1
|
Jurić I, Kelam N, Racetin A, Filipović N, Čarić D, Rošin M, Vukojević K. WNT Signaling Factors as Potential Synovial Inflammation Moderators in Patients with Hip Osteoarthritis. Biomedicines 2025; 13:995. [PMID: 40299569 PMCID: PMC12025112 DOI: 10.3390/biomedicines13040995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025] Open
Abstract
Background: The main feature of osteoarthritis (OA) is the deterioration of articular cartilage, but numerous studies have demonstrated the role of synovial inflammation in the early stages of the disease, leading to further progression of OA. The WNT signaling pathway is involved in numerous activities in joint tissue, but there is a lack of evidence considering the role of WNT in OA synovitis. Our research aims to investigate the expression of WNT Family Member 5A/B (WNT5A/B), β-catenin, acetyl-α-tubulin, Dishevelled-1 (DVL-1), and Inversin (INV) in the synovial membrane of osteoarthritis (OA) hips. Methods: The immunohistochemical expressions of the aforementioned proteins in the synovial membrane were analyzed and compared with samples of control group participants with fractured femoral necks. Results: The immunoexpression of acetyl-α-tubulin was significantly increased in the intima (p < 0.0001) and subintima (p < 0.0001) of the group with OA compared with the intima and subintima of the control group. At the same time, acetyl-α-tubulin was also more highly expressed in the intima of the OA group than in the subintima of the OA group (p < 0.05); we found the same expression pattern in the control group (p < 0.0001). The differential analysis of the GEO dataset did not show significant differences between the osteoarthritis (OA) and control groups in the expression of TUBA1A. β-catenin was significantly increased in the subintima (p < 0.01) of the group with OA compared to the subintima of the control group. WNT expression has significantly higher positivity in the subintima than in the intima, especially in the control group (p < 0.01). WNT5A and WNT5B were significantly down-regulated in OA compared to the control in the differential analysis of the GEO dataset. The expression of INV and DVL-1 in our study and the differential analysis of the GEO dataset did not differ significantly between the osteoarthritis (OA) and control groups. Conclusions: Based on our results, we suggest that acetyl-α-tubulin and β-catenin might be involved in synovial membrane inflammation in OA and serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Ivana Jurić
- Department of Emergency Medicine, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia;
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (N.F.)
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (N.F.)
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (N.F.)
| | - Davor Čarić
- Surgery Department, Orthopaedics and Traumatology Division, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (D.Č.); (M.R.)
| | - Matko Rošin
- Surgery Department, Orthopaedics and Traumatology Division, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (D.Č.); (M.R.)
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (N.F.)
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
- Mediterranean Institute for Life Sciences, University of Split, Meštrovićevo Šetalište 45, 21000 Split, Croatia
| |
Collapse
|
2
|
Villegas Villarroel M, Huber C, Baujat G, Bonnard A, Collet C, Cormier-Daire V. Loss-of-function of DDR1 is responsible for a chondrodysplasia with multiple dislocations. J Bone Miner Res 2025; 40:362-371. [PMID: 39714220 DOI: 10.1093/jbmr/zjae205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Chondrodysplasias with multiple dislocations are rare skeletal disorders characterized by hyperlaxity, joint dislocations, and growth retardation. Chondrodysplasias with multiple dislocations have been linked to pathogenic variants in genes encoding proteins involved in the proteoglycan (PG) biosynthesis. In this study, by exome sequencing analysis, we identified a homozygous nonsense variant (NM_001297654.2: c.1825C>T, p.Arg609*) in the discoidin domain receptor 1 (DDR1) gene in a patient presenting joint dislocations, hyperlaxity, and cerebellar hypoplasia. Functional studies revealed decreased PG production in patient fibroblasts. We further demonstrated that DDR1 inhibition impaired the Indian Hedgehog signaling pathway in chondrocytes, decreased differentiation and mineralization in osteoblasts, and disrupted p38 MAPK signaling in both cell types. Additionally, we showed that DDR1 inhibition affected the noncanonical WNT signaling pathway in human skeletal cells and decreased PG production in chondrocytes. These findings suggest that DDR1 is a new gene involved in the group of chondrodysplasias with multiple dislocations and highlights its essential role in human skeletal and brain development.
Collapse
Affiliation(s)
- Miriam Villegas Villarroel
- Reference Center for Skeletal Dysplasia, INSERM UMR 1163, Paris Cité University, Imagine Institute, Necker Enfants Malades Hospital (AP-HP), 75015 Paris, France
| | - Céline Huber
- Reference Center for Skeletal Dysplasia, INSERM UMR 1163, Paris Cité University, Imagine Institute, Necker Enfants Malades Hospital (AP-HP), 75015 Paris, France
| | - Geneviève Baujat
- Reference Center for Skeletal Dysplasia, INSERM UMR 1163, Paris Cité University, Imagine Institute, Necker Enfants Malades Hospital (AP-HP), 75015 Paris, France
| | - Adeline Bonnard
- Department of Molecular Genetics, Robert Debré Hospital (AP-HP), 75019 Paris, France
| | - Corinne Collet
- Reference Center for Skeletal Dysplasia, INSERM UMR 1163, Paris Cité University, Imagine Institute, Necker Enfants Malades Hospital (AP-HP), 75015 Paris, France
| | - Valérie Cormier-Daire
- Reference Center for Skeletal Dysplasia, INSERM UMR 1163, Paris Cité University, Imagine Institute, Necker Enfants Malades Hospital (AP-HP), 75015 Paris, France
| |
Collapse
|
3
|
Xiong F, Chevalier Y, Klar RM. Parallel Chondrogenesis and Osteogenesis Tissue Morphogenesis in Muscle Tissue via Combinations of TGF-β Supergene Family Members. Cartilage 2025; 16:71-88. [PMID: 37714817 PMCID: PMC11744598 DOI: 10.1177/19476035231196224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/20/2023] [Accepted: 08/04/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVE This study aimed to decipher the temporal and spatial signaling code for clinical cartilage and bone regeneration. We investigated the effects of continuous equal dosages of a single, dual, or triplicate growth factor combination of bone morphogenetic protein (BMP)-2, transforming growth factor (TGF)-β3, and/or BMP-7 on muscle tissue over a culturing period. The hypothesis was that specific growth factor combinations at specific time points direct tissue transformation toward endochondral bone or cartilage formation. DESIGN The harvested muscle tissues from F-344 adult male rats were cultured in 96-well plates maintained in a specific medium and cultured at specific conditions. And the multidimensional and multi-time point analyses were performed at both the genetic and protein levels. RESULTS The results insinuate that the application of growth factor stimulates a chaotic tissue response that does not follow a chronological signaling cascade. Both osteogenic and chondrogenic genes showed upregulation after induction, a similar result was also observed in the semiquantitative analysis after immunohistochemical staining against different antigens. CONCLUSIONS The study showed that multiple TGF-β superfamily proteins applied to tissue stimulate developmental tissue processes that do not follow current tissue formation rules. The findings contribute to the understanding of the chronological order of signals and expression patterns needed to achieve chondrogenesis, articular chondrogenesis, or osteogenesis, which is crucial for the development of treatments that can regrow bone and articular cartilage clinically.
Collapse
Affiliation(s)
- Fei Xiong
- Wuxi Hand Surgery Hospital, Wuxi, China
| | - Yan Chevalier
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital, LMU Munich, Germany
| | - Roland M. Klar
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, USA
| |
Collapse
|
4
|
Schmidt S, Klampfleuthner FAM, Renkawitz T, Diederichs S. Cause and chondroprotective effects of prostaglandin E2 secretion during mesenchymal stromal cell chondrogenesis. Eur J Cell Biol 2024; 103:151412. [PMID: 38608422 DOI: 10.1016/j.ejcb.2024.151412] [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: 02/16/2024] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) that are promising for cartilage tissue engineering secrete high amounts of prostaglandin E2 (PGE2), an immunoactive mediator involved in endochondral bone development. This study aimed to identify drivers of PGE2 and its role in the inadvertent MSC misdifferentiation into hypertrophic chondrocytes. PGE2 release, which rose in the first three weeks of MSC chondrogenesis, was jointly stimulated by endogenous BMP, WNT, and hedgehog activity that supported the exogenous stimulation by TGF-β1 and insulin to overcome the PGE2 inhibition by dexamethasone. Experiments with PGE2 treatment or the inhibitor celecoxib or specific receptor antagonists demonstrated that PGE2, although driven by prohypertrophic signals, exerted broad autocrine antihypertrophic effects. This chondroprotective effect makes PGE2 not only a promising option for future combinatorial approaches to direct MSC tissue engineering approaches into chondral instead of endochondral development but could potentially have implications for the use of COX-2-selective inhibitors in osteoarthritis pain management.
Collapse
Affiliation(s)
- Sven Schmidt
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg, Germany
| | - Felicia A M Klampfleuthner
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg, Germany
| | - Tobias Renkawitz
- Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Solvig Diederichs
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg, Germany.
| |
Collapse
|
5
|
Chang L, Chen Q, Wang B, Liu J, Zhang M, Zhu W, Jiang J. Single cell RNA analysis uncovers the cell differentiation and functionalization for air breathing of frog lung. Commun Biol 2024; 7:665. [PMID: 38816547 PMCID: PMC11139932 DOI: 10.1038/s42003-024-06369-1] [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: 10/11/2023] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
The evolution and development of vertebrate lungs have been widely studied due to their significance in terrestrial adaptation. Amphibians possess the most primitive lungs among tetrapods, underscoring their evolutionary importance in bridging the transition from aquatic to terrestrial life. However, the intricate process of cell differentiation during amphibian lung development remains poorly understood. Using single-cell RNA sequencing, we identify 13 cell types in the developing lungs of a land-dwelling frog (Microhyla fissipes). We elucidate the differentiation trajectories and mechanisms of mesenchymal cells, identifying five cell fates and their respective driver genes. Using temporal dynamics analyses, we reveal the gene expression switches of epithelial cells, which facilitate air breathing during metamorphosis. Furthermore, by integrating the published data from another amphibian and two terrestrial mammals, we illuminate both conserved and divergent cellular repertoires during the evolution of tetrapod lungs. These findings uncover the frog lung cell differentiation trajectories and functionalization for breathing in air and provide valuable insights into the cell-type evolution of vertebrate lungs.
Collapse
Affiliation(s)
- Liming Chang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiheng Chen
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bin Wang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
| | - Jiongyu Liu
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
| | - Meihua Zhang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
| | - Wei Zhu
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jianping Jiang
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Voga M. Modulation of Canine Adipose-Derived Mesenchymal Stem/Medicinal Signalling Cells with Ascorbic Acid: Effect on Proliferation and Chondrogenic Differentiation on Standard Plastic and Silk Fibroin Surfaces. Bioengineering (Basel) 2024; 11:513. [PMID: 38790380 PMCID: PMC11118827 DOI: 10.3390/bioengineering11050513] [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: 04/29/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Ascorbic acid (AA) plays a crucial role in both the proliferation and chondrogenic differentiation potential of mesenchymal stem/medicinal signalling cells (MSCs); these are both key aspects of their general therapeutic use and their increasing use in veterinary medicine. Current immunomodulatory therapies require efficient expansion of MSCs in the laboratory, while emerging tissue regeneration strategies, such as cartilage or bone repair, aim to use differentiated MSCs and modulate the expression of chondrogenic and hypertrophic markers. Our aim was to investigate whether the addition of AA to the growth medium enhances the proliferation of canine adipose-derived MSCs (cAMSCs) grown on standard plastic surfaces and whether it affects chondrogenic differentiation potential on silk fibroin (SF) films. We assessed cell viability with trypan blue and proliferation potential by calculating population doubling. Chondrogenic induction on SF films was assessed by Alcian blue staining and gene expression analysis of chondrogenic and hypertrophic genes. The results showed that growth medium with AA significantly enhanced the proliferation of cAMSCs without affecting cell viability and modulated the expression of chondrogenic and hypertrophic genes of cAMSCs grown on SF films. Our results suggest that AA may be used in growth medium for expansion of cAMSCs and, at the same time, provide the basis for future studies to investigate the role of AA and SF in chondrogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Metka Voga
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Diederichs S, Dreher SI, Nüesch SA, Schmidt S, Merle C, Richter W. Mesenchymal stromal cell chondrogenesis under ALK1/2/3-specific BMP inhibition: a revision of the prohypertrophic signalling network concept. Stem Cell Res Ther 2024; 15:98. [PMID: 38581019 PMCID: PMC10998299 DOI: 10.1186/s13287-024-03710-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/27/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND In vitro chondrogenesis of mesenchymal stromal cells (MSCs) driven by the essential chondro-inducer transforming growth factor (TGF)-β is instable and yields undesired hypertrophic cartilage predisposed to bone formation in vivo. TGF-β can non-canonically activate bone morphogenetic protein-associated ALK1/2/3 receptors. These have been accused of driving hypertrophic MSC misdifferentiation, but data remained conflicting. We here tested the antihypertrophic capacity of two highly specific ALK1/2/3 inhibitors - compound A (CompA) and LDN-212854 (LDN21) - in order to reveal potential prohypertrophic contributions of these BMP/non-canonical TGF-β receptors during MSC in vitro chondrogenesis. METHODS Standard chondrogenic pellet cultures of human bone marrow-derived MSCs were treated with TGF-β and CompA (500 nM) or LDN21 (500 nM). Daily 6-hour pulses of parathyroid hormone-related peptide (PTHrP[1-34], 2.5 nM, from day 7) served as potent antihypertrophic control treatment. Day 28 samples were subcutaneously implanted into immunodeficient mice. RESULTS All groups underwent strong chondrogenesis, but GAG/DNA deposition and ACAN expression were slightly but significantly reduced by ALK inhibition compared to solvent controls along with a mild decrease of the hypertrophy markers IHH-, SPP1-mRNA, and Alkaline phosphatase (ALP) activity. When corrected for the degree of chondrogenesis (COL2A1 expression), only pulsed PTHrP but not ALK1/2/3 inhibition qualified as antihypertrophic treatment. In vivo, all subcutaneous cartilaginous implants mineralized within 8 weeks, but PTHrP pretreated samples formed less bone and attracted significantly less haematopoietic marrow than ALK1/2/3 inhibitor groups. CONCLUSIONS Overall, our data show that BMP-ALK1/2/3 inhibition cannot program mesenchymal stromal cells toward stable chondrogenesis. BMP-ALK1/2/3 signalling is no driver of hypertrophic MSC misdifferentiation and BMP receptor induction is not an adverse prohypertrophic side effect of TGF-β that leads to endochondral MSC misdifferentiation. Instead, the prohypertrophic network comprises misregulated PTHrP/hedgehog signalling and WNT activity, and a potential contribution of TGF-β-ALK4/5-mediated SMAD1/5/9 signalling should be further investigated to decide about its postulated prohypertrophic activity. This will help to successfully engineer cartilage replacement tissues from MSCs in vitro and translate these into clinical cartilage regenerative therapies.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany.
