1
|
Cometa S, Busto F, Scalia AC, Castellaneta A, Gentile P, Cochis A, Manfredi M, Borrini V, Rimondini L, De Giglio E. Effectiveness of gellan gum scaffolds loaded with Boswellia serrata extract for in-situ modulation of pro-inflammatory pathways affecting cartilage healing. Int J Biol Macromol 2024; 277:134079. [PMID: 39038574 DOI: 10.1016/j.ijbiomac.2024.134079] [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: 11/06/2023] [Revised: 05/09/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
In this study, we developed a composite hydrogel based on Gellan gum containing Boswellia serrata extract (BSE). BSE was either incorporated directly or loaded into an MgAl-layered double hydroxide (LDH) clay to create a multifunctional cartilage substitute. This composite was designed to provide anti-inflammatory properties while enhancing chondrogenesis. Additionally, LDH was exploited to facilitate the loading of hydrophobic BSE components and to improve the hydrogel's mechanical properties. A calcination process was also adopted on LDH to increase BSE loading. Physicochemical and mechanical characterizations were performed by spectroscopic (XPS and FTIR), thermogravimetric, rheological, compression test, weight loss and morphological (SEM) investigations. RPLC-ESI-FTMS was employed to investigate the boswellic acids release in simulated synovial fluid. The composites were cytocompatible and capable of supporting the mesenchymal stem cells (hMSC) growth in a 3D-conformation. Loading BSE resulted in the modulation of the pro-inflammatory cascade by down-regulating COX2, PGE2 and IL1β. Chondrogenesis studies demonstrated an enhanced differentiation, leading to the up-regulation of COL 2 and ACAN. This effect was attributed to the efficacy of BSE in reducing the inflammation through PGE2 down-regulation and IL10 up-regulation. Proteomics studies confirmed gene expression findings by revealing an anti-inflammatory protein signature during chondrogenesis of the cells cultivated onto loaded specimens. Concluding, BSE-loaded composites hold promise as a tool for the in-situ modulation of the inflammatory cascade while preserving cartilage healing.
Collapse
Affiliation(s)
| | - Francesco Busto
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy; INSTM, National Consortium of Materials Science and Technology, Via G. Giusti 9, 50121 Florence, Italy.
| | - Alessandro C Scalia
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Andrea Castellaneta
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
| | - Piergiorgio Gentile
- Newcastle University, School of Engineering, Claremont Road, NE1 7RU Newcastle upon Tyne, United Kingdom.
| | - Andrea Cochis
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Vittoria Borrini
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Lia Rimondini
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Elvira De Giglio
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy; INSTM, National Consortium of Materials Science and Technology, Via G. Giusti 9, 50121 Florence, Italy.
| |
Collapse
|
2
|
Volk-Draper L, Athaiya S, Espinosa Gonzalez M, Bhattarai N, Wilber A, Ran S. Tumor microenvironment restricts IL-10 induced multipotent progenitors to myeloid-lymphatic phenotype. PLoS One 2024; 19:e0298465. [PMID: 38640116 PMCID: PMC11029653 DOI: 10.1371/journal.pone.0298465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/24/2024] [Indexed: 04/21/2024] Open
Abstract
Lymphangiogenesis is induced by local pro-lymphatic growth factors and bone marrow (BM)-derived myeloid-lymphatic endothelial cell progenitors (M-LECP). We previously showed that M-LECP play a significant role in lymphangiogenesis and lymph node metastasis in clinical breast cancer (BC) and experimental BC models. We also showed that differentiation of mouse and human M-LECP can be induced through sequential activation of colony stimulating factor-1 (CSF-1) and Toll-like receptor-4 (TLR4) pathways. This treatment activates the autocrine interleukin-10 (IL-10) pathway that, in turn, induces myeloid immunosuppressive M2 phenotype along with lymphatic-specific proteins. Because IL-10 is implicated in differentiation of numerous lineages, we sought to determine whether this pathway specifically promotes the lymphatic phenotype or multipotent progenitors that can give rise to M-LECP among other lineages. Analyses of BM cells activated either by CSF-1/TLR4 ligands in vitro or orthotopic breast tumors in vivo showed expansion of stem/progenitor population and coincident upregulation of markers for at least four lineages including M2-macrophage, lymphatic endothelial, erythroid, and T-cells. Induction of cell plasticity and multipotency was IL-10 dependent as indicated by significant reduction of stem cell markers and those for multiple lineages in differentiated cells treated with anti-IL-10 receptor (IL-10R) antibody or derived from IL-10R knockout mice. However, multipotent CD11b+/Lyve-1+/Ter-119+/CD3e+ progenitors detected in BM appeared to split into a predominant myeloid-lymphatic fraction and minor subsets expressing erythroid and T-cell markers upon establishing tumor residence. Each sub-population was detected at a distinct intratumoral site. This study provides direct evidence for differences in maturation status between the BM progenitors and those reaching tumor destination. The study results suggest preferential tumor bias towards expansion of myeloid-lymphatic cells while underscoring the role of IL-10 in early BM production of multipotent progenitors that give rise to both hematopoietic and endothelial lineages.
Collapse
Affiliation(s)
- Lisa Volk-Draper
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Shaswati Athaiya
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Maria Espinosa Gonzalez
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Nihit Bhattarai
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Sophia Ran
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| |
Collapse
|
3
|
Wisniewska E, Laue D, Spinnen J, Kuhrt L, Kohl B, Bußmann P, Meier C, Schulze-Tanzil G, Ertel W, Jagielski M. Infrapatellar Fat Pad Modulates Osteoarthritis-Associated Cytokine and MMP Expression in Human Articular Chondrocytes. Cells 2023; 12:2850. [PMID: 38132170 PMCID: PMC10741519 DOI: 10.3390/cells12242850] [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: 11/15/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Osteoarthritis (OA) most frequently affects the knee joint and is associated with an elevated expression of cytokines and extracellular cartilage matrix (ECM), degrading enzymes such as matrix metalloproteinases (MMPs). Differences in gene expression of the intra-articularly located infrapatellar fat pad (IPFP) and other fatty tissue suggest its autonomous function, yet its role in OA pathogenesis remains unknown. Human IPFPs and articular cartilage were collected from OA patients undergoing total knee arthroplasty, and biopsies from the IPFP of healthy patients harvested during knee arthroscopy served as controls (CO). Isolated chondrocytes were co-cultured with either osteoarthritic (OA) or CO-IPFPs in a transwell system. Chondrocyte expression of MMP1, -3, -13, type 1 and 2 collagens, interleukin IL1β, IL6, IL10, and tumor necrosis factor TNFα was analyzed by RTD-PCR at day 0 and day 2, and TNFα secretion was analyzed by ELISA. The cytokine release in IPFPs was assessed by an array. Results: Both IPFPs (CO, OA) significantly reduced the expression of type 2 collagen and TNFα in chondrocytes. On the other hand, only CO-IPFP suppressed the expression of type 1 collagen and significantly induced the MMP13 expression. On the contrary, IL1β and IL6 were significantly induced when exposed to OA-IPFP. Conclusions: The partial loss of the suppressive effect on type 1 collagen gene expression found for OA-IPFP shows the pathological remodeling and dedifferentiation potential of the OA-IPFP on the chondrocytes. However, the significant suppression of TNFα implies that the OA- and CO-IPFP could also exhibit a protective role in the knee joint, preventing the progress of inflammation.
