1
|
Rácz K, Segal Y, Lénárt K, Fillér C, Tóth A, Szegeczki V, Gergely P, Zákány R, Reglődi D, Juhász T. Cartilage degradation is followed by PAC1 receptor reduction in articular cartilage of human knee joints. GeroScience 2025:10.1007/s11357-025-01689-4. [PMID: 40369255 DOI: 10.1007/s11357-025-01689-4] [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: 02/21/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide expressed in the nervous system and also in various peripheral tissues, including the musculoskeletal system. PACAP has an important function in the regulation of chondrogenesis and plays a protective role in cartilage oxidative and mechanical stress. PACAP knockout (KO) mice show early signs of aging and osteoarthritis in knee joint articular cartilage. Its specific, most potent receptor is the PAC1 receptor, the activation of which leads to enhanced Sox9 expression and subsequently, it increases the expression of collagen type II, glucosaminoglycans and aggrecan. In the present study, we investigated articular cartilage of human knee joints taken from cadavers of varying ages. Thickness and extracellular matrix content of articular cartilage of knee joints decreases with aging. The cartilage degeneration process most likely begins between the ages of 40 to 50. Expression of PAC1 receptor decreases in parallel with the reduction of cartilage thickness, leading to subsequent reduced Sox9 expression with cartilage specific matrix production. In summary, we found correlation in the reduction of cartilage thickness and quality together with PAC1 receptor expression and activity.
Collapse
Affiliation(s)
- Kálmán Rácz
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Yonatan Segal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Kinga Lénárt
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Csaba Fillér
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Anna Tóth
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Vince Szegeczki
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Péter Gergely
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Dóra Reglődi
- Department of Anatomy, Medical School, HUN-REN-PTE PACAP Research Team, University of Pécs, Szigeti Út 12, 7624, Pécs, Hungary
| | - Tamás Juhász
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary.
| |
Collapse
|
2
|
Liu W, Feng M, Xu P. From regeneration to osteoarthritis in the knee joint: The role shift of cartilage-derived progenitor cells. Front Cell Dev Biol 2022; 10:1010818. [PMID: 36340024 PMCID: PMC9630655 DOI: 10.3389/fcell.2022.1010818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/30/2022] [Indexed: 12/02/2022] Open
Abstract
A mount of growing evidence has proven that cartilage-derived progenitor cells (CPCs) harbor strong proliferation, migration, andmultiple differentiation potentials over the past 2 decades. CPCs in the stage of immature tissue play an important role in cartilage development process and injured cartilage repair in the young and active people. However, during maturation and aging, cartilage defects cannot be completely repaired by CPCs in vivo. Recently, tissue engineering has revealed that repaired cartilage defects with sufficient stem cell resources under good condition and bioactive scaffolds in vitro and in vivo. Chronic inflammation in the knee joint limit the proliferation and chondrogenesis abilities of CPCs, which further hampered cartilage healing and regeneration. Neocartilage formation was observed in the varus deformity of osteoarthritis (OA) patients treated with offloading technologies, which raises the possibility that organisms could rebuild cartilage structures spontaneously. In addition, nutritionmetabolismdysregulation, including glucose and free fatty acid dysregulation, could influence both chondrogenesis and cartilage formation. There are a few reviews about the advantages of CPCs for cartilage repair, but few focused on the reasons why CPCs could not repair the cartilage as they do in immature status. A wide spectrum of CPCs was generated by different techniques and exhibited substantial differences. We recently reported that CPCs maybe are as internal inflammation sources during cartilage inflammaging. In this review, we further streamlined the changes of CPCs from immature development to maturation and from healthy status to OA advancement. The key words including “cartilage derived stem cells”, “cartilage progenitor cells”, “chondroprogenitor cells”, “chondroprogenitors” were set for latest literature searching in PubMed and Web of Science. The articles were then screened through titles, abstracts, and the full texts in sequence. The internal environment including long-term inflammation, extendedmechanical loading, and nutritional elements intake and external deleterious factors were summarized. Taken together, these results provide a comprehensive understanding of the underlying mechanism of CPC proliferation and differentiation during development, maturation, aging, injury, and cartilage regeneration in vivo.