| | - Simon I Dreher
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| | - Sarah Anna Nüesch
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| | - Sven Schmidt
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| | - Christian Merle
- Orthopaedic University Hospital, Heidelberg University Hospital, Heidelberg, Germany
- Orthopädische Klinik Paulinenhilfe, Diakonieklinikum Stuttgart, Stuttgart, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| |
Collapse
|
8
|
Hasani-Sadrabadi MM, Yuan W, Ferreira LDAQ, Liu Z, Shen J, Sarrión P, Sharifi F, Malek-Khatabi A, Dashtimoghadam E, Yu B, Ansari S, Moshaverinia A. Precise Engineering of Growth Factor Presentation Using Extracellular Microenvironment-Mimicking Microfluidic Microparticles. ACS Biomater Sci Eng 2024; 10:1686-1696. [PMID: 38347681 DOI: 10.1021/acsbiomaterials.3c01922] [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: 03/12/2024]
Abstract
One of the main challenges in tissue engineering is finding a way to deliver specific growth factors (GFs) with precise spatiotemporal control over their presentation. Here, we report a novel strategy for generating microscale carriers with enhanced affinity for high content loading suitable for the sustained and localized delivery of GFs. Our developed microparticles can be injected locally and sustainably release encapsulated growth factors for up to 28 days. Fine-tuning of particles' size, affinity, microstructures, and release kinetics is achieved using a microfluidic system along with bioconjugation techniques. We also describe an innovative 3D micromixer platform to control the formation of core-shell particles based on superaffinity using a polymer-peptide conjugate for further tuning of release kinetics and delayed degradation. Chitosan shells block the burst release of encapsulated GFs and enable their sustained delivery for up to 10 days. The matched release profiles and degradation provide the local tissues with biomimetic, developmental-biologic-compatible signals to maximize regenerative effects. The versatility of this approach is verified using three different therapeutic proteins, including human bone morphogenetic protein-2 (rhBMP-2), vascular endothelial growth factor (VEGF), and stromal cell-derived factor 1 (SDF-1α). As in vivo morphogenesis is typically driven by the combined action of several growth factors, the proposed technique can be developed to generate a library of GF-loaded particles with designated release profiles.
Collapse
Affiliation(s)
- Mohammad Mahdi Hasani-Sadrabadi
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, California 90095, United States
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, California 90095, United States
| | - Weihao Yuan
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Luiza de Almeida Queiroz Ferreira
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, California 90095, United States
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270, Brazil
| | - Zeyang Liu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, California 90095, United States
| | - Jun Shen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Patricia Sarrión
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Fatemeh Sharifi
- Department of Chemical Engineering, Sharif University of Technology, Tehran 11365, Iran
| | - Atefeh Malek-Khatabi
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran
| | - Erfan Dashtimoghadam
- Department of Chemistry and Physics, Troy University, Troy, Alabama 36082, United States
- Center for Materials and Manufacturing Sciences, Troy University, Troy, Alabama 36082, United States
| | - Bo Yu
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Section of Prosthodontics, School of Dentistry, University of California, Los Angeles, California 90095, United States
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
9
|
Velot É, Balmayor ER, Bertoni L, Chubinskaya S, Cicuttini F, de Girolamo L, Demoor M, Grigolo B, Jones E, Kon E, Lisignoli G, Murphy M, Noël D, Vinatier C, van Osch GJVM, Cucchiarini M. Women's contribution to stem cell research for osteoarthritis: an opinion paper. Front Cell Dev Biol 2023; 11:1209047. [PMID: 38174070 PMCID: PMC10762903 DOI: 10.3389/fcell.2023.1209047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/18/2023] [Indexed: 01/05/2024] Open
Affiliation(s)
- Émilie Velot
- Laboratory of Molecular Engineering and Articular Physiopathology (IMoPA), French National Centre for Scientific Research, University of Lorraine, Nancy, France
| | - Elizabeth R. Balmayor
- Experimental Orthopaedics and Trauma Surgery, Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN, United States
| | - Lélia Bertoni
- CIRALE, USC 957, BPLC, École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | - Flavia Cicuttini
- Musculoskeletal Unit, Monash University and Rheumatology, Alfred Hospital, Melbourne, VIC, Australia
| | - Laura de Girolamo
- IRCCS Ospedale Galeazzi - Sant'Ambrogio, Orthopaedic Biotechnology Laboratory, Milan, Italy
| | - Magali Demoor
- Normandie University, UNICAEN, BIOTARGEN, Caen, France
| | - Brunella Grigolo
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio RAMSES, Bologna, Italy
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom
| | - Elizaveta Kon
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department ofBiomedical Sciences, Humanitas University, Milan, Italy
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Bologna, Italy
| | - Mary Murphy
- Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Danièle Noël
- IRMB, University of Montpellier, Inserm, CHU Montpellier, Montpellier, France
| | - Claire Vinatier
- Nantes Université, Oniris, INSERM, Regenerative Medicine and Skeleton, Nantes, France
| | - Gerjo J. V. M. van Osch
- Department of Orthopaedics and Sports Medicine and Department of Otorhinolaryngology, Department of Biomechanical Engineering, University Medical Center Rotterdam, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, Netherlands
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
10
|
Carrasco ME, Thaler R, Nardocci G, Dudakovic A, van Wijnen AJ. Inhibition of Ezh2 redistributes bivalent domains within transcriptional regulators associated with WNT and Hedgehog pathways in osteoblasts. J Biol Chem 2023; 299:105155. [PMID: 37572850 PMCID: PMC10506106 DOI: 10.1016/j.jbc.2023.105155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/19/2023] [Accepted: 08/03/2023] [Indexed: 08/14/2023] Open
Abstract
Bivalent epigenomic regulatory domains containing both activating histone 3 lysine 4 (H3K4me3) and repressive lysine 27 (H3K27me3) trimethylation are associated with key developmental genes. These bivalent domains repress transcription in the absence of differentiation signals but maintain regulatory genes in a poised state to allow for timely activation. Previous studies demonstrated that enhancer of zeste homolog 2 (Ezh2), a histone 3 lysine 27 (H3K27) methyltransferase, suppresses osteogenic differentiation and that inhibition of Ezh2 enhances commitment of osteoblast progenitors in vitro and bone formation in vivo. Here, we examined the mechanistic effects of Tazemetostat (EPZ6438), an Food and Drug Administration approved Ezh2 inhibitor for epithelioid sarcoma treatment, because this drug could potentially be repurposed to stimulate osteogenesis for clinical indications. We find that Tazemetostat reduces H3K27me3 marks in bivalent domains in enhancers required for bone formation and stimulates maturation of MC3T3 preosteoblasts. Furthermore, Tazemetostat activates bivalent genes associated with the Wingless/integrated (WNT), adenylyl cyclase (cAMP), and Hedgehog (Hh) signaling pathways based on transcriptomic (RNA-seq) and epigenomic (chromatin immunoprecipitation [ChIP]-seq) data. Functional analyses using selective pathway inhibitors and silencing RNAs demonstrate that the WNT and Hh pathways modulate osteogenic differentiation after Ezh2 inhibition. Strikingly, we show that loss of the Hh-responsive transcriptional regulator Gli1, but not Gli2, synergizes with Tazemetostat to accelerate osteoblast differentiation. These studies establish epigenetic cooperativity of Ezh2, Hh-Gli1 signaling, and bivalent regulatory genes in suppressing osteogenesis. Our findings may have important translational ramifications for anabolic applications requiring bone mass accrual and/or reversal of bone loss.
Collapse
Affiliation(s)
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Gino Nardocci
- Program in Molecular Biology and Bioinformatics, Faculty of Medicine, Center for Biomedical Research and Innovation (CIIB), Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA.
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, Vermont, USA.
| |
Collapse
|
11
|
Hammersen T, Buchert J, Zietzschmann S, Diederichs S, Richter W. Inverse Regulation of Cartilage Neogenesis at Physiologically Relevant Calcium Conditions by Human Articular Chondrocytes and Mesenchymal Stromal Cells. Cells 2023; 12:1659. [PMID: 37371129 DOI: 10.3390/cells12121659] [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: 05/19/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Elaborate bioreactor cultivation or expensive growth factor supplementation can enhance extracellular matrix production in engineered neocartilage to provide sufficient mechanical resistance. We here investigated whether raising extracellular calcium levels in chondrogenic cultures to physiologically relevant levels would provide a simple and inexpensive alternative to enhance cartilage neogenesis from human articular chondrocytes (AC) or bone marrow-derived mesenchymal stromal cells (BMSC). Interestingly, AC and BMSC-derived chondrocytes showed an opposite response to a calcium increase from 1.8 mM to 8 mM by which glycosaminoglycan (GAG) and collagen type II production were elevated during BMSC chondrogenesis but depressed in AC, leading to two-fold higher GAG/DNA values in BMSC-based neocartilage compared to the AC group. According to control treatments with Mg2+ or sucrose, these effects were specific for CaCl2 rather than divalent cations or osmolarity. Importantly, undesired pro-hypertrophic traits were not stimulated by calcium treatment. Specific induction of PTHrP mRNA and protein by 8.0mM calcium only in AC, along with negative effects of recombinant PTHrP1-34 on cartilage matrix production, suggested that the PTHrP pathway contributed to the detrimental effects in AC-based neocartilage. Altogether, raising extracellular calcium levels was discovered as a novel, simple and inexpensive stimulator for BMSC-based cartilage neogenesis without the need for special bioreactors, whereas such conditions should be avoided for AC.
Collapse
Affiliation(s)
- Tim Hammersen
- Research Center for Experimental Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, 69118 Heidelberg, Germany
| | - Justyna Buchert
- Research Center for Experimental Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, 69118 Heidelberg, Germany
| | - Severin Zietzschmann
- Orthopaedic Hospital, Department of Orthopaedics, Heidelberg University Hospital, 69118 Heidelberg, Germany
| | - Solvig Diederichs
- Research Center for Experimental Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, 69118 Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, 69118 Heidelberg, Germany
| |
Collapse
|
12
|
Sountoulidis A, Marco Salas S, Braun E, Avenel C, Bergenstråhle J, Theelke J, Vicari M, Czarnewski P, Liontos A, Abalo X, Andrusivová Ž, Mirzazadeh R, Asp M, Li X, Hu L, Sariyar S, Martinez Casals A, Ayoglu B, Firsova A, Michaëlsson J, Lundberg E, Wählby C, Sundström E, Linnarsson S, Lundeberg J, Nilsson M, Samakovlis C. A topographic atlas defines developmental origins of cell heterogeneity in the human embryonic lung. Nat Cell Biol 2023; 25:351-365. [PMID: 36646791 PMCID: PMC9928586 DOI: 10.1038/s41556-022-01064-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/23/2022] [Indexed: 01/18/2023]
Abstract
The lung contains numerous specialized cell types with distinct roles in tissue function and integrity. To clarify the origins and mechanisms generating cell heterogeneity, we created a comprehensive topographic atlas of early human lung development. Here we report 83 cell states and several spatially resolved developmental trajectories and predict cell interactions within defined tissue niches. We integrated single-cell RNA sequencing and spatially resolved transcriptomics into a web-based, open platform for interactive exploration. We show distinct gene expression programmes, accompanying sequential events of cell differentiation and maturation of the secretory and neuroendocrine cell types in proximal epithelium. We define the origin of airway fibroblasts associated with airway smooth muscle in bronchovascular bundles and describe a trajectory of Schwann cell progenitors to intrinsic parasympathetic neurons controlling bronchoconstriction. Our atlas provides a rich resource for further research and a reference for defining deviations from homeostatic and repair mechanisms leading to pulmonary diseases.