Collapse
Affiliation(s)
- Ewa Wisniewska
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Dominik Laue
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Jacob Spinnen
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Leonard Kuhrt
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Benjamin Kohl
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Patricia Bußmann
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Carola Meier
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University (PMU), Prof.-Ernst Nathan Strasse 1, 90419 Nuremberg, Germany;
| | - Wolfgang Ertel
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| | - Michal Jagielski
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; (E.W.); (D.L.); (J.S.); (L.K.); (B.K.); (P.B.); (C.M.); (W.E.)
| |
Collapse
|
4
|
Guo J, Ye W, Wu X, Huang H, Li B, Sun Z, Ren Z, Yang Z. TNF-α activates RELA expression via TNFRSF1B to upregulate OPA1 expression and inhibit chondrogenic differentiation of human adipose stem cells. J Orthop Surg Res 2023; 18:430. [PMID: 37312126 DOI: 10.1186/s13018-023-03846-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Tumor necrosis factor-alpha (TNF-α), one of the pro-inflammatory cytokines mediating the local inflammatory process in joints, inhibits cartilage formation and has a detrimental effect on stem cell-based cartilage regeneration for the treatment of osteoarthritis (OA). However, the mechanisms behind this inhibitory effect are still poorly understood. Mitochondrial morphological changes mediated by mitochondrial fusion and fission are highly plastic, are quite sensitive to environmental stimuli and play a crucial role in maintaining cell structure and function. In our study, chondrogenic differentiated human adipose stem cells (hADSCs) were exposed to TNF-α and the effect of TNF-α on the ability of hADSCs to chondrogenic differentiate and on mitochondrial fusion and fission was observed and analyzed. The aim was to investigate the role and mechanisms of mitochondrial fusion and fission regulation in the chondrogenic differentiation of hADSCs under normal conditions and under exposure to TNF-α. METHODS We used flow cytometry to identify hADSCs immunophenotypes CD29, CD44, CD34, CD45, and HLA-DR. Alcian blue staining and Sirius red staining were used to observe the formation of proteoglycans and collagen during the chondrogenic differentiation of hADSCs, respectively. The mRNA and protein expression levels of the cartilage formation marker SOX9, type II collagen (COL2A1), and Aggrecan were measured by real-time fluorescent quantitative PCR (RT-qPCR) and western blot, respectively. The fluorescent probes MitoTracker® Red CMXRos and JC-1 were used to visualize mitochondria morphology and detect mitochondrial membrane electricity (MMP). Affymetrix PrimeView™ chips were used for gene expression profiling. RESULTS The results showed that the chondrogenic differentiation of hADSCs was inhibited in the presence of TNF-α that optic atrophy 1 (OPA1) expression was significantly upregulated and mitochondria were prolonged and interconnected during this process. Gene microarray and RT-qPCR data showed that the presence of TNF-α led to increased expression of TNFα receptor 2 (TNFRSF1B) and RELA during chondrogenic differentiation of hADSCs. CONCLUSIONS TNF-α inhibits chondrogenic differentiation of human adipose stem cells by activating RELA expression through TNFRSF1B upregulating OPA1 expression thereby increasing mitochondrial fusion.
Collapse
Affiliation(s)
- Jiajia Guo
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Wang Ye
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xinglin Wu
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Haifeng Huang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Bo Li
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Zeyu Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Zhijing Ren
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| | - Zhen Yang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
5
|
Lamparelli EP, Ciardulli MC, Giudice V, Scala P, Vitolo R, Dale TP, Selleri C, Forsyth NR, Maffulli N, Della Porta G. 3D in-vitro cultures of human bone marrow and Wharton’s jelly derived mesenchymal stromal cells show high chondrogenic potential. Front Bioeng Biotechnol 2022; 10:986310. [PMID: 36225603 PMCID: PMC9549977 DOI: 10.3389/fbioe.2022.986310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
In this study, chondrogenic potentials of 3D high-density cultures of Bone Marrow (BM) and Wharton’s Jelly (WJ)-derived mesenchymal stromal cells (MSCs) was investigated by chondrogenesis- and cytokine-related gene expression over a 16-day culture period supplemented with human transforming growth factor (hTGF)-β1 at 10 ng/ml. In BM-MSC 3D models, a marked upregulation of chondrogenesis-related genes, such as SOX9, COL2A1, and ACAN (all p < 0.05) and formation of spherical pellets with structured type II collagen fibers were observed. Similarly, WJ-based high-density culture appeared higher in size and more regular in shape, with a significant overexpression of COL2A1 and ACAN (all p < 0.05) at day 16. Moreover, a similar upregulation trend was documented for IL-6 and IL-10 expression in both BM and WJ 3D systems. In conclusion, MSC-based high-density cultures can be considered a promising in vitro model of cartilage regeneration and tissue engineering. Moreover, our data support the use of WJ-MSCs as a valid alternative for chondrogenic commitment of stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Erwin Pavel Lamparelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | | | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, SA, Italy
| | - Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | - Rosa Vitolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | - Tina Patricia Dale
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire, United Kingdom
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, SA, Italy
| | - Nicholas Robert Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire, United Kingdom
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
- Centre for Sport and Exercise Medicine, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
- Research Centre for Biomaterials BIONAM, Università di Salerno, Fisciano, SA, Italy
- *Correspondence: Giovanna Della Porta,
| |
Collapse
|
6
|
Bhogoju S, Khan S, Subramanian A. Continuous Low-Intensity Ultrasound Preserves Chondrogenesis of Mesenchymal Stromal Cells in the Presence of Cytokines by Inhibiting NFκB Activation. Biomolecules 2022; 12:434. [PMID: 35327626 PMCID: PMC8946190 DOI: 10.3390/biom12030434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Proinflammatory joint environment, coupled with impeded chondrogenic differentiation of mesenchymal stromal cells (MSCs), led to inferior cartilage repair outcomes. Nuclear translocation of phosphorylated-NFκB downregulates SOX9 and hinders the chondrogenesis of MSCs. Strategies that minimize the deleterious effects of NFκB, while promoting MSC chondrogenesis, are of interest. This study establishes the ability of continuous low-intensity ultrasound (cLIUS) to preserve MSC chondrogenesis in a proinflammatory environment. MSCs were seeded in alginate:collagen hydrogels and cultured for 21 days in an ultrasound-assisted bioreactor (5.0 MHz, 2.5 Vpp; 4 applications/day) in the presence of IL1β and evaluated by qRT-PCR and immunofluorescence. The differential expression of markers associated with the NFκB pathway was assessed upon a single exposure of cLIUS and assayed by Western blotting, qRT-PCR, and immunofluorescence. Mitochondrial potential was evaluated by tetramethylrhodamine methyl ester (TMRM) assay. The chondroinductive potential of cLIUS was noted by the increased expression of SOX9 and COLII. cLIUS extended its chondroprotective effects by stabilizing the NFκB complex in the cytoplasm via engaging the IκBα feedback mechanism, thus preventing its nuclear translocation. cLIUS acted as a mitochondrial protective agent by restoring the mitochondrial potential and the mitochondrial mRNA expression in a proinflammatory environment. Altogether, our results demonstrated the potential of cLIUS for cartilage repair and regeneration under proinflammatory conditions.
