-9.00016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Persichetti A, Milanetti E, Palmisano B, di Filippo A, Spica E, Donsante S, Coletta I, Venti MDS, Ippolito E, Corsi A, Riminucci M, Raimondo D. Nanostring technology on Fibrous Dysplasia bone biopsies. A pilot study suggesting different histology-related molecular profiles. Bone Rep 2021; 16:101156. [PMID: 34950753 PMCID: PMC8671863 DOI: 10.1016/j.bonr.2021.101156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/02/2022] Open
Abstract
Identifying the molecular networks that underlie Fibrous Dysplasia (FD) is key to understand the pathogenesis of the disease, to refine current diagnostic approaches and to develop efficacious therapies. In this study, we used the NanoString nCounter Analysis System to investigate the gene signature of a series of nine Formalin Fixed Decalcified and Paraffin-Embedded (FFDPE) bone biopsies from seven FD patients. We analyzed the expression level of 770 genes. Unsupervised clustering analysis demonstrated partitioning into two clusters with distinct patterns of gene expression. Differentially expressed genes included growth factors, components of the Wnt signaling system, interleukins and some of their cognate receptors, ephrin ligands, matrix metalloproteinases, neurotrophins and genes encoding components of the cAMP-dependent protein kinase. Interestingly, two tissue samples obtained from the same skeletal site of one patient one year apart failed to segregate in the same cluster. Retrospective histological review of the samples revealed different microscopic aspects in the two groups. The results of our pilot study suggest that the genetic signature of FD is heterogeneous and varies according to the histology and, likely, to the age of the lesion. In addition, they show that the Nanostring technology is a valuable tool for molecular translational studies on archival FFDPE material in FD and other rare bone diseases. We used the NanoString technology to analyze Formalin Fixed Decalcified Paraffin Embedded (FFDPE) Fibrous Dysplasia samples. We show that Fibrous Dysplasia lesions may have different molecular profiles consistent with its histological heterogeneity. NanoString technology is a valuable tool for molecular studies on rare bone diseases by using FFDPE archival material.
Collapse
Affiliation(s)
- Agnese Persichetti
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Edoardo Milanetti
- Department of Physics, Piazzale Aldo Moro 5, 00185 Rome, Italy.,Center for Life Nano Science@Sapienza, Italian Institute of Technology, Viale Regina Elena 291, 00161 Rome, Italy
| | - Biagio Palmisano
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | | | - Emanuela Spica
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Ilenia Coletta
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | | | - Ernesto Ippolito
- Department of Orthopaedic Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| | - Domenico Raimondo
- Department of Molecular Medicine, Viale Regina Elena, 324, 00161 Rome, Italy
| |
Collapse
|
49
|
de Castro LF, Sworder BJ, Mui B, Futrega K, Berendsen A, Phillips MD, Burbach NJ, Cherman N, Kuznetsov S, Gabet Y, Holmbeck K, Robey PG. Secreted frizzled related-protein 2 (Sfrp2) deficiency decreases adult skeletal stem cell function in mice. Bone Res 2021; 9:49. [PMID: 34857734 PMCID: PMC8639730 DOI: 10.1038/s41413-021-00169-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/21/2021] [Accepted: 06/27/2021] [Indexed: 12/18/2022] Open
Abstract
In a previous transcriptomic study of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived "mesenchymal stem cells"), SFRP2 was highly over-represented in a subset of multipotent BMSCs (skeletal stem cells, SSCs), which recreate a bone/marrow organ in an in vivo ectopic bone formation assay. SFRPs modulate WNT signaling, which is essential to maintain skeletal homeostasis, but the specific role of SFRP2 in BMSCs/SSCs is unclear. Here, we evaluated Sfrp2 deficiency on BMSC/SSC function in models of skeletal organogenesis and regeneration. The skeleton of Sfrp2-deficient (KO) mice is overtly normal; but their BMSCs/SSCs exhibit reduced colony-forming efficiency, reflecting low SSC self-renewal/abundancy. Sfrp2 KO BMSCs/SSCs formed less trabecular bone than those from WT littermates in the ectopic bone formation assay. Moreover, regeneration of a cortical drilled hole defect was dramatically impaired in Sfrp2 KO mice. Sfrp2-deficient BMSCs/SSCs exhibited poor in vitro osteogenic differentiation as measured by Runx2 and Osterix expression and calcium accumulation. Interestingly, activation of the Wnt co-receptor, Lrp6, and expression of Wnt target genes, Axin2, C-myc and Cyclin D1, were reduced in Sfrp2-deficient BMSCs/SSCs. Addition of recombinant Sfrp2 restored most of these activities, suggesting that Sfrp2 acts as a Wnt agonist. We demonstrate that Sfrp2 plays a role in self-renewal of SSCs and in the recruitment and differentiation of adult SSCs during bone healing. SFRP2 is also a useful marker of BMSC/SSC multipotency, and a factor to potentially improve the quality of ex vivo expanded BMSC/SSC products.