Collapse
Affiliation(s)
- Wenguang Liu
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Meng Feng
- Department of Orthopedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Peng Xu
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Peng Xu,
| |
Collapse
|
3
|
Gene Expression and Chondrogenic Potential of Cartilage Cells: Osteoarthritis Grade Differences. Int J Mol Sci 2022; 23:ijms231810610. [PMID: 36142513 PMCID: PMC9504485 DOI: 10.3390/ijms231810610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Recent data suggest that cells isolated from osteoarthritic (OA) cartilage express mesenchymal progenitor cell (MPC) markers that have the capacity to form hyaline-like cartilage tissue. Whether or not these cells are influenced by the severity of OA remains unexplored. Therefore, we analyzed MPC marker expression and chondrogenetic potential of cells from mild, moderate and severe OA tissue. Human osteoarthritic tibial plateaus were obtained from 25 patients undergoing total knee replacement. Each sample was classified as mild, moderate or severe OA according to OARSI scoring. mRNA expression levels of MPC markers—CD105, CD166, Notch 1, Sox9; mature chondrocyte markers—Aggrecan (Acan), Col II A1, hypertrophic chondrocyte and osteoarthritis-related markers—Col I A1, MMP-13 and ALPL were measured at the tissue level (day 0), after 2 weeks of in vitro expansion (day 14) and following chondrogenic in vitro re-differentiation (day 35). Pellet matrix composition after in vitro chondrogenesis of different OA-derived cells was tested for proteoglycans, collagen II and I by safranin O and immunofluorescence staining. Multiple MPC markers were found in OA cartilage resident tissue within a single OA joint with no significant difference between grades except for Notch1, which was higher in severe OA tissues. Expression levels of CD105 and Notch 1 were comparable between OA cartilage-derived cells of different disease grades and bone marrow mesenchymal stem cell (BM-MSC) line (healthy control). However, the MPC marker Sox 9 was conserved after in vitro expansion and significantly higher in OA cartilage-derived cells compared to its levels in the BM-MSC. The in vitro expansion of cartilage-derived cells resulted in enrichment while re–differentiation in reduction of MPC markers for all three analyzed grades. However, only moderate OA-derived cells after the in vitro chondrogenesis resulted in the formation of hyaline cartilage-like tissue. The latter tissue samples were also highly positive for collagen II and proteoglycans with no expression of osteoarthritis-related markers (collagen I, ALPL and MMP13). MPC marker expression did not differ between OA grades at the tissue level. Interestingly after in vitro re-differentiation, only moderate OA-derived cells showed the capacity to form hyaline cartilage-like tissue. These findings may have implications for clinical practice to understand the intrinsic repair capacity of articular cartilage in OA tissues and raises the possibility of these progenitor cells as a candidate for articular cartilage repair.
Collapse
|
4
|
Matheus HR, Özdemir ŞD, Guastaldi FPS. Stem cell-based therapies for temporomandibular joint osteoarthritis and regeneration of cartilage/osteochondral defects: a systematic review of preclinical experiments. Osteoarthritis Cartilage 2022; 30:1174-1185. [PMID: 35597373 DOI: 10.1016/j.joca.2022.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVES The aim of this systematic review was to assess the effects of stem cell-based therapies on the treatment of Temporomandibular Joint Osteoarthritis (TMJ-OA) and the regeneration of cartilage/osteochondral defects. METHODS Data on preclinical studies evaluating the effectiveness of stem cell-based therapies for treating Temporomandibular Disorders (TMDs) were extracted from PubMed, Web of Science, and Cochrane Library and the grey literature by three independent reviewers. A manual search was performed in the databases, the reference list of review studies, and relevant journals in the field. Compliance with the ARRIVE guidelines was evaluated for quality assessment. SYRCLE's risk of bias tool for animal experimental studies was assessed to define internal validity. RESULTS After applying the inclusion and exclusion criteria, 10 studies were included in the qualitative synthesis. Regardless of cell origin, stem cell-based therapeutic approaches induced protective, anti-inflammatory, and chondroregenerative potential in the treatment of TMJ-OA. Regeneration of the cartilage layer on the surface of the condyle was achieved when stem cells were directly flushed into the defect or when delivered within a carrier. CONCLUSION Stem cell-based therapies may be considered a promising approach for the treatment of TMJ-OA and for the regeneration of full-thickness cartilage and osteochondral defects in the TMJ. Human studies shall be performed to validate these results found in animals.
Collapse
Affiliation(s)
- H R Matheus
- Skeletal Biology Research Center, Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA, USA; Department of Diagnosis and Surgery - Periodontics Division, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil.
| | - Ş D Özdemir
- Skeletal Biology Research Center, Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA, USA; Istanbul Medipol University, School of Dentistry, İstanbul, Turkey.
| | - F P S Guastaldi
- Skeletal Biology Research Center, Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
5
|
Yuan Z, Liu S, Song W, Liu Y, Bi G, Xie R, Ren L. Galactose Enhances Chondrogenic Differentiation of ATDC5 and Cartilage Matrix Formation by Chondrocytes. Front Mol Biosci 2022; 9:850778. [PMID: 35615738 PMCID: PMC9124793 DOI: 10.3389/fmolb.2022.850778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/07/2022] [Indexed: 12/01/2022] Open
Abstract
Galactose, an important carbohydrate nutrient, is involved in several types of cellular metabolism, participating in physiological activities such as glycosaminoglycan (GAG) synthesis, glycosylation, and intercellular recognition. The regulatory effects of galactose on osteoarthritis have attracted increased attention. In this study, in vitro cell models of ATDC5 and chondrocytes were prepared and cultured with different concentrations of galactose to evaluate its capacity on chondrogenesis and cartilage matrix formation. The cell proliferation assay demonstrated that galactose was nontoxic to both ATDC5 cells and chondrocytes. RT-PCR and immunofluorescence staining indicated that the gene expressions of cartilage matrix type II collagen and aggrecan were significantly upregulated with increasing galactose concentration and the expression and accumulation of the extracellular matrix (ECM) protein. Overall, these results indicated that a galactose concentration below 8 mM exhibited the best effect on promoting chondrogenesis, which entitles galactose as having considerable potential for cartilage repair and regeneration.