Collapse
Affiliation(s)
| | | | - Anuradha Subramanian
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL 35899, USA; (S.B.); (S.K.)
| |
Collapse
|
7
|
Genç D, Sezer Kürkçü M, Yiğittürk G, Günaydın B, Elbe H, Aladağ A, Çöl B, Tarhan EF. Synovial fluid niche promoted differentiation of dental follicle mesenchymal stem cells toward chondrogenesis in rheumatoid arthritis. Arch Rheumatol 2021; 37:94-109. [PMID: 35949879 PMCID: PMC9326389 DOI: 10.46497/archrheumatol.2022.8891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022] Open
Abstract
Objectives
In this study, we aimed to investigate the differentiation potential of dental follicle mesenchymal stem cells (MSCs) in the synovial fluid (SF) niche of early-onset or end-stage rheumatoid arthritis (RA). Patients and methods
Between May 2020 and January 2021, six patients (1 male, 5 females; mean age: 57.5±11.2 years; range, 49 to 65 years) who were diagnosed with RA with the indication of SF aspiration were included in the study. The third passage dental follicle stem cells (DFSCs) were cocultured with fresh SF samples of end-stage or early-onset RA patients in micromass culture system for 21 days. SF samples were analyzed for secreted cytokines. Chondrogenic markers (CD49e, CD49f) were analyzed in DFSCs, gene expression analysis was performed for the expressions of Col I, Col II, Aggrecan and Sox-9, and histochemical analysis was performed by staining three-dimensional pellets with anti-collagen II antibody. The neutralization assay was performed with anti-interleukin (IL)-6, anti-interferon-gamma (IFN-g), and anti-IL-1beta(b). Results
The high levels of IL-1b and IL-6 were observed in end-stage RA patients’ SF samples compared to the early-onset patients (p<0.05). The CD49e and CD49f expressions in DFSCs were significantly higher in the SF samples of end-stage RA patients (p<0.05). Also, the Col II, Sox-9 and Aggrecan messenger ribonucleic acid (mRNA) expressions increased in the DFSCs, when cultured with end-stage RA patients’ SF samples (p<0.01). Collagen-II expression in histochemical analysis of micromass pellets was higher in the DFSCs cultured with end-stage RA patients’ SF samples. The neutralization of IL-6 significantly decreased the CD49e and CD49f expressions (p<0.05). Conclusion
The high levels of IL-6 in SF niche of end-stage RA patients were found to differentiate DFSCs toward chondrogenesis. Based on these findings, DFSCs can be used as a new cell-based treatment in RA patients for the cartilage damage.
Collapse
Affiliation(s)
- Deniz Genç
- Department of Pediatric Health and Diseases Nursing, Muğla Sıtkı Koçman University, Faculty of Health Sciences, Muğla, Turkey
| | - Merve Sezer Kürkçü
- Muğla Sıtkı Koçman University, Research Laboratories Center, Muğla, Turkey
| | - Gürkan Yiğittürk
- Department of Histology and Embryology, Muğla Sıtkı Koçman University, Faculty of Medicine, Muğla, Turkey
| | - Burcu Günaydın
- Department of Histology and Embryology, Muğla Sıtkı Koçman University, Institute of Health Sciences, Muğla, Turkey
| | - Hülya Elbe
- Department of Histology and Embryology, Muğla Sıtkı Koçman University, Faculty of Medicine, Muğla, Turkey
| | - Akın Aladağ
- Muğla Sıtkı Koçman University, Faculty of Dentistry, Muğla, Turkey
| | - Bekir Çöl
- Department of Biology, Muğla Sıtkı Koçman University, Faculty of Science, Muğla, Turkey
| | - Emine Figen Tarhan
- Department of Rheumatology, Muğla Sıtkı Koçman University, Faculty of Medicine, Muğla, Turkey
| |
Collapse
|
8
|
Huynh PD, Vu NB, To XHV, Le TM. Culture and Differentiation of Human Umbilical Cord-Derived Mesenchymal Stem Cells on Growth Factor-Rich Fibrin Scaffolds to Produce Engineered Cartilages. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021:193-208. [PMID: 34739721 DOI: 10.1007/5584_2021_670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION After injuries, the cartilage healing capacity is limited owing to its nature as a particular connective tissue without blood vessels, lymphatics, or nerves. The creation of artificial cartilage tissue mimics the biological properties of native cartilage and can reduce the need for donated tissue. Fibrin is a type of biodegradable scaffold that has great potential in tissue engineering applications. It can become good material for cell adhesion and proliferation in vitro. Therefore, this study aimed to create a cartilage tissue in vitro using umbilical cord-derived mesenchymal stem cells (UCMSC) and growth factor-rich fibrin (GRF) scaffolds. METHODS UCMSCs were isolated and expanded, and platelet-rich plasma (PRP) preparations were performed following previously published protocols. PRP was activated (aPRP) by a 0.45-μm syringe filter to release growth factors inside the platelets. Each 2.105 of the UCMSCs were suspended in 2 ml of aPRP to make the mixture of MSC and PRP (MSC-PRP). Then, Ca2+ solution was added to this mixture to produce the fibril scaffold with UCMSCs inside. UCMSCs' adhesion and proliferation inside the scaffold were evaluated by observation under inverted microscopy, H-E staining, MTT assays, and scanning electron microscopy (SEM). The fibril structure containing UCMSCs was cultured, and chondrogenesis was induced using commercial chondrogenesis media for 21 days (iMSC-GRF). The differentiation in efficacy toward cartilage was evaluated based on the accumulation of aggrecan (acan), glycosaminoglycans (GAGs), and collagen type II (Col II). RESULTS The results showed that we successfully created a cartilage tissue with some characteristics that mimic the properties of natural cartilage. The engineered cartilage tissue was positive with some cartilage protein, such as acan, GAG, and Coll II. In vitro cartilage presented some natural chondrocyte-like cells. The artificial cartilage tissue was positive for CD14, CD34, CD90, CD105, and HLA-DR and negative for CD44, CD45, and CD73. CONCLUSION These results showed that using UCMSCs and growth factor-rich fibril from platelet-rich plasma was feasible to produce engineered cartilage tissue for further experiments or clinical usage.