Collapse
Affiliation(s)
- Alexandros Sountoulidis
- Science for Life Laboratory, Solna, Sweden
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sergio Marco Salas
- Science for Life Laboratory, Solna, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Emelie Braun
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Christophe Avenel
- Department of Information Technology, Uppsala University, Uppsala, Sweden
- BioImage Informatics Facility, Science for Life Laboratory, SciLifeLab, Sweden
| | - Joseph Bergenstråhle
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jonas Theelke
- Science for Life Laboratory, Solna, Sweden
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marco Vicari
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Paulo Czarnewski
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Andreas Liontos
- Science for Life Laboratory, Solna, Sweden
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Xesus Abalo
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Žaneta Andrusivová
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Reza Mirzazadeh
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Michaela Asp
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lijuan Hu
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Sanem Sariyar
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Anna Martinez Casals
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Burcu Ayoglu
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Alexandra Firsova
- Science for Life Laboratory, Solna, Sweden
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Emma Lundberg
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Carolina Wählby
- Department of Information Technology, Uppsala University, Uppsala, Sweden
- BioImage Informatics Facility, Science for Life Laboratory, SciLifeLab, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Sten Linnarsson
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Solna, Sweden.
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| | - Christos Samakovlis
- Science for Life Laboratory, Solna, Sweden.
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
- Molecular Pneumology, Cardiopulmonary Institute, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
13
|
Liang T, Li P, Liang A, Zhu Y, Qiu X, Qiu J, Peng Y, Huang D, Gao W, Gao B. Identifying the key genes regulating mesenchymal stem cells chondrogenic differentiation: an in vitro study. BMC Musculoskelet Disord 2022; 23:985. [PMCID: PMC9664719 DOI: 10.1186/s12891-022-05958-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Mesenchymal stem cells (MSCs) possess the potential to differentiate into chondrocytes, which makes them an ideal source for healing cartilage defects. Here, we seek to identify the essential genes participating in MSCs chondrogenesis.
Methods
Human MSCs were induced for chondrogenesis for 7, 14, and 21 days using a high-density micromass culture system, and RNA was extracted for RNA-seq.
Results
A total of 6247 differentially expressed genes (DEGs) were identified on day 7, and 85 DEGs were identified on day 14. However, no significant DEGs was identified on day 21. The top 30 DEGs at day 7, including COL9A3, COL10A1, and CILP2, are closely related to extracellular matrix organization. While the top 30 DEGs at day 14 revealed that inflammation-related genes were enriched, including CXCL8, TLR2, and CCL20. We also conducted protein–protein interaction (PPI) networks analysis using the search tool for the retrieval of interacting genes (STRING) database and identified key hub genes, including CXCL8, TLR2, CCL20, and MMP3. The transcriptional factors were also analyzed, identifying the top 5 TFs: LEF1, FOXO1, RORA, BHLHE41, and SOX5. We demonstrated one particular TF, RORA, in promoting early MSCs chondrogenesis.
Conclusions
Taken together, our results suggested that these DEGs may have a complex effect on MSCs chondrogenesis both synergistically and solitarily.
Collapse
|
14
|
Stage-Dependent Activity and Pro-Chondrogenic Function of PI3K/AKT during Cartilage Neogenesis from Mesenchymal Stromal Cells. Cells 2022; 11:cells11192965. [PMID: 36230927 PMCID: PMC9563299 DOI: 10.3390/cells11192965] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Differentiating mesenchymal stromal cells (MSCs) into articular chondrocytes (ACs) for application in clinical cartilage regeneration requires a profound understanding of signaling pathways regulating stem cell chondrogenesis and hypertrophic degeneration. Classifying endochondral signals into drivers of chondrogenic speed versus hypertrophy, we here focused on insulin/insulin-like growth factor 1 (IGF1)-induced phosphoinositide 3-kinase (PI3K)/AKT signaling. Aware of its proliferative function during early but not late MSC chondrogenesis, we aimed to unravel the late pro-chondrogenic versus pro-hypertrophic PI3K/AKT role. PI3K/AKT activity in human MSC and AC chondrogenic 3D cultures was assessed via Western blot detection of phosphorylated AKT. The effects of PI3K inhibition with LY294002 on chondrogenesis and hypertrophy were assessed via histology, qPCR, the quantification of proteoglycans, and alkaline phosphatase activity. Being repressed by ACs, PI3K/AKT activity transiently rose in differentiating MSCs independent of TGFβ or endogenous BMP/WNT activity and climaxed around day 21. PI3K/AKT inhibition from day 21 on equally reduced chondrocyte and hypertrophy markers. Proving important for TGFβ-induced SMAD2 phosphorylation and SOX9 accumulation, PI3K/AKT activity was here identified as a required stage-dependent driver of chondrogenic speed but not of hypertrophy. Thus, future attempts to improve MSC chondrogenesis will depend on the adequate stimulation and upregulation of PI3K/AKT activity to generate high-quality cartilage from human MSCs.
Collapse
|
15
|
The effect of matrix stiffness on the chondrogenic differentiation of mesenchymal stem cells. J Mol Histol 2022; 53:805-816. [PMID: 36029427 DOI: 10.1007/s10735-022-10094-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 07/24/2022] [Indexed: 10/15/2022]
Abstract
Articular cartilage is one of the most important weight-bearing components in human body, thus the chondrogenesis of stem cells is reactive to many intracellular and extracellular mechanical signals. As a unique physical cue, matrix stiffness plays an integral role in commitment of stem cell fate. However, when examining the downstream effects of matrix stiffness, most studies used different soluble factors to assist physical inducing process, which may mask the chondrogenic effects of matrix stiffness. Here we fabricated polyacrylamide (PAAm) hydrogels with gradient stiffness to unravel the role of matrix stiffness in chondrogenic process of mesenchymal stem cells (MSCs), with or without TGF-β3 as induction factor. The results showed that with micromass culture mimicking relatively high cell density in vivo, the chondrogenic differentiation of MSCs can be promoted by soft substrates (about 0.5 kPa) independently with assembled cytoskeleton. Further analysis indicated that addition of TGF-β3 generally increased expression level of cartilage-related markers and masked the stiffness-derived expression pattern of hypertrophic markers. These results demonstrate how mechanical cues experienced in developmental context regulate commitment of stem cell fate and have significant impact on the design of tissue regeneration materials.
Collapse
|
16
|
Cai S, Zou Y, Zhao Y, Lin H, Zheng D, Xu L, Lu Y. Mechanical stress reduces secreted frizzled-related protein expression and promotes temporomandibular joint osteoarthritis via Wnt/β-catenin signaling. Bone 2022; 161:116445. [PMID: 35589066 DOI: 10.1016/j.bone.2022.116445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/24/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022]
Abstract
AIMS Mechanical stress overload in the temporomandibular joint (TMJ) is an important cause of TMJ osteoarthritis (TMJOA). Whether secreted frizzled-related proteins (SFRPs) play important roles in the development of mechanical stress-induced TMJOA remains controversial. In this study, we investigated the roles of the Wnt/β-catenin signaling and SFRPs in the progression of mechanical stress-induced TMJOA. METHODS We investigated the progression of mechanical stress-induced TMJOA using an in vivo model via modified increased occlusal vertical dimension (iOVD) malocclusion and an in vitro model in which isolated chondrocytes were subjected to mechanical stress. The effects of inhibition of Wnt/β-catenin signal on TMJOA induced by mechanical stress were studied by in vitro drug added and in vivo intra-articular injection of XAV-939. TMJOA progression, Wnt/β-catenin signaling and SFRPs was assessed by Cone beam computed tomography (CBCT) analysis, histochemical and immunohistochemical (IHC) staining, quantitative real-time PCR (qRT-PCR), Western blotting (WB), and immunofluorescence (IF) staining. RESULTS Our in vivo results showed that iOVD-induced mechanical stress in the TMJ disrupted mandible growth, induced OA-like changes in TMJ cartilage, and increased OA-related cytokine expression. In addition, iOVD activated Wnt/β-catenin signaling and suppressed Sfrp1, Sfrp3, and Sfrp4 expression in condylar cartilage. Moreover, our in vitro study showed that stress disrupted homeostasis, activated Wnt/β-catenin signaling and inhibited SFRP3 and SFRP4 expression in chondrocytes. Suppression of Wnt/β-catenin signaling with XAV-939 promoted SFRP3 and SFRP4 expression and rescued mechanical stress-induced cartilage degeneration in vivo and in vitro. CONCLUSIONS Our work suggests that mechanical stress reduces SFRPs expression both in vivo and in vitro and promotes TMJOA via Wnt/β-catenin signaling. Suppression of Wnt/β-catenin signaling promotes SFRPs expression, especially SFRP3 and SFRP4 expression, and rescues mechanical stress-induced cartilage degeneration. Wnt/β-catenin signaling and SFRPs may represent potential therapeutic targets for TMJOA.
Collapse
Affiliation(s)
- Senxin Cai
- Department of Orthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yuchun Zou
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yong Zhao
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China; Department of Pathology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Hanyu Lin
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Linyu Xu
- Department of Orthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| | - Youguang Lu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
17
|
Kim JG, Rim YA, Ju JH. The Role of Transforming Growth Factor Beta in Joint Homeostasis and Cartilage Regeneration. Tissue Eng Part C Methods 2022; 28:570-587. [PMID: 35331016 DOI: 10.1089/ten.tec.2022.0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) is an important regulator of joint homeostasis, of which dysregulation is closely associated with the development of osteoarthritis (OA). In normal conditions, its biological functions in a joint environment are joint protective, but it can be dramatically altered in different contexts, making its therapeutic application a challenge. However, with the deeper insights into the TGF-β functions, it has been proven that TGF-β augments cartilage regeneration by chondrocytes, and differentiates both the precursor cells of chondrocytes and stem cells into cartilage-generating chondrocytes. Following documentation of the therapeutic efficacy of chondrocytes augmented by TGF-β in the last decade, there is an ongoing phase III clinical trial examining the therapeutic efficacy of a mixture of allogeneic chondrocytes and TGF-β-overexpressing cells. To prepare cartilage-restoring chondrocytes from induced pluripotent stem cells (iPSCs), the stem cells are differentiated mainly using TGF-β with some other growth factors. Of note, clinical trials evaluating the therapeutic efficacy of iPSCs for OA are scheduled this year. Mesenchymal stromal stem cells (MSCs) have inherent limitations in that they differentiate into the osteochondral pathway, resulting in the production of poor-quality cartilage. Despite the established essential role of TGF-β in chondrogenic differentiation of MSCs, whether the coordinated use of TGF-β in MSC-based therapy for degenerated cartilage is effective is unknown. We herein reviewed the general characteristics and mechanism of action of TGF-β in a joint environment. Furthermore, we discussed the core interaction of TGF-β with principal cells of OA cell-based therapies, the chondrocytes, MSCs, and iPSCs. Impact Statement Transforming growth factor-beta (TGF-β) has been widely used as a core regulator to improve or formulate therapeutic regenerative cells for degenerative joints. It differentiates stem cells into chondrocytes and improves the chondrogenic potential of differentiated chondrocytes. Herein, we discussed the overall characteristics of TGF-β and reviewed the comprehension and utilization of TGF-β in cell-based therapy for degenerative joint disease.
Collapse
Affiliation(s)
- Jung Gon Kim
- Division of Rheumatology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, 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
|
18
|
Xie M, Zhang Y, Xiong Z, Hines S, Shangjiang Y, Clark KL, Tan S, Alexander PG, Lin H. Generation of hyaline-like cartilage tissue from human mesenchymal stromal cells within the self-generated extracellular matrix. Acta Biomater 2022; 149:150-166. [PMID: 35779770 DOI: 10.1016/j.actbio.2022.06.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022]
Abstract
Chondrocytic hypertrophy, a phenotype not observed in healthy hyaline cartilage, is often concomitant with the chondrogenesis of human mesenchymal stromal cells (hMSCs). This undesired feature represents one of the major obstacles in applying hMSCs for hyaline cartilage repair. Previously, we developed a method to induce hMSC chondrogenesis within self-generated extracellular matrix (mECM), which formed a cartilage tissue with a lower hypertrophy level than conventional hMSC pellets. In this study, we aimed to test the utility of hypoxia and insulin-like growth factor-1 (IGF1) on further reducing hypertrophy. MSC-mECM constructs were first subjected to chondrogenic culture in normoxic or hypoxic (5%) conditions. The results indicated that hMSC-derived cartilage formed in hypoxic culture displayed a significantly reduced hypertrophy level than normoxic culture. However, hMSC chondrogenesis was also suppressed under hypoxic culture, partially due to the reduced activity of the IGF1 pathway. IGF1 was then supplemented in the chondrogenic medium, which promoted remarkable hMSC chondrogenesis under hypoxic culture. Interestingly, the IGF1-enhanced hMSC chondrogenesis, under hypoxic culture, was not at the expense of promoting significantly increased hypertrophy. Lastly, the cartilage tissues created by hMSCs with different conditions were implanted into osteochondral defect in rats. The results indicated that the tissue formed under hypoxic condition and induced with IGF1-supplemented chondrogenic medium displayed the best reparative results with minimal hypertrophy level. Our results demonstrate a new method to generate hyaline cartilage-like tissue from hMSCs without using exogenous scaffolds, which further pave the road for the clinical application of hMSC-based cartilage tissue engineering. STATEMENT OF SIGNIFICANCE: In this study, hyaline cartilage-like tissues were generated from human mesenchymal stromal cells (hMSCs), which displayed robust capacity in repairing the osteochondral defect in rats. In particular, the extracellular matrix created by hMSCs was used, so no exogenous scaffold was needed. Through a series of optimization, we defined that hypoxic culture and supplementation of insulin-like growth factor-1 (IGF-1) in chondrogenic medium resulted in robust cartilage formation with minimal hypertrophy. We also demonstrated that hypoxic culture suppressed chondrogenesis and hypertrophy through modulating the Wnt/β-catenin and IGF1 pathways, respectively. Our results demonstrate a new method to generate hyaline cartilage-like tissue from hMSCs without using exogenous scaffolds, which will further pave the road for the clinical application of hMSCs-based cartilage tissue engineering.