Collapse
Affiliation(s)
- Luis Fernandez de Castro
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, USA.
| | - Brian J. Sworder
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA ,grid.189504.10000 0004 1936 7558Department of Molecular Medicine, Boston University, Boston, MA USA
| | - Byron Mui
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Kathryn Futrega
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Agnes Berendsen
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Matthew D. Phillips
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Nathan J. Burbach
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA ,grid.17635.360000000419368657School of Dentistry, University of Minnesota—Twin Cities, Minneapolis, MN USA
| | - Natasha Cherman
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Sergei Kuznetsov
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Yankel Gabet
- grid.12136.370000 0004 1937 0546Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Kenn Holmbeck
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Pamela G. Robey
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
50
|
Zhou Q, Ren X, Oberoi MK, Bedar M, Caprini RM, Dewey MJ, Kolliopoulos V, Yamaguchi DT, Harley BA, Lee JC. β-Catenin Limits Osteogenesis on Regenerative Materials in a Stiffness-Dependent Manner. Adv Healthc Mater 2021; 10:e2101467. [PMID: 34585526 PMCID: PMC8665088 DOI: 10.1002/adhm.202101467] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/22/2021] [Indexed: 12/30/2022]
Abstract
Targeted refinement of regenerative materials requires mechanistic understanding of cell-material interactions. The nanoparticulate mineralized collagen glycosaminoglycan (MC-GAG) scaffold is shown to promote skull regeneration in vivo without additive exogenous growth factors or progenitor cells, suggesting potential for clinical translation. This work evaluates modulation of MC-GAG stiffness on canonical Wnt (cWnt) signaling. Primary human bone marrow-derived mesenchymal stem cells (hMSCs) are differentiated on two MC-GAG scaffolds (noncrosslinked, NX-MC, 0.3 kPa vs conventionally crosslinked, MC, 3.9 kPa). hMSCs increase expression of activated β-catenin, the major cWnt intracellular mediator, and the mechanosensitive YAP protein with near complete subcellular colocalization on stiffer MC scaffolds. Overall Wnt pathway inhibition reduces activated β-catenin and osteogenic differentiation, while elevating BMP4 and phosphorylated Smad1/5 (p-Smad1/5) expression on MC, but not NX-MC. Unlike Wnt pathway downregulation, isolated canonical Wnt inhibition with β-catenin knockdown increases osteogenic differentiation and mineralization specifically on the stiffer MC. β-catenin knockdown also increases p-Smad1/5, Runx2, and BMP4 expression only on the stiffer MC material. Thus, while stiffness-induced activation of the Wnt and mechanotransduction pathways promotes osteogenesis on MC-GAG, activated β-catenin is a limiting agent and may serve as a useful target or readout for optimal modulation of stiffness in skeletal regenerative materials.
Collapse
Affiliation(s)
- Qi Zhou
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Xiaoyan Ren
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Michelle K. Oberoi
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Meiwand Bedar
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Rachel M. Caprini
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| | - Marley J. Dewey
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Vasiliki Kolliopoulos
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Dean T. Yamaguchi
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073
- UCLA Molecular Biology Institute, Los Angeles, CA 90095
| |
Collapse
|