Collapse
Affiliation(s)
- Phat Duc Huynh
- Laboratory of Stem Cell Research and Application, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ngoc Bich Vu
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
- Stem Cell Institute, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| | - Xuan Hoang-Viet To
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Stem Cell Institute, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thuan Minh Le
- Laboratory of Stem Cell Research and Application, University of Science Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
9
|
Jauković A, Kukolj T, Obradović H, Okić-Đorđević I, Mojsilović S, Bugarski D. Inflammatory niche: Mesenchymal stromal cell priming by soluble mediators. World J Stem Cells 2020; 12:922-937. [PMID: 33033555 PMCID: PMC7524701 DOI: 10.4252/wjsc.v12.i9.922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/13/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are adult stem cells of stromal origin that possess self-renewal capacity and the ability to differentiate into multiple mesodermal cell lineages. They play a critical role in tissue homeostasis and wound healing, as well as in regulating the inflammatory microenvironment through interactions with immune cells. Hence, MSCs have garnered great attention as promising candidates for tissue regeneration and cell therapy. Because the inflammatory niche plays a key role in triggering the reparative and immunomodulatory functions of MSCs, priming of MSCs with bioactive molecules has been proposed as a way to foster the therapeutic potential of these cells. In this paper, we review how soluble mediators of the inflammatory niche (cytokines and alarmins) influence the regenerative and immunomodulatory capacity of MSCs, highlighting the major advantages and concerns regarding the therapeutic potential of these inflammatory primed MSCs. The data summarized in this review may provide a significant starting point for future research on priming MSCs and establishing standardized methods for the application of preconditioned MSCs in cell therapy.
Collapse
Affiliation(s)
- Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Hristina Obradović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Ivana Okić-Đorđević
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Slavko Mojsilović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| |
Collapse
|
10
|
Farivar S, Ramezankhani R, Mohajerani E, Ghazimoradi MH, Shiari R. Gene Expression Analysis of Chondrogenic Markers in Hair Follicle Dermal Papillae Cells Under the Effect of Laser Photobiomodulation and the Synovial Fluid. J Lasers Med Sci 2019; 10:171-178. [PMID: 31749941 DOI: 10.15171/jlms.2019.27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Introduction: Regarding the limited ability of the damaged cartilage cells to self-renew, which is due to their specific tissue structure, subtle damages can usually cause diseases such as osteoarthritis. In this work, using laser photobiomodulation and an interesting source of growth factors cocktail called the synovial fluid, we analyzed the chondrogenic marker genes in treated hair follicle dermal papilla cells as an accessible source of cells with relatively high differentiation potential. Methods: Dermal papilla cells were isolated from rat whisker hair follicle (Rattus norvegicus) and established cell cultures were treated with a laser (gallium aluminum arsenide diode Laser (λ=780 nm, 30 mW) at 5 J/cm2 ), the synovial fluid, and a combination of both. After 1, 4, 7, and 14 days, the morphological changes were evaluated and the expression levels of four chondrocyte marker genes (Col2a1, Sox-9, Col10a1, and Runx-2) were assessed by the quantitative real-time polymerase chain reaction. Results: It was monitored that treating cells with laser irradiation can accelerate the rate of proliferation of cells. The morphology of the cells treated with the synovial fluid altered considerably as in the fourth day they surprisingly looked like cultured articular chondrocytes. The gene expression analysis showed that all genes were up-regulated until the day 14 following the treatments although not equally in all the cell groups. Moreover, the cell groups treated with both irradiation and the synovial fluid had a significantly augmented expression in gene markers. Conclusion: Based on the gene expression levels and the morphological changes, we concluded that the synovial fluid can have the potential to make the dermal papilla cells to most likely mimic the chondrogenic and/or osteogenic differentiation, although this process seems to be augmented by the irradiation of the low-level laser.
Collapse
Affiliation(s)
- Shirin Farivar
- Department of Molecular and Cell Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, General Campus, Tehran, Iran
| | - Roya Ramezankhani
- Department of Molecular and Cell Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, General Campus, Tehran, Iran
| | - Ezedin Mohajerani
- Laser and Plasma Research Institute, Shahid Beheshti University, General Campus, Tehran, Iran
| | - Mohammad Hosein Ghazimoradi
- Department of Molecular and Cell Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, General Campus, Tehran, Iran
| | - Reza Shiari
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Trevisol TC, Langbehn RK, Battiston S, Immich APS. Nonwoven membranes for tissue engineering: an overview of cartilage, epithelium, and bone regeneration. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 30:1026-1049. [PMID: 31106705 DOI: 10.1080/09205063.2019.1620592] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Scaffold-type biomaterials are crucial for application in tissue engineering. Among them, the use of a nonwoven scaffold has grown in recent years and has been widely investigated for the regeneration of different types of tissues. Several polymers, whether they are synthetic, biopolymers or both, have been used to produce a scaffold that can mimic the natural tissue to which it will be applied to. The scaffolds used in tissue engineering must be biocompatible and allow cell adhesion and proliferation to be applied in tissue engineering. In addition, the scaffolds should maintain the mechanical properties and architecture of the desired tissue. Nonwoven fabrics have produced good results and are more extensively applied for the regeneration of cartilage, epithelial and bone tissues. Recent advances in tissue engineering have shown promising results, however, no ideal material or standardization parameters and characteristics of the materials were obtained. The present review provides an overview of the application of nonwoven scaffolds, including the main results obtained regarding the properties of the biomaterials and their applications in vitro and in vivo, focusing on the cartilaginous, the epithelium, and bone tissue regeneration.
Collapse
Affiliation(s)
- Thalles Canton Trevisol
- a Department of Chemical and Food Engineering, Technological Center , Federal University of Santa Catarina , Florianópolis , Brazil
| | - Rayane Kunert Langbehn
- a Department of Chemical and Food Engineering, Technological Center , Federal University of Santa Catarina , Florianópolis , Brazil
| | - Suellen Battiston
- a Department of Chemical and Food Engineering, Technological Center , Federal University of Santa Catarina , Florianópolis , Brazil
| | - Ana Paula Serafini Immich
- b Department of Textile Engineering, Blumenau campus , Federal University of Santa Catarina , Blumenau , Brazil
| |
Collapse
|
12
|
Zhang T, Yao Y. Effects of inflammatory cytokines on bone/cartilage repair. J Cell Biochem 2019; 120:6841-6850. [PMID: 30335899 DOI: 10.1002/jcb.27953] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/02/2018] [Indexed: 01/24/2023]
Abstract
Many inflammatory factors can affect cell behaviors and work as a form of inter-regulatory networks through the inflammatory pathway. Inflammatory cytokines are critical for triggering bone regeneration after fracture or bone injury. Also, inflammatory cytokines play an important role in cartilage repair. The synergistic or antagonistic effects of both proinflammatory and anti-inflammatory cytokines have a great influence on fracture healing. This review discusses key inflammatory cytokines and signaling pathways involved in bone or cartilage repair.