Collapse
Affiliation(s)
- Mingsheng Xie
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yiqian Zhang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zixuan Xiong
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410008, China
| | - Sophie Hines
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA
| | - Yingzi Shangjiang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA
| | - Karen L Clark
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA
| | - Susheng Tan
- Department of Electrical and Computer Engineering, Swanson School of Engineering, and Petersen Institute of NanoScience and Engineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Peter G Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15219, USA.
| |
Collapse
|
19
|
Wang X, Guan Y, Xiang S, Clark KL, Alexander PG, Simonian LE, Deng Y, Lin H. Role of Canonical Wnt/β-Catenin Pathway in Regulating Chondrocytic Hypertrophy in Mesenchymal Stem Cell-Based Cartilage Tissue Engineering. Front Cell Dev Biol 2022; 10:812081. [PMID: 35141220 PMCID: PMC8820467 DOI: 10.3389/fcell.2022.812081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/06/2022] [Indexed: 01/14/2023] Open
Abstract
In the past 3 decades, the cartilage repair potential of mesenchymal stromal cells, or mesenchymal stem cells (MSCs), has been widely examined in animal studies. Unfortunately, the phenotype and physical properties of MSC-derived cartilage tissue are not comparable to native hyaline cartilage. In particular, chondrocytic hypertrophy, a phenotype that is not observed in healthy hyaline cartilage, is concomitant with MSC chondrogenesis. Given that hypertrophic chondrocytes potentially undergo apoptosis or convert into osteoblasts, this undesired phenotype needs to be prevented or minimized before MSCs can be used to repair cartilage injuries in the clinic. In this review, we first provide an overview of chondrocytic hypertrophy and briefly summarize current methods for suppressing hypertrophy in MSC-derived cartilage. We then highlight recent progress on modulating the canonical Wnt/β-catenin pathway for inhibiting hypertrophy. Specially, we discuss the potential crosstalk between Wnt/β-catenin with other pathways in regulating hypertrophy. Lastly, we explore future perspectives to further understand the role of Wnt/β-catenin in chondrocytic hypertrophy.
Collapse
Affiliation(s)
- Xueqi Wang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yiming Guan
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shiyu Xiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Karen L. Clark
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Peter G. Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Lauren E. Simonian
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yuhao Deng
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Hang Lin, ; Yuhao Deng,
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- *Correspondence: Hang Lin, ; Yuhao Deng,
| |
Collapse
|
20
|
Deng Y, Zhang X, Li R, Li Z, Yang B, Shi P, Zhang H, Wang C, Wen C, Li G, Bian L. Biomaterial-mediated presentation of wnt5a mimetic ligands enhances chondrogenesis and metabolism of stem cells by activating non-canonical Wnt signaling. Biomaterials 2021; 281:121316. [PMID: 34959028 DOI: 10.1016/j.biomaterials.2021.121316] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022]
Abstract
The presentation of development-relevant bioactive cues by biomaterial scaffolds is essential to the guided differentiation of seeded human mesenchymal stem cells (hMSCs) and subsequent tissue regeneration. Wnt5a is a critical non-canonical Wnt signaling ligand and plays a key role in the development of musculoskeletal tissues including cartilage. Herein we investigate the efficacy of biofunctionalizing the hyaluronic acid hydrogel with a synthetic Wnt5a mimetic ligand (Foxy5 peptide) to promote the chondrogenesis of hMSCs and the potential underlying molecular mechanism. Our findings show that the conjugation of Foxy5 peptide in the hydrogels activates non-canonical Wnt signaling of encapsulated hMSCs via the upregulation expression of PLCE1, CaMKII-β, and downstream NFATc1, leading to enhanced expression of chondrogenic markers such as SOX9. The decoration of Foxy5 peptide also promotes the metabolic activities of encapsulated hMSCs as evidenced by upregulated gene expression of mitochondrial complex components and glucose metabolism biomarkers, leading to enhanced ATP biosynthesis. Furthermore, the conjugation of Foxy5 peptide activates the non-canonical Wnt, PI3K-PDK-AKT and IKK/NF-κB signaling pathways, thereby inhibiting the hypertrophy of the chondrogenically induced hMSCs in the hydrogels under both in vitro and in vivo conditions. This enhanced chondrogenesis and attenuated hypertrophy of hMSCs by the biomaterial-mediated bioactive cue presentation facilitates the potential clinical translation of hMSCs for cartilage regeneration. Our work provides valuable guidance to the rational design of bio-inductive scaffolds for various applications in regenerative medicine.
Collapse
Affiliation(s)
- Yingrui Deng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China
| | - Xiaoting Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China
| | - Rui Li
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Honglu Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Macau SAR, China
| | - Chunyi Wen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China.
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China; Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, PR China.
| |
Collapse
|
21
|
Melnik S, Hofmann N, Gabler J, Hecht N, Richter W. MiR-181a Targets RSPO2 and Regulates Bone Morphogenetic Protein - WNT Signaling Crosstalk During Chondrogenic Differentiation of Mesenchymal Stromal Cells. Front Cell Dev Biol 2021; 9:747057. [PMID: 34778258 PMCID: PMC8586458 DOI: 10.3389/fcell.2021.747057] [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: 07/25/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Mechanisms of WNT and bone morphogenetic protein (BMP) signaling crosstalk is in the focus of multiple biological studies, and it also has been discovered to play important roles in human mesenchymal stromal cells (MSC) that are of great interest for neocartilage engineering due to their high chondrogenic differentiation potential. However, MSC-derived chondrocytes undergo hypertrophic degeneration that impedes their clinical application for cartilage regeneration. In our previous study, we established that several microRNAs (miRs) are differentially expressed between articular chondrocytes (AC) - and MSC-derived neocartilage, with miR-181a being the most prominent candidate as key microRNA involved in the regulation of a balance between chondral and endochondral differentiation. The aim of this study was the identification of precise mRNA targets and signaling pathways regulated by miR-181a in MSC during chondrogenesis. MiR-181a was upregulated during chondrogenesis of MSC, along with an increase of the hypertrophic phenotype in resulting cartilaginous tissue. By in silico analysis combined with miR reporter assay, the WNT signaling activator and BMP signaling repressor RSPO2 was suggested as a target of miR-181a. Further validation experiments confirmed that miR-181a targets RSPO2 mRNA in MSC. It was found that in human MSC miR-181a activated BMP signaling manifested by the accumulation of SOX9 protein and increased phosphorylation of SMAD1/5/9. These effects, together with the concomitant reduction of canonical WNT signaling induced by miR-181a mimic, were in accordance with the effects expected by the loss of RSPO2, thus indicating the causative link between miR-181a and RSPO2. Moreover, we observed that a tight correlation between miR-181a and miR-218 expression levels in healthy human cartilage tissue was disrupted in osteoarthritis (OA) highlighting the importance of the WNT-BMP signaling crosstalk for preventing OA.
Collapse
Affiliation(s)
- Svitlana Melnik
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Nina Hofmann
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jessica Gabler
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicole Hecht
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
22
|
Lees-Shepard JB, Flint K, Fisher M, Omi M, Richard K, Antony M, Chen PJ, Yadav S, Threadgill D, Maihle NJ, Dealy CN. Cross-talk between EGFR and BMP signals regulates chondrocyte maturation during endochondral ossification. Dev Dyn 2021; 251:75-94. [PMID: 34773433 DOI: 10.1002/dvdy.438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Progressive maturation of growth plate chondrocytes drives long bone growth during endochondral ossification. Signals from the epidermal growth factor receptor (EGFR), and from bone morphogenetic protein-2 (BMP2), are required for normal chondrocyte maturation. Here, we investigated cross-talk between EGFR and BMP2 signals in developing and adult growth plates. RESULTS Using in vivo mouse models of conditional cartilage-targeted EGFR or BMP2 loss, we show that canonical BMP signal activation is increased in the hypertrophic chondrocytes of EGFR-deficient growth plates; whereas EGFR signal activation is increased in the reserve, prehypertrophic and hypertrophic chondrocytes of BMP2-deficient growth plates. EGFR-deficient chondrocytes displayed increased BMP signal activation in vitro, accompanied by increased expression of IHH, COL10A1, and RUNX2. Hypertrophic differentiation and BMP signal activation were suppressed in normal chondrocyte cultures treated with the EGFR ligand betacellulin, effects that were partially blocked by simultaneous treatment with BMP2 or a chemical EGFR antagonist. CONCLUSIONS Cross-talk between EGFR and BMP2 signals occurs during chondrocyte maturation. In the reserve and prehypertrophic zones, BMP2 signals unilaterally suppress EGFR activity; in the hypertrophic zone, EGFR and BMP2 signals repress each other. This cross-talk may play a role in regulating chondrocyte maturation in developing and adult growth plates.
Collapse
Affiliation(s)
- John B Lees-Shepard
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Kaitlyn Flint
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Melanie Fisher
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Minoru Omi
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Kelsey Richard
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Michelle Antony
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Po Jung Chen
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Sumit Yadav
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David Threadgill
- Department of Veterinary Pathology, Texas A&M University, College Station, Texas, USA.,Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, USA
| | - Nita J Maihle
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA.,Department of Cell & Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Caroline N Dealy
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA.,Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT, USA.,Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA.,Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
23
|
Non-thermal atmospheric pressure plasma activates Wnt/β-catenin signaling in dermal papilla cells. Sci Rep 2021; 11:16125. [PMID: 34373562 PMCID: PMC8352944 DOI: 10.1038/s41598-021-95650-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 07/28/2021] [Indexed: 12/03/2022] Open
Abstract
There is an unmet need for novel, non-pharmacological therapeutics to treat alopecia. Recent studies have shown the potential biological benefits of non-thermal atmospheric pressure plasma (NTAPP), including wound healing, angiogenesis, and the proliferation of stem cells. We hypothesized that NTAPP might have a stimulatory effect on hair growth or regeneration. We designed an NTAPP-generating apparatus which is applicable to in vitro and in vivo experiments. The human dermal papilla (DP) cells, isolated fresh hair follicles, and mouse back skin were exposed with the NTAPP. Biological outcomes were measured using RNA-sequencing, RT-PCR, Western blots, and immunostaining. The NTAPP treatment increased the expression levels of Wnt/β-catenin pathway-related genes (AMER3, CCND1, LEF1, and LRG1) and proteins (β-catenin, p-GSK3β, and cyclin D1) in human DP cells. In contrast, inhibitors of Wnt/β-catenin signaling, endo-IWR1 and IWP2, attenuated the levels of cyclin D1, p-GSK3β, and β-catenin proteins induced by NTAPP. Furthermore, we observed that NTAPP induced the activation of β-catenin in DP cells of hair follicles and the mRNA levels of target genes of the β-catenin signaling pathway (CCND1, LEF1, and TCF4). NTAPP-treated mice exhibited markedly increased anagen induction, hair growth, and the protein levels of β-catenin, p-GSK3β, p-AKT, and cyclin D1. NTAPP stimulates hair growth via activation of the Wnt/β-catenin signaling pathway in DP cells. These findings collectively suggest that NTAPP may be a potentially safe and non-pharmacological therapeutic intervention for alopecia.
Collapse
|
24
|
Tang RF, Zhou XZ, Niu L, Qi YY. Type I collagen scaffold with WNT5A plasmid for in situ cartilage tissue engineering. Biomed Mater Eng 2021; 33:65-76. [PMID: 34366316 DOI: 10.3233/bme-211277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cartilage tissue lacks the ability to heal. Cartilage tissue engineering using cell-free scaffolds has been increasingly used in recent years. OBJECTIVE This study describes the use of a type I collagen scaffold combined with WNT5A plasmid to promote chondrocyte proliferation and differentiation in a rabbit osteochondral defect model. METHODS Type I collagen was extracted and fabricated into a collagen scaffold. To improve gene transfection efficiency, a cationic chitosan derivative N,N,N-trimethyl chitosan chloride (TMC) vector was used. A solution of TMC/WNT5A complexes was adsorbed to the collagen scaffold to prepare a WNT5A scaffold. Osteochondral defects were created in the femoral condyles of rabbits. The rabbits were divided into defect, scaffold, and scaffold with WNT5A groups. At 6 and 12 weeks after creation of the osteochondral defects, samples were collected from all groups for macroscopic observation and gene expression analysis. RESULTS Samples from the defect group exhibited incomplete cartilage repair, while those from the scaffold and scaffold with WNT5A groups exhibited "preliminary cartilage" covering the defect. Cartilage regeneration was superior in the scaffold with WNT5A group compared to the scaffold group. Safranin O staining revealed more proteoglycans in the scaffold and scaffold with WNT5A groups compared to the defect group. The expression levels of aggrecan, collagen type II, and SOX9 genes were significantly higher in the scaffold with WNT5A group compared to the other two groups. CONCLUSIONS Type I collagen scaffold showed effective adsorption and guided the three-dimensional arrangement of stem cells. WNT5A plasmid promoted cartilage repair by stimulating the expression of aggrecan, type II collagen, and SOX9 genes and proteins, as well as inhibiting cartilage hypertrophy.
Collapse
Affiliation(s)
- Ruo-Fu Tang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Lie Niu
- Department of Orthopedics, Dongping People's Hospital, ShanDong, China
| | - Yi-Ying Qi
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| |
Collapse
|
25
|
Xie H, Zhou L, Chen Z, Zhao H. The Effect of Wnt Family Member 5a Gene Silencing on the Proliferation of Achondroplasia Using a DNAzymes-CoOOH Nanocomposite. J Biomed Nanotechnol 2021; 17:1426-1434. [PMID: 34446145 DOI: 10.1166/jbn.2021.3119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Achondroplasia is a kind of congenital dysplasia due to the defect of endochondral ossification. Achondroplasia is considered to be a protein folding disease leading to endoplasmic reticulum stress. Endoplasmic reticulum stress may lead to disease by affecting the function and survival state of chondrocytes, but the specific mechanism requires further study. In this study, bioinformatics methods, online database mining, screening of differentially expressed genes for pathway enrichment, and interaction analysis were conducted to detect the Wnt family member 5a (Wnt5a) gene. Additionally, we designed a novel DNAzymes-based nanocomposite that can simultaneously silence Wnt5a genes in chondrocytes. The nanocomposite was composed of amino-functionalized cobalt oxyhydroxide nanoflakes modified by DNAzymes that target the Wnt5a gene. Further, we conducted in vitro experiments to verify that Wnt5a can mediate the mitogen-activated protein kinase signaling pathway through the endoplasmic reticulum stress pathway to affect the proliferation of chondrocytes.
Collapse
Affiliation(s)
- Hairui Xie
- Department of Pediatrics Center, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, Guangdong, PR China
| | - Lili Zhou
- Department of Pediatrics Center, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, Guangdong, PR China
| | - Zhijiang Chen
- Department of Pediatrics Center, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, Guangdong, PR China
| | - Hong Zhao
- Department of Pediatrics Center, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, Guangdong, PR China
| |
Collapse
|
26
|
Guan Z, Chen S, Pan F, Fan L, Sun D. Effects of Gene Delivery Approaches on Differentiation Potential and Gene Function of Mesenchymal Stem Cells. IEEE Trans Biomed Eng 2021; 69:83-95. [PMID: 34101578 DOI: 10.1109/tbme.2021.3087129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Introduction of a gene to mesenchymal stem cells (MSCs) is a well-known strategy to purposely manipulate the cell fate and further enhance therapeutic performance in cell-based therapy. Viral and chemical approaches for gene delivery interfere with differentiation potential. Although microinjection as a physical delivery method is commonly used for transfection, its influence on MSC cell fate is not fully understood. The current study aimed to evaluate the effects of four nonviral gene delivery methods on stem cell multi-potency. The four delivery methods are robotic microinjection, polyethylenimine (PEI), cationic liposome (cLipo), and calcium phosphate nanoparticles (CaP). Among the four methods, microinjection has exhibited the highest transfection efficiency of ~60%, while the three others showed lower efficiency of 10-25%. Robotic microinjection preserved fibroblast-like cell morphology, stress fibre intactness, and mature focal adhesion complex, while PEI caused severe cytotoxicity. No marked differentiation bias was observed after microinjection and cLipo treatment. By contrast, CaP-treated MSCs exhibited excessive osteogenesis, while PEI-treated MSCs showed excessive adipogenesis. Robotic microinjection system was used to inject the CRISPR/Cas9-encoding plasmid to knock out PPAR gene in MSCs, and the robotic microinjection did not interfere with PPAR function in differentiation commitment. Meanwhile, the bias in osteo-adipogenic differentiation exhibited in CaP and PEI-treated MSCs after PPAR knockout via chemical carriers. Our results indicate that gene delivery vehicles variously disturb MSCs differentiation and interfere with exogenous gene function. Our findings further suggest that robotic microinjection offers a promise of generating genetically modified MSCs without disrupting stem cell multi-potency and therapeutic gene function.
Collapse
|
27
|
Riquier S, Mathieu M, Bessiere C, Boureux A, Ruffle F, Lemaitre JM, Djouad F, Gilbert N, Commes T. Long non-coding RNA exploration for mesenchymal stem cell characterisation. BMC Genomics 2021; 22:412. [PMID: 34088266 PMCID: PMC8178833 DOI: 10.1186/s12864-020-07289-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The development of RNA sequencing (RNAseq) and the corresponding emergence of public datasets have created new avenues of transcriptional marker search. The long non-coding RNAs (lncRNAs) constitute an emerging class of transcripts with a potential for high tissue specificity and function. Therefore, we tested the biomarker potential of lncRNAs on Mesenchymal Stem Cells (MSCs), a complex type of adult multipotent stem cells of diverse tissue origins, that is frequently used in clinics but which is lacking extensive characterization. RESULTS We developed a dedicated bioinformatics pipeline for the purpose of building a cell-specific catalogue of unannotated lncRNAs. The pipeline performs ab initio transcript identification, pseudoalignment and uses new methodologies such as a specific k-mer approach for naive quantification of expression in numerous RNAseq data. We next applied it on MSCs, and our pipeline was able to highlight novel lncRNAs with high cell specificity. Furthermore, with original and efficient approaches for functional prediction, we demonstrated that each candidate represents one specific state of MSCs biology. CONCLUSIONS We showed that our approach can be employed to harness lncRNAs as cell markers. More specifically, our results suggest different candidates as potential actors in MSCs biology and propose promising directions for future experimental investigations.
Collapse
Affiliation(s)
- Sébastien Riquier
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Marc Mathieu
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Chloé Bessiere
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Anthony Boureux
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Florence Ruffle
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Jean-Marc Lemaitre
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Farida Djouad
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Nicolas Gilbert
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Thérèse Commes
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| |
Collapse
|
28
|
Chen H, Tan XN, Hu S, Liu RQ, Peng LH, Li YM, Wu P. Molecular Mechanisms of Chondrocyte Proliferation and Differentiation. Front Cell Dev Biol 2021; 9:664168. [PMID: 34124045 PMCID: PMC8194090 DOI: 10.3389/fcell.2021.664168] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022] Open
Abstract
Cartilage is a kind of connective tissue that buffers pressure and is essential to protect joint movement. It is difficult to self-recover once cartilage is damaged due to the lack of blood vessels, lymph, and nerve tissues. Repair of cartilage injury is mainly achieved by stimulating chondrocyte proliferation and extracellular matrix (ECM) synthesis. Cartilage homeostasis involves the regulation of multiple growth factors and the transduction of cellular signals. It is a very complicated process that has not been elucidated in detail. In this review, we summarized a variety of signaling molecules related to chondrocytes function. Especially, we described the correlation between chondrocyte-specific regulatory factors and cell signaling molecules. It has potential significance for guiding the treatment of cartilage injury.
Collapse
Affiliation(s)
- Hui Chen
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| | - Xiao-Ning Tan
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China
| | - Shi Hu
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China.,Center for Bionic Sensing and Intelligence, Institute of Bio-medical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ren-Qin Liu
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| | - Li-Hong Peng
- School of Computer, Hunan University of Technology, Zhuzhou, China
| | - Yong-Min Li
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China
| | - Ping Wu
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| |
Collapse
|
29
|
Saeidinezhad M, Razban V, Safizadeh H, Ezzatabadipour M. Effects of maternal consumption of morphine on rat skeletal system development. BMC Musculoskelet Disord 2021; 22:435. [PMID: 33985485 PMCID: PMC8120720 DOI: 10.1186/s12891-021-04321-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/05/2021] [Indexed: 11/19/2022] Open
Abstract
Background Opioid abuse is among the most ubiquitous issues world-wide, and when it happens in mothers, it puts them at risk of diseases that can be transferred to the next generation. Previous studies have indicated that morphine addiction during pregnancy could inhibit development in rat embryos and infants. The present study focused on the effects of maternal consumption of morphine on rat skeletal system development and also investigate the molecular pathway of chondrogenesis and osteogenesis of infants from control and addicted rat groups. Methods Thirty-two female rats were randomly assigned to four groups. The groups consisted of one- and seven-day-old female infants which were born of morphine-dependent mothers and a control group for each of them. Experimental groups received oral morphine at the final dose of 0.4 mg/ml/day. Withdrawal signs were confirmation of morphine dependency. Female rats were crossed with male rats and coupling time was recorded. Fixed bones of all groups were processed and then stained by hematoxyline-eosin method. Thickness and cell number of proximal and distal growth plate of bones were measured. The cartilage and bone cells were stained by alcian blue/alizarin red method. Additionally, the gene expression of alkaline phosphatase, osteocalcin, and COLL2 and SOX9 gene expression were studied immuno-histochemically. Results Unfavorable effects of morphine on histological measurements were observed in one-day and seven-day infants, with more effects on seven-day infants. The thickness and cell number of the proximal and distal growth plate of morphine-dependent rat offsprings were reduced significantly. Furthermore, morphine reduced growth of primary and secondary ossification centers, and thus, longitudinal bone growth was reduced. Moreover, a decrease in the alkaline phosphatase, osteocalcin, COLL2 and SOX9 gene expression, and the number of stained cells was observed. More adverse effects of morphine in seven-day infants compared to one-day infants which showed the time dependent of morphine to the time length of administration. Conclusion Histochemistry and immunohistochemistry findings on cartilage and bone matrix formation, as well as protein expression of chondrogenic and osteogenic markers suggest that morphine dependence in pregnant mothers may impair intra-cartilaginous osteogenesis in post-natal rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-021-04321-6.
Collapse
Affiliation(s)
- Maryam Saeidinezhad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Somayeh Cross-road, Sajad Boulevard, Ebnesina Street, Kerman, 7619813159, Iran.,Department of Anatomical Sciences, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Stem cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hosein Safizadeh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Somayeh Cross-road, Sajad Boulevard, Ebnesina Street, Kerman, 7619813159, Iran
| | - Massood Ezzatabadipour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Somayeh Cross-road, Sajad Boulevard, Ebnesina Street, Kerman, 7619813159, Iran. .,Department of Anatomical Sciences, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
30
|
Ling Y, Zhang W, Wang P, Xie W, Yang W, Wang DA, Fan C. Three-dimensional (3D) hydrogel serves as a platform to identify potential markers of chondrocyte dedifferentiation by combining RNA sequencing. Bioact Mater 2021; 6:2914-2926. [PMID: 33718672 PMCID: PMC7917462 DOI: 10.1016/j.bioactmat.2021.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Dedifferentiation of chondrocyte greatly restricts its function and application, however, it is poorly understood except a small number of canonical markers. The non-cell-adhesive property endows polysaccharide hydrogel with the ability to maintain chondrocyte phenotype, which can serve as a platform to identify new molecular markers and therapeutic targets of chondrocyte dedifferentiation. In this study, the high-throughput RNA sequencing (RNA-seq) was first performed on articular chondrocytes at primary (P0) and passage 1 (P1) stages to explore the global alteration of gene expression along with chondrocyte dedifferentiation. Significantly, several potential marker genes, such as PFKFB3, KDM6B, had been identified via comparatively analyzing their expression in P0 and P1 chondrocytes as well as in 3D constructs (i.e. chondrocyte-laden alginate hydrogel and HA-MA hydrogel) at both mRNA and protein level. Besides, the changes in cellular morphology and enriched pathway of differentially expressed genes during chondrocyte dedifferentiation was studied in detail. This study developed the use of hydrogel as a platform to investigate chondrocyte dedifferentiation; the results provided new molecular markers and potential therapeutic targets of chondrocyte dedifferentiation.