Collapse
Affiliation(s)
- Tingshuai Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
13
|
Liu J, Jiang T, Li C, Wu Y, He M, Zhao J, Zheng L, Zhang X. Bioconjugated Carbon Dots for Delivery of siTnfα to Enhance Chondrogenesis of Mesenchymal Stem Cells by Suppression of Inflammation. Stem Cells Transl Med 2019; 8:724-736. [PMID: 30919586 PMCID: PMC6591550 DOI: 10.1002/sctm.18-0289] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/20/2019] [Indexed: 01/05/2023] Open
Abstract
Although a promising strategy, the mesenchymal stem cell (MSC)‐based therapy of cartilage defects is sometimes accompanied with chronic inflammation during the remodeling status, which may hinder cartilage regeneration. During this process, the inflammatory cytokine tumor necrosis factor α (TNFα) plays an important role and may be a potential target. In this study, we investigated the effect of Tnfα RNA interference by introducing a functional and highly safe carbon dot (CD)‐SMCC nanovector synthesized by bioconjugation of CDs with a protein crosslinker, sulfosuccinimidyl‐4‐(N‐maleimidomethyl) cyclohexane‐1‐carboxylate (sulfo‐SMCC), as the vehicle of the silenced TNFα (siTnfα) on chondrogenesis of MSCs. The results showed that CD‐SMCC displayed intense fluorescence with well‐dispersed and positively charged properties, which favored effective binding and delivering of siTnfα into the MSCs. CD‐SMCC‐siTnfα nanoformula also exhibited considerably high transfection efficiency and nearly no cytotoxicity, which is preferred over commercial polyethyleneimine. Interference of Tnfα by CD‐SMCC‐siTnfα markedly promoted the chondrogenesis of MSCs, as indicated by upregulating cartilage‐specific markers. Furthermore, in vivo exploration indicated that CD‐SMCC‐siTnfα transfected MSCs accelerated cartilage regeneration. In conclusion, this study demonstrated that in combination with the novel CD‐SMCC nanovector, targeting Tnfα may facilitate stem cell‐based therapy of cartilage defects. stem cells translational medicine2019;8:724&736
Collapse
Affiliation(s)
- Jianwei Liu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China.,Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China.,Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Chun Li
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Yang Wu
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Maolin He
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China.,Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China.,Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People's Republic of China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
14
|
New Approach for Differentiation of Bone Marrow Mesenchymal Stem Cells Toward Chondrocyte Cells With Overexpression of MicroRNA-140. ASAIO J 2018; 64:662-672. [DOI: 10.1097/mat.0000000000000688] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
15
|
Žigon-Branc S, Barlič A, Knežević M, Jeras M, Vunjak-Novakovic G. Testing the potency of anti-TNF-α and anti-IL-1β drugs using spheroid cultures of human osteoarthritic chondrocytes and donor-matched chondrogenically differentiated mesenchymal stem cells. Biotechnol Prog 2018; 34:1045-1058. [PMID: 29536646 PMCID: PMC6138577 DOI: 10.1002/btpr.2629] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 03/07/2018] [Indexed: 12/31/2022]
Abstract
Inflammation plays a major role in progression of rheumatoid arthritis, a disease treated with antagonists of tumor necrosis factor-alpha (TNF-α) and interleukin 1β (IL-1β). New in vitro testing systems are needed to evaluate efficacies of new anti-inflammatory biological drugs, ideally in a patient-specific manner. To address this need, we studied microspheroids containing 10,000 human osteoarthritic primary chondrocytes (OACs) or chondrogenically differentiated mesenchymal stem cells (MSCs), obtained from three donors. Hypothesizing that this system can recapitulate clinically observed effects of anti-inflammatory drugs, spheroids were exposed to TNF-α, IL-1β, or to supernatant containing secretome from activated macrophages (MCM). The anti-inflammatory efficacies of anti-TNF-α biologicals adalimumab, infliximab, and etanercept, and the anti-IL-1β agent anakinra were assessed in short-term microspheroid and long-term macrospheroid cultures (100,000 OACs). While gene and protein expressions were evaluated in microspheroids, diameters, amounts of DNA, glycosaminoglycans, and hydroxiproline were measured in macrospheroids. The tested drugs significantly decreased the inflammation induced by TNF-α or IL-1β. The differences in potency of anti-TNF-α biologicals at 24 h and 3 weeks after their addition to inflamed spheroids were comparable, showing high predictability of short-term cultures. Moreover, the data obtained with microspheroids grown from OACs and chondrogenically differentiated MSCs were comparable, suggesting that MSCs could be used for this type of in vitro testing. We propose that in vitro gene expression measured after the first 24 h in cultures of chondrogenically differentiated MSCs can be used to determine the functionality of anti-TNF-α drugs in personalized and preclinical studies. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1045-1058, 2018.
Collapse
Affiliation(s)
- Sara Žigon-Branc
- Department of Biomedical Engineering, Columbia University, New York NY, USA
- Educell Cell Therapy Service Ltd, Trzin, Slovenia
| | | | | | - Matjaž Jeras
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Slovenia
- Celica Biomedical Ltd., Ljubljana, Slovenia
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York NY, USA
- Department of Medicine, Columbia University, New York NY, USA
| |
Collapse
|
16
|
Enhanced chondrogenesis differentiation of human induced pluripotent stem cells by MicroRNA-140 and transforming growth factor beta 3 (TGFβ3). Biologicals 2018; 52:30-36. [DOI: 10.1016/j.biologicals.2018.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/07/2017] [Accepted: 01/26/2018] [Indexed: 12/17/2022] Open
|
17
|
Chondrogenic potential of IL-10 in mechanically injured cartilage and cellularized collagen ACI grafts. Osteoarthritis Cartilage 2018; 26:264-275. [PMID: 29169959 DOI: 10.1016/j.joca.2017.11.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 11/07/2017] [Accepted: 11/11/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The application of adjunctive mediators in Autologous chondrocyte implantation (ACI) techniques might be useful for improving the dedifferentiated chondrocyte phenotype, to support neocartilage formation and inhibit post-traumatic cartilage destruction. In this study we examined if (a) interleukin 10 treatment can cause chondrogenic phenotype stabilization and matrix preservation in mechanically injured cartilage and if (b) IL-10 can promote chondrogenesis in a clinically applied collagen scaffold for ACI treatment. MATERIALS AND METHODS For (a) bovine articular cartilage was harvested, subjected to an axial unconfined injury and treated with bovine IL-10 (1-10,000 pg/ng/ml). For (b) a post-operatively remaining ACI graft was treated with human IL-10. Expression levels of type I/II/X collagen, SOX9 and aggrecan were measured by qPCR (a,b). After 3 weeks cell death was analyzed (nuclear blebbing and TUNEL assay) and matrix composition was determined by GAG measurements and immunohistochemistry (aggrecan, type I/II collagen, hyaluronic acid). STATISTICS One way ANOVA analysis with Bonferroni's correction. RESULTS (a) IL-10 stabilized the chondrogenic phenotype after injurious compression and preserved matrix integrity. This was indicated by elevated expression of chondrogenic markers COL2A1, ACAN, SOX9, while COL1A1 and COL10A1 were reduced. An increased GAG content paralleled this and histological staining of type 2 collagen, aggrecan and toluidine blue were enhanced after 3 weeks. (b) IL-10 [100 pg/ml] improved the chondrogenic differentiation of human chondrocytes, which was accompanied by cartilaginous matrix formation after 3 weeks of incubation. CONCLUSION Interleukin-10 is a versatile adjuvant candidate to control the post-injurious environment in cartilage defects and promote chondrogenesis in ACI grafts.