Collapse
Affiliation(s)
- Yang Ling
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, PR China.,Department of Human Anatomy Histology and Embryology, School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Weiyuan Zhang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Peiyan Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Wanhua Xie
- The Precise Medicine Center, Shenyang Medical College, Shenyang, 110034, Liaoning, PR China
| | - Wei Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, PR China.,School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China.,Shenzhen Research Institute, City University of Hong Kong, Shenzhen Hi-tech Industrial Park, Shenzhen, Guangdong, 518057, PR China.,Karolinska Institute Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong, China
| | - Changjiang Fan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, PR China.,Department of Human Anatomy Histology and Embryology, School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| |
Collapse
|
31
|
Saghati S, Nasrabadi HT, Khoshfetrat AB, Moharamzadeh K, Hassani A, Mohammadi SM, Rahbarghazi R, Fathi Karkan S. Tissue Engineering Strategies to Increase Osteochondral Regeneration of Stem Cells; a Close Look at Different Modalities. Stem Cell Rev Rep 2021; 17:1294-1311. [PMID: 33547591 DOI: 10.1007/s12015-021-10130-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
The homeostasis of osteochondral tissue is tightly controlled by articular cartilage chondrocytes and underlying subchondral bone osteoblasts via different internal and external clues. As a correlate, the osteochondral region is frequently exposed to physical forces and mechanical pressure. On this basis, distinct sets of substrates and physicochemical properties of the surrounding matrix affect the regeneration capacity of chondrocytes and osteoblasts. Stem cells are touted as an alternative cell source for the alleviation of osteochondral diseases. These cells appropriately respond to the physicochemical properties of different biomaterials. This review aimed to address some of the essential factors which participate in the chondrogenic and osteogenic capacity of stem cells. Elements consisted of biomechanical forces, electrical fields, and biochemical and physical properties of the extracellular matrix are the major determinant of stem cell differentiation capacity. It is suggested that an additional certain mechanism related to signal-transduction pathways could also mediate the chondro-osteogenic differentiation of stem cells. The discovery of these clues can enable us to modulate the regeneration capacity of stem cells in osteochondral injuries and lead to the improvement of more operative approaches using tissue engineering modalities.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Baradar Khoshfetrat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine (HBMCDM), Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Seyedeh Momeneh Mohammadi
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sonia Fathi Karkan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Wu CL, Dicks A, Steward N, Tang R, Katz DB, Choi YR, Guilak F. Single cell transcriptomic analysis of human pluripotent stem cell chondrogenesis. Nat Commun 2021; 12:362. [PMID: 33441552 PMCID: PMC7806634 DOI: 10.1038/s41467-020-20598-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 12/10/2020] [Indexed: 01/29/2023] Open
Abstract
The therapeutic application of human induced pluripotent stem cells (hiPSCs) for cartilage regeneration is largely hindered by the low yield of chondrocytes accompanied by unpredictable and heterogeneous off-target differentiation of cells during chondrogenesis. Here, we combine bulk RNA sequencing, single cell RNA sequencing, and bioinformatic analyses, including weighted gene co-expression analysis (WGCNA), to investigate the gene regulatory networks regulating hiPSC differentiation under chondrogenic conditions. We identify specific WNTs and MITF as hub genes governing the generation of off-target differentiation into neural cells and melanocytes during hiPSC chondrogenesis. With heterocellular signaling models, we further show that WNT signaling produced by off-target cells is responsible for inducing chondrocyte hypertrophy. By targeting WNTs and MITF, we eliminate these cell lineages, significantly enhancing the yield and homogeneity of hiPSC-derived chondrocytes. Collectively, our findings identify the trajectories and molecular mechanisms governing cell fate decision in hiPSC chondrogenesis, as well as dynamic transcriptome profiles orchestrating chondrocyte proliferation and differentiation.
Collapse
Affiliation(s)
- Chia-Lung Wu
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, 14627, USA
| | - Amanda Dicks
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA
- Dept. of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, 63110, USA
| | - Nancy Steward
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA
| | - Ruhang Tang
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA
| | - Dakota B Katz
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA
- Dept. of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, 63110, USA
| | - Yun-Rak Choi
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA
- Dept. of Orthopaedic Surgery, Yonsei University, Seoul, South Korea
| | - Farshid Guilak
- Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, 63110, USA.
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, 63110, USA.
- Dept. of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
33
|
Futrega K, Music E, Robey PG, Gronthos S, Crawford R, Saifzadeh S, Klein TJ, Doran MR. Characterisation of ovine bone marrow-derived stromal cells (oBMSC) and evaluation of chondrogenically induced micro-pellets for cartilage tissue repair in vivo. Stem Cell Res Ther 2021; 12:26. [PMID: 33413652 PMCID: PMC7791713 DOI: 10.1186/s13287-020-02045-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract Bone marrow stromal cells (BMSC) show promise in cartilage repair, and sheep are the most common large animal pre-clinical model. Objective The objective of this study was to characterise ovine BMSC (oBMSC) in vitro, and to evaluate the capacity of chondrogenic micro-pellets manufactured from oBMSC or ovine articular chondrocytes (oACh) to repair osteochondral defects in sheep. Design oBMSC were characterised for surface marker expression using flow cytometry and evaluated for tri-lineage differentiation capacity. oBMSC micro-pellets were manufactured in a microwell platform, and chondrogenesis was compared at 2%, 5%, and 20% O2. The capacity of cartilage micro-pellets manufactured from oBMSC or oACh to repair osteochondral defects in adult sheep was evaluated in an 8-week pilot study. Results Expanded oBMSC were positive for CD44 and CD146 and negative for CD45. The common adipogenic induction ingredient, 3-Isobutyl-1-methylxanthine (IBMX), was toxic to oBMSC, but adipogenesis could be restored by excluding IBMX from the medium. BMSC chondrogenesis was optimal in a 2% O2 atmosphere. Micro-pellets formed from oBMSC or oACh appeared morphologically similar, but hypertrophic genes were elevated in oBMSC micro-pellets. While oACh micro-pellets formed cartilage-like repair tissue in sheep, oBMSC micro-pellets did not. Conclusion The sensitivity of oBMSC, compared to human BMSC, to IBMX in standard adipogenic assays highlights species-associated differences. Micro-pellets manufactured from oACh were more effective than micro-pellets manufactured from oBMSC in the repair of osteochondral defects in sheep. While oBMSC can be driven to form cartilage-like tissue in vitro, the effective use of these cells in cartilage repair will depend on the successful mitigation of hypertrophy and tissue integration. Supplementary information The online version contains supplementary material available at 10.1186/s13287-020-02045-3.
Collapse
Affiliation(s)
- K Futrega
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA.,Translational Research Institute (TRI), Brisbane, Queensland, Australia
| | - E Music
- Translational Research Institute (TRI), Brisbane, Queensland, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - P G Robey
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - S Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - R Crawford
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - S Saifzadeh
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - T J Klein
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - M R Doran
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia. .,National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA. .,Translational Research Institute (TRI), Brisbane, Queensland, Australia. .,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia. .,Mater Research Institute - University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, Queensland, Australia.
| |
Collapse
|
34
|
Anderson-Baron M, Liang Y, Kunze M, Mulet-Sierra A, Osswald M, Ansari K, Seikaly H, Adesida AB. Suppression of Hypertrophy During in vitro Chondrogenesis of Cocultures of Human Mesenchymal Stem Cells and Nasal Chondrocytes Correlates With Lack of in vivo Calcification and Vascular Invasion. Front Bioeng Biotechnol 2021; 8:572356. [PMID: 33469528 PMCID: PMC7813892 DOI: 10.3389/fbioe.2020.572356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023] Open
Abstract
Objective Human nasal septal chondrocytes (NC) are a promising minimally invasive derivable chondrogenic cell source for cartilage repair. However, the quality of NC-derived cartilage is variable between donors. Coculture of NC with mesenchymal stem cells (MSCs) mitigates the variability but with undesirable markers of chondrocyte hypertrophy, such as type X collagen, and the formation of unstable calcifying cartilage at ectopic sites. In contrast, monoculture NC forms non-calcifying stable cartilage. Formation of a stable NC-MSC coculture cartilage is crucial for clinical application. The aim of this study was to explore the utility of parathyroid hormone-related peptide (PTHrP) hormone to suppress chondrocyte hypertrophy in NC-MSC cocultures and form stable non-calcifying cartilage at ectopic sites. Methods Human NC and bone marrow MSCs, and cocultures of NC and MSC (1:3 ratio) were aggregated in pellet form and subjected to in vitro chondrogenesis for 3 weeks in chondrogenic medium in the presence and absence of PTHrP. Following in vitro chondrogenesis, the resulting pellets were implanted in immunodeficient athymic nude mice for 3 weeks. Results Coculture of NC and MSC resulted in synergistic cartilage matrix production. PTHrP suppressed the expression of hypertrophy marker, type X collagen (COL10A1), in a dose-dependent fashion without affecting the synergism in cartilage matrix synthesis, and in vivo calcification was eradicated with PTHrP. In contrast, cocultured control (CC) pellets without PTHrP treatment expressed COL10A1, calcified, and became vascularized in vivo.
Collapse
Affiliation(s)
- Matthew Anderson-Baron
- Division of Orthopaedic Surgery, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada.,Division of Surgical Research, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Division of Orthopaedic Surgery, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada.,Division of Surgical Research, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Division of Orthopaedic Surgery, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada.,Division of Surgical Research, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Division of Orthopaedic Surgery, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada.,Division of Surgical Research, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta Hospital, Edmonton, AB, Canada.,Institute for Reconstructive Sciences in Medicine, Misericordia Community Hospital, Edmonton, AB, Canada
| | - Khalid Ansari
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta Hospital, Edmonton, AB, Canada
| | - Hadi Seikaly
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta Hospital, Edmonton, AB, Canada
| | - Adetola B Adesida
- Division of Orthopaedic Surgery, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada.,Division of Surgical Research, Department of Surgery, Faculty of Medicine and Dentistry, Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, University of Alberta, Edmonton, AB, Canada.,Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
35
|
Futrega K, Robey PG, Klein TJ, Crawford RW, Doran MR. A single day of TGF-β1 exposure activates chondrogenic and hypertrophic differentiation pathways in bone marrow-derived stromal cells. Commun Biol 2021; 4:29. [PMID: 33398032 PMCID: PMC7782775 DOI: 10.1038/s42003-020-01520-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023] Open
Abstract
Virtually all bone marrow-derived stromal cell (BMSC) chondrogenic induction cultures include greater than 2 weeks exposure to transforming growth factor-β (TGF-β), but fail to generate cartilage-like tissue suitable for joint repair. Herein we used a micro-pellet model (5 × 103 BMSC each) to determine the duration of TGF-β1 exposure required to initiate differentiation machinery, and to characterize the role of intrinsic programming. We found that a single day of TGF-β1 exposure was sufficient to trigger BMSC chondrogenic differentiation and tissue formation, similar to 21 days of TGF-β1 exposure. Despite cessation of TGF-β1 exposure following 24 hours, intrinsic programming mediated further chondrogenic and hypertrophic BMSC differentiation. These important behaviors are obfuscated by diffusion gradients and heterogeneity in commonly used macro-pellet models (2 × 105 BMSC each). Use of more homogenous micro-pellet models will enable identification of the critical differentiation cues required, likely in the first 24-hours, to generate high quality cartilage-like tissue from BMSC.
Collapse
Affiliation(s)
- Kathryn Futrega
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD, USA
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
- Translational Research Institute (TRI), Brisbane, Queensland, Australia
| | - Pamela G Robey
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD, USA
| | - Travis J Klein
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Ross W Crawford
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Michael R Doran
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD, USA.
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.
- Translational Research Institute (TRI), Brisbane, Queensland, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.
- Mater Research Institute, University of Queensland (UQ), Brisbane, Queensland, Australia.
| |
Collapse
|
36
|
BMSC-derived exosomes from congenital polydactyly tissue alleviate osteoarthritis by promoting chondrocyte proliferation. Cell Death Discov 2020; 6:142. [PMID: 33303743 PMCID: PMC7730395 DOI: 10.1038/s41420-020-00374-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
In the past decade, mesenchymal stem cells (MSCs) have been widely used for the treatment of osteoarthritis (OA), and exosomes may play a major role. Here, we acquired a special kind of MSCs from the bone marrow of surgically resected tissue from the hand of a patient with polydactyly. Experiments were focused on the role of polydactyly bone marrow-derived MSCs (pBMSCs) in osteoarthritis. The results showed that the pBMSCs had a greater ability than the BMSCs to differentiate into chondrocytes. Mechanistically, the expression of BMP4 was significantly higher in the pBMSCs than it was in the BMSCs. Furthermore, we showed that the migration and proliferation of chondrocytes were stimulated by exosomes secreted by pBMSC (pBMSC-EXOs). Notably, the downregulation of BMP4 in pBMSCs by siRNA inhibited both the chondrogenic differentiation potential of the MSCs and the function of the chondrocytes. In addition, the injection of pBMSC-EXOs and BMSC-EXOs attenuated OA in an OA mouse model, but the pBMSC-EXOs had a superior therapeutic effect compared with that of the BMSC-EXOs. Taken together, the data indicate that pBMSCs have greater ability to differentiate into chondrocytes and regulate chondrocyte formation through BMP4 signaling. Therefore, pBMSC-EXOs may represent a novel treatment for OA.
Collapse
|
37
|
Melnik S, Gabler J, Dreher SI, Hecht N, Hofmann N, Großner T, Richter W. MiR-218 affects hypertrophic differentiation of human mesenchymal stromal cells during chondrogenesis via targeting RUNX2, MEF2C, and COL10A1. Stem Cell Res Ther 2020; 11:532. [PMID: 33303006 PMCID: PMC7727242 DOI: 10.1186/s13287-020-02026-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
Background Human mesenchymal stromal cells (MSC) hold hopes for cartilage regenerative therapy due to their chondrogenic differentiation potential. However, undesirable occurrence of calcification after ectopic transplantation, known as hypertrophic degeneration, remains the major obstacle limiting application of MSC in cartilage tissue regeneration approaches. There is growing evidence that microRNAs (miRs) play essential roles in post-transcriptional regulation of hypertrophic differentiation during chondrogenesis. Aim of the study was to identify new miR candidates involved in repression of hypertrophy-related targets. Methods The miR expression profile in human articular chondrocytes (AC) was compared to that in hypertrophic chondrocytes derived from human MSC by microarray analysis, and miR expression was validated by qPCR. Putative targets were searched by in silico analysis and validated by miR reporter assay in HEK293T, by functional assays (western blotting and ALP-activity) in transiently transfected SaOS-2 cells, and by a miR pulldown assay in human MSC. The expression profile of miR-218 was assessed by qPCR during in vitro chondrogenesis of MSC and re-differentiation of AC. MSC were transfected with miR-218 mimic, and differentiation outcome was assessed over 28 days. MiR-218 expression was quantified in healthy and osteoarthritic cartilage of patients. Results Within the top 15 miRs differentially expressed between chondral AC versus endochondral MSC differentiation, miR-218 was selected as a candidate miR predicted to target hypertrophy-related genes. MiR-218 was downregulated during chondrogenesis of MSC and showed a negative correlation to hypertrophic markers, such as COL10A1 and MEF2C. It was confirmed in SaOS-2 cells that miR-218 directly targets hypertrophy-related COL10A1, MEF2C, and RUNX2, as a gain of ectopic miR-218 mimic caused drop in MEF2C and RUNX2 protein accumulation, with attenuation of COL10A1 expression and significant concomitant reduction of ALP activity. A miR pulldown assay confirmed that miR-218 directly targets RUNX2, MEF2C in human MSC. Additionally, the gain of miR-218 in human MSC attenuated hypertrophic markers (MEF2C, RUNX2, COL10A1, ALPL), although with no boost of chondrogenic markers (GAG deposition, COL2A1) due to activation of WNT/β-catenin signaling. Moreover, no correlation between miR-218 expression and a pathologic phenotype in the cartilage of osteoarthritis (OA) patients was found. Conclusions Although miR-218 was shown to target pro-hypertrophic markers MEF2C, COL10A1, and RUNX2 in human MSC during chondrogenic differentiation, overall, it could not significantly reduce the hypertrophic phenotype or boost chondrogenesis. This could be explained by a concomitant activation of WNT/β-catenin signaling counteracting the anti-hypertrophic effects of miR-218. Therefore, to achieve a full inhibition of the endochondral pathway, a whole class of anti-hypertrophic miRs, including miR-218, needs to be taken into consideration.
Collapse
Affiliation(s)
- Svitlana Melnik
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jessica Gabler
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Simon I Dreher
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicole Hecht
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Nina Hofmann
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Großner
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
38
|
Dennis JE, Whitney GA, Rai J, Fernandes RJ, Kean TJ. Physioxia Stimulates Extracellular Matrix Deposition and Increases Mechanical Properties of Human Chondrocyte-Derived Tissue-Engineered Cartilage. Front Bioeng Biotechnol 2020; 8:590743. [PMID: 33282851 PMCID: PMC7691651 DOI: 10.3389/fbioe.2020.590743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Cartilage tissue has been recalcitrant to tissue engineering approaches. In this study, human chondrocytes were formed into self-assembled cartilage sheets, cultured in physiologic (5%) and atmospheric (20%) oxygen conditions and underwent biochemical, histological and biomechanical analysis at 1- and 2-months. The results indicated that sheets formed at physiological oxygen tension were thicker, contained greater amounts of glycosaminoglycans (GAGs) and type II collagen, and had greater compressive and tensile properties than those cultured in atmospheric oxygen. In all cases, cartilage sheets stained throughout for extracellular matrix components. Type II-IX-XI collagen heteropolymer formed in the neo-cartilage and fibrils were stabilized by trivalent pyridinoline cross-links. Collagen cross-links were not significantly affected by oxygen tension but increased with time in culture. Physiological oxygen tension and longer culture periods both served to increase extracellular matrix components. The foremost correlation was found between compressive stiffness and the GAG to collagen ratio.
Collapse
Affiliation(s)
| | | | - Jyoti Rai
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | - Russell J Fernandes
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | - Thomas J Kean
- Benaroya Research Institute, Seattle, WA, United States
| |
Collapse
|
39
|
Lv S, Xu J, Chen L, Wu H, Feng W, Zheng Y, Li P, Zhang H, Zhang L, Chi G, Li Y. MicroRNA-27b targets CBFB to inhibit differentiation of human bone marrow mesenchymal stem cells into hypertrophic chondrocytes. Stem Cell Res Ther 2020; 11:392. [PMID: 32917285 PMCID: PMC7488425 DOI: 10.1186/s13287-020-01909-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/09/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background Human bone marrow-derived mesenchymal stem cells (hBMSCs) have chondrocyte differentiation potential and are considered to be a cell source for cell-transplantation-mediated repair of cartilage defects, including those associated with osteoarthritis (OA). However, chondrocyte hypertrophic differentiation is a major obstacle for the application of hBMSCs in articular cartilage defect treatment. We have previously shown that microRNA-27b (miR-27b) inhibits hypertrophy of chondrocytes from rat knee cartilage. In this study, we investigated the role of miR-27b in chondrocyte hypertrophic differentiation of hBMSCs. Methods Chondrogenic marker and microRNA expression in hBMSC chondrogenic pellets were evaluated using RT-qPCR and immunohistochemistry. The hBMSCs were transfected with miR-27b before inducing differentiation. Gene and protein expression levels were analyzed using RT-qPCR and western blot. Coimmunoprecipitation was used to confirm interaction between CBFB and RUNX2. Luciferase reporter assays were used to demonstrate that CBFB is a miR-27b target. Chondrogenic differentiation was evaluated in hBMSCs treated with shRNA targeting CBFB. Chondrogenic hBMSC pellets overexpressing miR-27b were implanted into cartilage lesions in model rats; therapeutic effects were assessed based on histology and immunohistochemistry. Results The hBMSCs showed typical MSC differentiation potentials. During chondrogenic differentiation, collagen 2 and 10 (COL2 and COL10), SOX9, and RUNX2 expression was upregulated. Expression of miR-140, miR-143, and miR-181a increased over time, whereas miR-27b and miR-221 were downregulated. Cartilage derived from hBMSC and overexpressing miR-27b exhibited higher expression of COL2 and SOX9, but lower expression of COL10, RUNX2, and CBFB than did the control cartilage. CBFB and RUNX2 formed a complex, and CBFB was identified as a novel miR-27b target. CBFB knockdown by shRNA during hBMSC chondrogenic differentiation led to significantly increased COL2 and SOX9 expression and decreased COL10 expression. Finally, miR-27b-overexpressing hBMSC chondrogenic pellets had better hyaline cartilage morphology and reduced expression of hypertrophic markers and tend to increase repair efficacy in vivo. Conclusion MiR-27b plays an important role in preventing hypertrophic chondrogenesis of hBMSCs by targeting CBFB and is essential for maintaining a hyaline cartilage state. This study provides new insights into the mechanism of hBMSC chondrocyte differentiation and will aid in the development of strategies for treating cartilage injury based on hBMSC transplantation.
Collapse
Affiliation(s)
- Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Lin Chen
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.,Department of Gastrointestinal Surgery, Sino-Japanese Friendship Hospital of Jilin University, Changchun, 130021, China
| | - Haitao Wu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.,Department of Oncology, the First Hospital of Jilin University, Changchun, 130021, China
| | - Wei Feng
- Department of Bone and Joint, the First Hospital of Jilin University, Changchun, 130021, China
| | - Yangyang Zheng
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Pengdong Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Haiying Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Lihong Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
40
|
Qi J, Tang X, Li W, Chen W, Yao G, Sun L. Mesenchymal stem cells inhibited the differentiation of MDSCs via COX2/PGE2 in experimental sialadenitis. Stem Cell Res Ther 2020; 11:325. [PMID: 32727564 PMCID: PMC7391592 DOI: 10.1186/s13287-020-01837-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/25/2020] [Accepted: 07/16/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) can regulate innate and adaptive immune systems through interacting with immune cells directly and secreting multiple soluble factors. Due to their immunosuppressive properties, MSC transplantation has been applied to treat many clinical and experimental autoimmune diseases. However, the therapeutic effects and mechanisms by which MSCs regulate myeloid cells in Sjögren’s syndrome (SS) still remain elusive. Methods The number and immune-suppressive activity of myeloid-derived suppressor cells (MDSCs), polymorphonuclear MDSCs (PMN-MDSCs), and monocytic MDSCs (M-MDSCs) were determined in non-obese diabetic (NOD) mice with sialadenitis and in NOD mice with human umbilical cord-derived MSC (UC-MSC) transplantation. Bone marrow cells were cultured with MSC-conditioned medium (MSC-CM) for 4 days. The number and immune-suppressive gene of MDSCs were detected by flow cytometry or qRT-PCR. Results The results showed that the number of MDSCs and PMN-MDSCs was higher and M-MDSCs were lower in NOD mice with sialadenitis. UC-MSCs ameliorated SS-like syndrome by reducing MDSCs, PMN-MDSCs, and M-MDSCs and promoting the suppressive ability of MDSCs significantly in NOD mice. UC-MSCs inhibited the differentiation of MDSCs. In addition, UC-MSCs enhanced the suppressive ability of MDSCs in vitro. Mechanistically, MSCs inhibited the differentiation of MDSCs and PMN-MDSCs via secreting prostaglandin E2 (PGE2) and inhibited the differentiation of M-MDSCs through secreting interferon-β (IFN-β). Conclusions Our findings suggested that MSCs alleviated SS-like symptoms by suppressing the aberrant accumulation and improving the suppressive function of MDSCs in NOD mice with sialadenitis.
Collapse
Affiliation(s)
- Jingjing Qi
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, People's Republic of China.,Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, People's Republic of China
| | - Wenchao Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, People's Republic of China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, People's Republic of China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, People's Republic of China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
41
|
Nakayama N, Pothiawala A, Lee JY, Matthias N, Umeda K, Ang BK, Huard J, Huang Y, Sun D. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020; 77:2543-2563. [PMID: 31915836 PMCID: PMC11104892 DOI: 10.1007/s00018-019-03445-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The cartilage of joints, such as meniscus and articular cartilage, is normally long lasting (i.e., permanent). However, once damaged, especially in large animals and humans, joint cartilage is not spontaneously repaired. Compensating the lack of repair activity by supplying cartilage-(re)forming cells, such as chondrocytes or mesenchymal stromal cells, or by transplanting a piece of normal cartilage, has been the basis of therapy for biological restoration of damaged joint cartilage. Unfortunately, current biological therapies face problems on a number of fronts. The joint cartilage is generated de novo from a specialized cell type, termed a 'joint progenitor' or 'interzone cell' during embryogenesis. Therefore, embryonic chondroprogenitors that mimic the property of joint progenitors might be the best type of cell for regenerating joint cartilage in the adult. Pluripotent stem cells (PSCs) are expected to differentiate in culture into any somatic cell type through processes that mimic embryogenesis, making human (h)PSCs a promising source of embryonic chondroprogenitors. The major research goals toward the clinical application of PSCs in joint cartilage regeneration are to (1) efficiently generate lineage-specific chondroprogenitors from hPSCs, (2) expand the chondroprogenitors to the number needed for therapy without loss of their chondrogenic activity, and (3) direct the in vivo or in vitro differentiation of the chondroprogenitors to articular or meniscal (i.e., permanent) chondrocytes rather than growth plate (i.e., transient) chondrocytes. This review is aimed at providing the current state of research toward meeting these goals. We also include our recent achievement of successful generation of "permanent-like" cartilage from long-term expandable, hPSC-derived ectomesenchymal chondroprogenitors.
Collapse
Affiliation(s)
- Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA.
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - Katsutsugu Umeda
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Department of Pediatrics, Kyoto University School of Medicine, Kyoto, Japan
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yun Huang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Deqiang Sun
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
42
|
Chu YC, Lim J, Hwang WH, Lin YX, Wang JL. Piezoelectric stimulation by ultrasound facilitates chondrogenesis of mesenchymal stem cells. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2020; 148:EL58. [PMID: 32752766 DOI: 10.1121/10.0001590] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
A cellular stimulation device utilizing an AT-cut quartz coverslip mounted on an ultrasonic live imaging chamber is developed to investigate the effect of piezoelectric stimulation. Two types of chambers deliver ultrasound at intensities ranging from 1 to 20 mW/cm2 to mesenchymal stem cells (MSCs) seeded on the quartz coverslip. The quartz coverslip imposes additionally localized electric charges as it vibrates with the stimulation. The device was applied to explore whether piezoelectric stimulation can facilitate chondrogenesis of MSCs. The results suggest piezoelectric stimulation drove clustering of MSCs and consequently facilitated chondrogenesis of MSCs without the use of differentiation media.