Collapse
|
18
|
Schwarz S, Mrosewski I, Silawal S, Schulze-Tanzil G. The interrelation of osteoarthritis and diabetes mellitus: considering the potential role of interleukin-10 and in vitro models for further analysis. Inflamm Res 2017; 67:285-300. [PMID: 29196771 DOI: 10.1007/s00011-017-1121-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 11/12/2017] [Accepted: 11/24/2017] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Today, not only the existence of an interrelation between obesity/adipositas and osteoarthritis (OA) but also the association of OA and diabetes mellitus (DM) are widely recognized. Nevertheless, shared influence factors facilitating OA development in DM patients still remain speculative up until now. To supplement the analysis of clinical data, appropriate in vitro models could help to identify shared pathogenetic pathways. Informative in vitro studies could later be complemented by in vivo data obtained from suitable animal models. MATERIALS AND METHODS Therefore, this detailed review of available literature was undertaken to discuss and compare the results of currently published in vitro studies focusing on the interrelation between OA, the metabolic syndrome and DM and to propose models to further study the molecular pathways. RESULTS The survey of literature presented here supports the hypothesis that the pathogenesis of OA in DM is based on imbalanced molecular pathways with a putative crucial role of antiinflammatory cytokines such as IL-10. CONCLUSION Future development of versatile micro-scaled in vitro models such as combining DM and OA on chip could allow the identification of common pathogenetic pathways and might help to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Silke Schwarz
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany.,Institute of Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Ingo Mrosewski
- MVZ Limbach Laboratories, Aroser Allee 84, 13407, Berlin, Germany
| | - Sandeep Silawal
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany.,Institute of Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Gundula Schulze-Tanzil
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany. .,Institute of Anatomy, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
19
|
Conoscenti G, Schneider T, Stoelzel K, Carfì Pavia F, Brucato V, Goegele C, La Carrubba V, Schulze-Tanzil G. PLLA scaffolds produced by thermally induced phase separation (TIPS) allow human chondrocyte growth and extracellular matrix formation dependent on pore size. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 80:449-459. [DOI: 10.1016/j.msec.2017.06.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 05/26/2017] [Accepted: 06/16/2017] [Indexed: 01/25/2023]
|
20
|
Behrendt P, Häfelein K, Preusse-Prange A, Bayer A, Seekamp A, Kurz B. IL-10 ameliorates TNF-α induced meniscus degeneration in mature meniscal tissue in vitro. BMC Musculoskelet Disord 2017; 18:197. [PMID: 28511649 PMCID: PMC5434535 DOI: 10.1186/s12891-017-1561-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022] Open
Abstract
Background Joint inflammation causes meniscus degeneration and can exacerbate post-traumatic meniscus injuries by extracellular matrix degradation, cellular de-differentiation and cell death. The aim of this study was to examine whether anti-inflammatory interleukin-10 exerts protective effects in an in vitro model of TNF-α-induced meniscus degeneration. Methods Meniscus tissue was harvested from the knees of adult cows. After 24 h of equilibrium explants were simultaneously treated with bovine TNF-α and IL-10. After an incubation time of 72 h cell death was measured histomorphometrically (nuclear blebbing, NB) and release of glycosaminoglycans (GAG, DMMB assay) and nitric oxide (NO, Griess-reagent) were analysed. Transcription levels (mRNA) of matrix degrading enzymes, collagen type X (COL10A1) and nitric oxide synthetase 2 (NOS2) were measured by quantitative real time PCR. TNF-α-dependent formation of the aggrecanase-specific aggrecan neoepitope NITEGE was visualised by immunostaining. Differences between groups were calculated using a one-way ANOVA with a Bonferroni post hoc test. Results Administration of IL-10 significantly prevented the TNF-α-related cell death (P .001), release of NO (P .003) and NOS2 expression (P .04). Release of GAG fragments (P .001), NITEGE formation and expression of MMP3 (P .007), -13 (P .02) and ADAMTS4 (P .001) were significantly reduced. The TNF-α-dependent increase in COL10A1 expression was also antagonized by IL-10 (P .02). Conclusion IL-10 prevented crucial mechanisms of meniscal degeneration induced by a key cytokine of OA, TNF-α. Administration of IL-10 might improve the biological regeneration and provide a treatment approach in degenerative meniscus injuries and in conditions of post-traumatic sports injuries.
Collapse
Affiliation(s)
- P Behrendt
- Department of Orthopaedics and Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - K Häfelein
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany
| | - A Preusse-Prange
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany
| | - A Bayer
- Department of Cardiovascular Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - A Seekamp
- Department of Orthopaedics and Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - B Kurz
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany
| |
Collapse
|
21
|
Goldberg A, Mitchell K, Soans J, Kim L, Zaidi R. The use of mesenchymal stem cells for cartilage repair and regeneration: a systematic review. J Orthop Surg Res 2017; 12:39. [PMID: 28279182 PMCID: PMC5345159 DOI: 10.1186/s13018-017-0534-y] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/13/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The management of articular cartilage defects presents many clinical challenges due to its avascular, aneural and alymphatic nature. Bone marrow stimulation techniques, such as microfracture, are the most frequently used method in clinical practice however the resulting mixed fibrocartilage tissue which is inferior to native hyaline cartilage. Other methods have shown promise but are far from perfect. There is an unmet need and growing interest in regenerative medicine and tissue engineering to improve the outcome for patients requiring cartilage repair. Many published reviews on cartilage repair only list human clinical trials, underestimating the wealth of basic sciences and animal studies that are precursors to future research. We therefore set out to perform a systematic review of the literature to assess the translation of stem cell therapy to explore what research had been carried out at each of the stages of translation from bench-top (in vitro), animal (pre-clinical) and human studies (clinical) and assemble an evidence-based cascade for the responsible introduction of stem cell therapy for cartilage defects. This review was conducted in accordance to PRISMA guidelines using CINHAL, MEDLINE, EMBASE, Scopus and Web of Knowledge databases from 1st January 1900 to 30th June 2015. In total, there were 2880 studies identified of which 252 studies were included for analysis (100 articles for in vitro studies, 111 studies for animal studies; and 31 studies for human studies). There was a huge variance in cell source in pre-clinical studies both of terms of animal used, location of harvest (fat, marrow, blood or synovium) and allogeneicity. The use of scaffolds, growth factors, number of cell passages and number of cells used was hugely heterogeneous. SHORT CONCLUSIONS This review offers a comprehensive assessment of the evidence behind the translation of basic science to the clinical practice of cartilage repair. It has revealed a lack of connectivity between the in vitro, pre-clinical and human data and a patchwork quilt of synergistic evidence. Drivers for progress in this space are largely driven by patient demand, surgeon inquisition and a regulatory framework that is learning at the same pace as new developments take place.