Collapse
Affiliation(s)
- Ya-Cherng Chu
- Department of Biomedical Engineering, National Taiwan University, Taipei, , , , ,
| | - Jormay Lim
- Department of Biomedical Engineering, National Taiwan University, Taipei, , , , ,
| | - Wen-Hao Hwang
- Department of Biomedical Engineering, National Taiwan University, Taipei, , , , ,
| | - Yu-Xuan Lin
- Department of Biomedical Engineering, National Taiwan University, Taipei, , , , ,
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, National Taiwan University, Taipei, , , , ,
| |
Collapse
|
43
|
LncRNA ENST00000563492 promoting the osteogenesis-angiogenesis coupling process in bone mesenchymal stem cells (BMSCs) by functions as a ceRNA for miR-205-5p. Cell Death Dis 2020; 11:486. [PMID: 32587236 PMCID: PMC7316863 DOI: 10.1038/s41419-020-2689-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022]
Abstract
Pain, physical dysfunction, and mental disorders caused by bone nonunion bring great burden to patients. Bone mesenchymal stem cells (BMSCs) isolated from bone nonunion patients with poor proliferation and osteogenic ability are compared with that from normal bone-healing patients. Long noncoding RNAs (lncRNAs) are a class of RNAs that are more than 200 nucleotides in length, lack an open-reading frame encoding a protein, and have little or no protein-coding function, and could regulate gene expression, which is involved in the regulation of important life activities, such as growth, development, aging, and death at epigenetic, transcriptional, and post-transcriptional levels. In this study, we intended to investigate the difference of lncRNA expression between patients with nonunion and normal fracture healing. Our result found that lncRNA ENST00000563492 was downregulated in bone nonunion tissues. LncRNA ENST00000563492 promotes osteogenic differentiation of BMSCs through upregulating the expression of CDH11. On the other hand, LncRNA ENST0000563492 could improve the osteogenesis–angiogenesis coupling process through enhancing the expression of VEGF during osteogenic differentiation of BMSCs. LncRNA ENST00000563492 functions as a ceRNA for miR-205-5p that was targeting CDH11 and VEGF. LncRNA ENST00000563492 could promote the osteogenesis of BMSCs in vivo. Our result indicated that lncRNA ENST00000563492 may be a new target for bone nonunion.
Collapse
|
44
|
Volleman TNE, Schol J, Morita K, Sakai D, Watanabe M. Wnt3a and wnt5a as Potential Chondrogenic Stimulators for Nucleus Pulposus Cell Induction: A Comprehensive Review. Neurospine 2020; 17:19-35. [PMID: 32252152 PMCID: PMC7136098 DOI: 10.14245/ns.2040040.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/18/2020] [Indexed: 12/20/2022] Open
Abstract
Low back pain remains a highly prevalent pathology engendering a tremendous socioeconomic burden. Low back pain is generally associated with intervertebral disc (IVD) degeneration, a process involving the deterioration of nucleus pulpous (NP) cells and IVD matrix. Scientific interest has directed efforts to restoring cell numbers as a strategy to enable IVD regeneration. Currently, mesenchymal stromal cells (MSCs) are being explored as cell therapy agents, due to their easy accessibility and differentiation potential. For enhancement of MSCs, growth factor supplementation is commonly applied to induce differentiation towards a chondrogenic (NP) cell phenotype. The wnt signaling pathways play a crucial role in chondrogenesis, nonetheless, literature appears to present controversies with regard to wnt3a and wnt5a for the induction of NP cells, chondrocytes, and MSCs. This review aims to summarize the reporting on wnt3a/wnt5a mediated NP cell differentiation, and to elucidate the mechanisms involved in wnt3a and wnt5a mediated chondrogenesis for potential application as cell therapy supplements for IVD regeneration. Our review suggests that wnt3a, subsequently replaced with a chondrogenic stimulating growth factor, can enhance the chondrogenic potential of MSCs in vitro. Contrariwise, wnt5a is suggested to play a role in maintaining cell potency of differentiated NP or chondrogenic cells.
Collapse
Affiliation(s)
- Tibo Nico Emmie Volleman
- Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jordy Schol
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Kosuke Morita
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
45
|
Zhang W, Xia Y, Ling Y, Yang W, Dong ZX, Wang DA, Fan C. A Transcriptome Sequencing Study on Genome-Wide Gene Expression Differences of 3D Cultured Chondrocytes in Hydrogel Scaffolds with Different Gel Density. Macromol Biosci 2020; 20:e2000028. [PMID: 32187455 DOI: 10.1002/mabi.202000028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022]
Abstract
Hydrogel is considered as a promising cell delivery vehicle in cartilage tissue engineering, whose tunable microenvironments may influence the function and fate of encapsulated chondrocytes. Here, the transcriptomes of chondrocytes that are encapsulated and cultured in hydrogel constructs respectively made of 0.8% and 4% alginate solution are investigated. Differences in chondrocyte transcriptome are detected via RNA-sequencing from these two cultural conditions. The differentially expressed genes (DEGs) are reflected in extracellular matrix (ECM) secretion, cell cycle, proliferation, cartilage development, and so on. Significantly, the expression of DEGs associated with cartilage ECM and cell proliferation are upregulated in 0.8% constructs; whilst the expressions of DEGs involved in cell cycle and matrix degradation are upregulated in 4% constructs. Moreover, interestingly, the expressions of chondrocyte hypertrophy markers are upregulated in 0.8% constructs; while 4% constructs seemingly favor the long-term maintenance of chondrocyte phenotype. Taken together, this study confirms on transcriptomic level that gel density affects gene expression and phenotype of the encapsulated chondrocytes; therefore, it may provide guidance for future design and fabrication of cartilage tissue engineering scaffolds.
Collapse
Affiliation(s)
- Weiyuan Zhang
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yujun Xia
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Ling
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Wei Yang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, P. R. China
| | - Zuo-Xiang Dong
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, 266021, P. R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong SAR
| | - Changjiang Fan
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China.,Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, P. R. China
| |
Collapse
|
46
|
Dreher SI, Fischer J, Walker T, Diederichs S, Richter W. Significance of MEF2C and RUNX3 Regulation for Endochondral Differentiation of Human Mesenchymal Progenitor Cells. Front Cell Dev Biol 2020; 8:81. [PMID: 32195247 PMCID: PMC7064729 DOI: 10.3389/fcell.2020.00081] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Guiding progenitor cell development between chondral versus endochondral pathways is still an unachieved task of cartilage neogenesis, and human mesenchymal progenitor cell (MPC) chondrogenesis is considered as a valuable model to better understand hypertrophic development of chondrocytes. Transcription factors Runx2, Runx3, and Mef2c play prominent roles for chondrocyte hypertrophy during mouse development, but little is known on the importance of these key fate-determining factors for endochondral development of human MPCs. The aim of this study was to unravel the regulation of RUNX2, RUNX3, and MEF2C during MPC chondrogenesis, the pathways driving their expression, and the downstream hypertrophic targets affected by their regulation. RUNX2, RUNX3, and MEF2C gene expression was differentially regulated during chondrogenesis of MPCs, but remained low and unregulated when non-hypertrophic articular chondrocytes were differentiated under the same conditions. RUNX3 and MEF2C mRNA and protein levels rose in parallel to hypertrophic marker upregulation, but surprisingly, RUNX2 gene expression changed only by trend and RUNX2 protein remained undetectable. While RUNX3 expression was driven by TGF-β and BMP signaling, MEF2C responded to WNT-, BMP-, and Hedgehog-pathway inhibition. MEF2C but not RUNX3 levels correlated significantly with COL10A1, IHH, and IBSP gene expression when hypertrophy was attenuated. IBSP was a downstream target of RUNX3 and MEF2C but not RUNX2 in SAOS-2 cells, underlining the capacity of RUNX3 and MEF2C to stimulate osteogenic marker expression in human cells. Conclusively, RUNX3 and MEF2C appeared more important than RUNX2 for human endochondral MPC chondrogenesis. Pathways altering the speed of chondrogenesis (FGF, TGF-β, BMP) affected RUNX2 or RUNX3, while pathways changing hypertrophy (WNT, PTHrP/HH) regulated mainly MEF2C. Taken together, reduction of MEF2C levels is a new goal to shift human cartilage neogenesis toward the chondral pathway.
Collapse
Affiliation(s)
- Simon I Dreher
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jennifer Fischer
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Walker
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
47
|
Diederichs S, Klampfleuthner FAM, Moradi B, Richter W. Chondral Differentiation of Induced Pluripotent Stem Cells Without Progression Into the Endochondral Pathway. Front Cell Dev Biol 2019; 7:270. [PMID: 31737632 PMCID: PMC6838640 DOI: 10.3389/fcell.2019.00270] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
A major problem with chondrocytes derived in vitro from stem cells is undesired hypertrophic degeneration, to which articular chondrocytes (ACs) are resistant. As progenitors of all adult tissues, induced pluripotent stem cells (iPSCs) are in theory able to form stable articular cartilage. In vitro differentiation of iPSCs into chondrocytes with an AC-phenotype and resistance to hypertrophy has not been demonstrated so far. Here, we present a novel protocol that succeeded in deriving chondrocytes from human iPSCs without using pro-hypertrophic bone-morphogenetic-proteins. IPSC-chondrocytes had a high cartilage formation capacity and deposited two-fold more proteoglycans per cell than adult ACs. Importantly, cartilage engineered from iPSC-chondrocytes had similar marginal expression of hypertrophic markers (COL10A1, PTH1R, IBSP, ALPL mRNAs) like cartilage from ACs. Collagen X was barely detectable in iPSC-cartilage and 30-fold lower than in hypertrophic cartilage derived from mesenchymal stromal cells (MSCs). Moreover, alkaline phosphatase (ALP) activity remained at basal AC-like levels throughout iPSC chondrogenesis, in contrast to a well-known significant upregulation in hypertrophic MSCs. In line, iPSC-cartilage subjected to mineralizing conditions in vitro showed barely any mineralization, while MSC-derived hypertrophic cartilage mineralized strongly. Low expression of Indian hedgehog (IHH) like in ACs but rising BMP7 expression like in MSCs suggested that phenotype stability was linked to the hedgehog rather than the bone morphogenetic protein (BMP) pathway. Taken together, unlimited amounts of AC-like chondrocytes with a high proteoglycan production reminiscent of juvenile chondrocytes and resistance to hypertrophy and mineralization can now be produced from human iPSCs in vitro. This opens new strategies for cartilage regeneration, disease modeling and pharmacological studies.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Center for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felicia A M Klampfleuthner
- Research Center for Experimental Orthopaedics, Center for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Babak Moradi
- Clinic for Orthopaedics and Trauma Surgery, Center for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Center for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
48
|
Huang X, Chen Z, Shi W, Zhang R, Li L, Liu H, Wu L. TMF inhibits miR-29a/Wnt/β-catenin signaling through upregulating Foxo3a activity in osteoarthritis chondrocytes. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2009-2019. [PMID: 31354246 PMCID: PMC6590397 DOI: 10.2147/dddt.s209694] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022]
Abstract
Background: miR-29a, a downstream factor of Wnt/β-catenin signaling, promotes the activity of the Wnt/β-catenin signaling in a positive feedback loop. Our previous work showed that 5,7,3ʹ,4ʹ-tetramethoxyflavone (TMF), a major constituent from Murraya exotica L., exhibited chondroprotective activity by inhibiting the activity of Wnt/β-catenin signaling. Purpose: To investigate whether TMF showed the inhibitory effects on miR-29a/β-catenin signaling by up regulation of Foxo3a expression. Methods: Rat knee OA models were duplicated by using Hulth’s method. TMF (5 μg/mL and 20 μg/mL) was used for administration to cultured cells, which were isolated from the rat cartilages. Analysis of chondrocytes apoptosis, gene expression, and protein expression were conducted. In addition, miR-29a mimics and pcDNA3.1(+)-Foxo3a vector were used for transfection, luciferase reporter assay for detecting the activity of Wnt/β-catenin signaling, and co-immunoprecipitation for determining proteins interaction. Results: TMF down regulated miR-29a/β-catenin signaling activity and cleaved caspase-3 expression and up regulated Foxo3a expression in OA rat cartilages. In vitro, miR-29a mimics down regulated the expression of Foxo3a and up regulated the activity of Wnt/β-catenin signaling and cleaved caspase-3 expression. TMF ameliorated miR-29a/β-catenin-induced chondrocytes apoptosis by up regulation of Foxo3a expression. Conclusion: TMF exhibited chondroprotective activity by up regulating Foxo3a expression and subsequently inhibiting miR-29a/Wnt/β-catenin signaling activity.
Collapse
Affiliation(s)
- Xianhua Huang
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| | - Weimei Shi
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| | - Rui Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| | - Linfu Li
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| | - Hai Liu
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, People's Republic of China
| |
Collapse
|