Collapse
Affiliation(s)
- Andy Goldberg
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| | - Katrina Mitchell
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| | - Julian Soans
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| | - Louise Kim
- Joint Research and Enterprise Office, St George’s University of London and St George’s University Hospitals NHS Foundation Trust, Hunter Wing, Cranmer Terrace, London, SW17 0RE UK
| | - Razi Zaidi
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| |
Collapse
|
22
|
Shioda M, Muneta T, Tsuji K, Mizuno M, Komori K, Koga H, Sekiya I. TNFα promotes proliferation of human synovial MSCs while maintaining chondrogenic potential. PLoS One 2017; 12:e0177771. [PMID: 28542363 PMCID: PMC5461123 DOI: 10.1371/journal.pone.0177771] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 05/03/2017] [Indexed: 12/21/2022] Open
Abstract
Synovial mesenchymal stem cells (MSCs) are a candidate cell source for cartilage and meniscus regeneration. If we can proliferate synovial MSCs more effectively, we can expand clinical applications to patients with large cartilage and meniscus lesions. TNFα is a pleiotropic cytokine that can affect the growth and differentiation of cells in the body. The purpose of this study was to examine the effect of TNFα on proliferation, chondrogenesis, and other properties of human synovial MSCs. Passage 1 human synovial MSCs from 2 donors were cultured with 2.5 x 10-12~10-7 g/ml, 10 fold dilution series of TNFα for 14 days, then the cell number and colony number was counted. The effect of the optimum dose of TNFα on proliferation was also examined in synovial MSCs from 6 donors. Chondrogenic potential of synovial MSCs pretreated with TNFα was evaluated in 6 donors. The expressions of 12 surface antigens were also examined in 3 donors.2.5 ng/ml and higher concentration of TNFα significantly increased cell number/dish and cell number/colony in both donors. The effect of 25 ng/ml TNFα was confirmed in all 6 donors. There was no significant difference in the weight, or amount of glycosaminoglycan and DNA of the cartilage pellets between the MSCs untreated and MSCs pretreated with 25 ng/ml TNFα. TNFα decreased expression rate of CD 105 and 140b in all 3 donors. TNFα promoted proliferation of synovial MSCs with increase of cell number/ colony. Pretreatment with TNFα did not affect chondrogenesis of synovial MSCs. However, TNFα affected some properties of synovial MSCs.
Collapse
Affiliation(s)
- Mikio Shioda
- Department of Joint Surgery and Sports Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Muneta
- Department of Joint Surgery and Sports Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuru Mizuno
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichiro Komori
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
23
|
The Signaling Pathways Involved in Chondrocyte Differentiation and Hypertrophic Differentiation. Stem Cells Int 2016; 2016:2470351. [PMID: 28074096 PMCID: PMC5198191 DOI: 10.1155/2016/2470351] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022] Open
Abstract
Chondrocytes communicate with each other mainly via diffusible signals rather than direct cell-to-cell contact. The chondrogenic differentiation of mesenchymal stem cells (MSCs) is well regulated by the interactions of varieties of growth factors, cytokines, and signaling molecules. A number of critical signaling molecules have been identified to regulate the differentiation of chondrocyte from mesenchymal progenitor cells to their terminal maturation of hypertrophic chondrocytes, including bone morphogenetic proteins (BMPs), SRY-related high-mobility group-box gene 9 (Sox9), parathyroid hormone-related peptide (PTHrP), Indian hedgehog (Ihh), fibroblast growth factor receptor 3 (FGFR3), and β-catenin. Except for these molecules, other factors such as adenosine, O2 tension, and reactive oxygen species (ROS) also have a vital role in cartilage formation and chondrocyte maturation. Here, we outlined the complex transcriptional network and the function of key factors in this network that determine and regulate the genetic program of chondrogenesis and chondrocyte differentiation.
Collapse
|
24
|
Behrendt P, Preusse-Prange A, Klüter T, Haake M, Rolauffs B, Grodzinsky AJ, Lippross S, Kurz B. IL-10 reduces apoptosis and extracellular matrix degradation after injurious compression of mature articular cartilage. Osteoarthritis Cartilage 2016; 24:1981-1988. [PMID: 27349464 DOI: 10.1016/j.joca.2016.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/25/2016] [Accepted: 06/13/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this study was to examine whether anti-inflammatory interleukin-10 (IL-10) exerts chondroprotective effects in an in vitro model of a single mechanical injury of mature articular cartilage. METHOD Articular cartilage was harvested from the femoro-patellar groove of adult cows (Bos taurus) and cultured w/o bovine IL-10. After 24 h of equilibration explants were subjected to an axial unconfined compression (50% strain, velocity 2 mm/s, held for 10 s). After 96 h cell death was measured histomorphometrically (nuclear blebbing, NB) and the release of glycosaminoglycans (GAG, DMMB assay) and nitric oxide (NO, Griess-reagent) were analyzed. mRNA levels of matrix degrading enzymes and nitric oxide synthetase were measured by quantitative real time PCR. Differences between groups were calculated using a one-way ANOVA with a Bonferroni post hoc test. RESULTS Injurious compression significantly increased the number of cells with NB, release of GAG and nitric oxide and expression of MMP-3, -13, ADAMTS-4 and NOS2. Administration of IL-10 significantly reduced the injury related cell death and release of GAG and NO, respectively. Expression of MMP-3, -13, ADAMTS-4 and NOS2 were significantly reduced. CONCLUSION Joint injury is a complex process involving specific mechanical effects on cartilage as well as induction of an inflammatory environment. IL-10 prevented crucial mechanisms of chondrodegeneration induced by an injurious single compression. IL-10 might be a multipurpose drug candidate for the treatment of cartilage-related sports injuries or osteoarthritis (OA).
Collapse
Affiliation(s)
- P Behrendt
- Department of Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Germany.
| | - A Preusse-Prange
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany.
| | - T Klüter
- Department of Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Germany.
| | - M Haake
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany.
| | - B Rolauffs
- Siegfried Weller Institute for Trauma Research & Clinic for Trauma and Restorative Surgery, BG Trauma Clinic Tuebingen, University of Tuebingen, Tuebingen, Germany; Department of Orthopedics and Trauma Surgery, Albert Ludwigs University of Freiburg, Freiburg, Germany.
| | - A J Grodzinsky
- Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - S Lippross
- Department of Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Germany.
| | - B Kurz
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany.
| |
Collapse
|
25
|
Koizumi K, Ebina K, Hart DA, Hirao M, Noguchi T, Sugita N, Yasui Y, Chijimatsu R, Yoshikawa H, Nakamura N. Synovial mesenchymal stem cells from osteo- or rheumatoid arthritis joints exhibit good potential for cartilage repair using a scaffold-free tissue engineering approach. Osteoarthritis Cartilage 2016; 24:1413-22. [PMID: 26973329 DOI: 10.1016/j.joca.2016.03.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To assess whether synovial mesenchymal stem cells (SMSCs) from patients with osteoarthritis (OA) or rheumatoid arthritis (RA) can be used as an alternative cell source for cartilage repair using allogenic tissue engineered construct (TEC). METHODS Twenty-five patients (17 female, average age 61.8 years) were divided according to their pathology (control trauma group; N = 6, OA group; N = 6) and RA patients were subdivided into two groups to evaluate the impact of biologics in accordance with whether treated with biologics [Bio(+)RA; N = 7] or not [Bio(-)RA; N = 6]. We compared the following characteristics among these groups: (1) The cell proliferation capacity of SMSCs; (2) The influence of passage number on features of SMSCs; (3) The weight and volume of TEC from the same number of SMSCs; (4) Inflammatory cytokine gene expressions levels of TEC; (5) The chondrogenic potential of TEC; and (6) Osteochondral repair using TEC in athymic nude rats. RESULTS SMSCs from the four groups exhibited equivalent features in the above evaluation items. In in vivo studies, the TEC-treated repair tissues for all groups exhibited significantly better outcomes than those for the untreated group and no significant differences among the four TEC groups. CONCLUSION SMSCs from OA or RA patients are no less appropriate for repairing cartilage than those from trauma patients and thus, may be an effective source for allogenic cell-based cartilage repair.
Collapse
Affiliation(s)
- K Koizumi
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - K Ebina
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - D A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Canada
| | - M Hirao
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - T Noguchi
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - N Sugita
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Y Yasui
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - R Chijimatsu
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - H Yoshikawa
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - N Nakamura
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan; Institute for Medical Science in Sports, Osaka Health Science University, 1-9-27, Tennma, Kita-ku, Osaka 530-0043, Japan.
| |
Collapse
|
26
|
Zayed MN, Schumacher J, Misk N, Dhar MS. Effects of pro-inflammatory cytokines on chondrogenesis of equine mesenchymal stromal cells derived from bone marrow or synovial fluid. Vet J 2016; 217:26-32. [PMID: 27810206 DOI: 10.1016/j.tvjl.2016.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022]
Abstract
Mesenchymal stromal cells (MSCs) have the capacity to differentiate into cells of mesenchymal lineage, such as chondrocytes, and have potential for use in regeneration of equine articular cartilage. MSCs instilled intra-articularly would be exposed to the inflamed environment associated with equine osteoarthritis (OA), which may compromise their function and ability to heal a cartilaginous defect. The aim of this study was to assess the ability of equine adult MSCs to differentiate into chondrocytes when stimulated with pro-inflammatory cytokines. MSCs derived from equine bone marrow (BM) and from synovial fluid (SF) were cultured in chondrogenic induction medium containing transforming growth factor (TGF)-β1. BM-derived MSCs (BMMSCs) and SF-derived MSCs (SFMSCs) were stimulated with 100 ng/mL interferon (IFN)-γ and 10 ng/mL tumor necrosis factor (TNF)-α. Chondrogenic differentiation was measured quantitatively with the glycosaminoglycan (GAG) assay and qualitatively by immunofluorescence (IF) for SOX-9, TGF-β1, aggrecan and collagen II. The viability of equine MSCs was maintained in the presence of IFN-γ and TNF-α, but production of GAGs from both types of MSCs was decreased in stimulated medium. Exposure of BMMSCs to pro-inflammatory cytokines reduced the levels of SOX-9, TGF-β1, aggrecan and collagen II, whereas exposure of SFMSCs to these cytokines reduced the levels of aggrecan only. These data suggest that pro-inflammatory cytokines do not affect proliferation of MSCs, but could inhibit chondrogenesis of MSCs.
Collapse
Affiliation(s)
- M N Zayed
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive c247, Knoxville, TN 37996, USA; Department of Animal Surgery, College of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - J Schumacher
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive c247, Knoxville, TN 37996, USA
| | - N Misk
- Department of Animal Surgery, College of Veterinary Medicine, Assuit University, Assuit 71526, Egypt
| | - M S Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive c247, Knoxville, TN 37996, USA.
| |
Collapse
|
27
|
Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation 2016; 92:41-51. [PMID: 27012163 DOI: 10.1016/j.diff.2016.02.005] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/15/2016] [Accepted: 02/25/2016] [Indexed: 11/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that represent a promising source for regenerative medicine. MSCs are capable of osteogenic, chondrogenic, adipogenic and myogenic differentiation. Efficacy of differentiated MSCs to regenerate cells in the injured tissues requires the ability to maintain the differentiation toward the desired cell fate. Since MSCs represent an attractive source for autologous transplantation, cellular and molecular signaling pathways and micro-environmental changes have been studied in order to understand the role of cytokines, chemokines, and transcription factors on the differentiation of MSCs. The differentiation of MSC into a mesenchymal lineage is genetically manipulated and promoted by specific transcription factors associated with a particular cell lineage. Recent studies have explored the integration of transcription factors, including Runx2, Sox9, PPARγ, MyoD, GATA4, and GATA6 in the differentiation of MSCs. Therefore, the overexpression of a single transcription factor in MSCs may promote trans-differentiation into specific cell lineage, which can be used for treatment of some diseases. In this review, we critically discussed and evaluated the role of transcription factors and related signaling pathways that affect the differentiation of MSCs toward adipocytes, chondrocytes, osteocytes, skeletal muscle cells, cardiomyocytes, and smooth muscle cells.
Collapse
Affiliation(s)
- Sami G Almalki
- Departments of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K Agrawal
- Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA.
| |
Collapse
|
28
|
Ham O, Lee CY, Kim R, Lee J, Oh S, Lee MY, Kim J, Hwang KC, Maeng LS, Chang W. Therapeutic Potential of Differentiated Mesenchymal Stem Cells for Treatment of Osteoarthritis. Int J Mol Sci 2015; 16:14961-78. [PMID: 26147426 PMCID: PMC4519882 DOI: 10.3390/ijms160714961] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/24/2015] [Accepted: 06/26/2015] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive, and irreversible degenerative joint disease. Conventional OA treatments often result in complications such as pain and limited activity. However, transplantation of mesenchymal stem cells (MSCs) has several beneficial effects such as paracrine effects, anti-inflammatory activity, and immunomodulatory capacity. In addition, MSCs can be differentiated into several cell types, including chondrocytes, osteocytes, endothelia, and adipocytes. Thus, transplantation of MSCs is a suggested therapeutic tool for treatment of OA. However, transplanted naïve MSCs can cause problems such as heterogeneous populations including differentiated MSCs and undifferentiated cells. To overcome this problem, new strategies for inducing differentiation of MSCs are needed. One possibility is the application of microRNA (miRNA) and small molecules, which regulate multiple molecular pathways and cellular processes such as differentiation. Here, we provide insight into possible strategies for cartilage regeneration by transplantation of differentiated MSCs to treat OA patients.
Collapse
Affiliation(s)
- Onju Ham
- Catholic Kwandong University International St. Mary's Hospital, Incheon 404-834, Korea.
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, 50 Yonsei-ro, Seodamun-gu, Seoul 120-759, Korea.
| | - Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735, Korea.
| | - Jihyun Lee
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735, Korea.
| | - Sekyung Oh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 702-701, Korea.
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women's University, Seoul 140-742, Korea.
| | - Ki-Chul Hwang
- Catholic Kwandong University International St. Mary's Hospital, Incheon 404-834, Korea.
| | - Lee-So Maeng
- Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, the Catholic University of Korea, College of Medicine, Incheon 403-720, Korea.
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735, Korea.
| |
Collapse
|