Collapse
Affiliation(s)
- Zhongrun Yuan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Sa Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Sa Liu, ; Renjian Xie, ; Li Ren,
| | - Wenjing Song
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Ying Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Gangyuan Bi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Renjian Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, China
- Jiangxi Key Laboratory of Medical Tissue Engineering Materials and Biofabrication, Gannan Medical University, Ganzhou, China
- *Correspondence: Sa Liu, ; Renjian Xie, ; Li Ren,
| | - Li Ren
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Sa Liu, ; Renjian Xie, ; Li Ren,
| |
Collapse
|
6
|
Xia P, Wang Q, Song J, Wang X, Wang X, Lin Q, Cheng K, Chen A, Li X. Low-Intensity Pulsed Ultrasound Enhances the Efficacy of Bone Marrow-Derived MSCs in Osteoarthritis Cartilage Repair by Regulating Autophagy-Mediated Exosome Release. Cartilage 2022; 13:19476035221093060. [PMID: 35438034 PMCID: PMC9137322 DOI: 10.1177/19476035221093060] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The present study explored whether low-intensity pulsed ultrasound (LIPUS) enhances the therapeutic efficacy of mesenchymal stem cells (MSCs) in osteoarthritis (OA) cartilage repair by regulating autophagy-mediated exosome release. DESIGN MSCs were isolated from the rat bone marrow and treated with rapamycin, 3-methyladenine, or LIPUS. The mechanism of the LIPUS-stimulated exosome release by MSCs was analyzed by inhibiting autophagy. In addition, the MSCs were co-cultured with OA chondrocytes and stimulated by LIPUS, with or without exosome release inhibitor intervention. The exosome release was detected through transmission electron microscopy (TEM), nanoparticle tracking analysis, and biomarker expression analysis. Autophagy was analyzed through TEM, autophagy-related gene expression analysis, and immunofluorescence analysis in vitro. Furthermore, a rat knee OA model was constructed and treated with MSCs, GW4869, and LIPUS. The cartilage repair was assessed through histopathological analysis and extracellular matrix protein expression analysis. RESULTS The in vitro results indicated that LIPUS promoted MSC exosome release by activating autophagy. The in vivo results demonstrated that LIPUS significantly enhanced the positive effects of MSCs on OA cartilage. These effects were significantly blocked by GW4869, an inhibitor of exosome release. CONCLUSIONS LIPUS can enhance the therapeutic efficacy of MSCs in OA cartilage repair, and the underlying mechanism is related to the increase in autophagy-mediated exosome release.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qi Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiulong Song
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xinwei Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qiang Lin
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kai Cheng
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Anliang Chen
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,Xueping Li, Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210012, China.
| |
Collapse
|
7
|
Zhang Y, Xu X, Zhou P, Liu Q, Zhang M, Yang H, Yu S, Zhang J, Huo W, Zhao Y, Wang M. Elder Mice Exhibit More Severe Degeneration and Milder Regeneration in Temporomandibular Joints Subjected to Bilateral Anterior Crossbite. Front Physiol 2021; 12:750468. [PMID: 34925057 PMCID: PMC8674645 DOI: 10.3389/fphys.2021.750468] [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: 08/13/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Temporomandibular joints (TMJs) have a biomechanical relationship with dental occlusion. Aberrant occlusion initiates degenerative remodeling responses in TMJ condyles. Aging is a promoting factor of osteoarthritis (OA) development. The aim of this study was to assess the effect of aging on degenerative remodeling in TMJ condyles in response to occlusal biomechanical stimulation caused by the installation of aberrant prostheses and observe rehabilitation after their removal. The experiments involved 84 female C57BL/6J mice (42 at 6 weeks old and 42 at 28 weeks old). A bilateral anterior crossbite (BAC) model was developed, and the TMJs were sampled at 3, 7, and 11 weeks. BAC was removed at 7 weeks in a subset of mice, which accepted BAC treatment at 6 week of age, and maintained for another 4 weeks after BAC removal. TMJ changes were assessed with micro-CT, histomorphology, immunohistochemistry (IHC), and immunofluorescence staining assays. The results showed that BAC induced typical OA-like TMJ lesions that were more severe in the elder groups as evaluated by the acellular zones, clustered chondrocytes, fissures between cartilage and subchondral bone, reductions in matrix amount and the cartilage thickness as revealed by histomorphological measurements, and subchondral bone loss as detected on micro-CT images. IHC indicated significant increases in cleaved caspase-3-expressing cells and decreases in ki67-positive cells in the BAC groups. There were obvious age-dependent changes in the numbers of superficial zone cells and CD90-expressing cells. Supportively, cleaved caspase-3-expressing cells obviously increased, while ki67-expressing cells significantly decreased with aging. In the elder BAC groups, the superficial zone cells such as CD90-expressing cells were greatly reduced. At 11 weeks, the superficial zone cells were almost non-existent, and there were clear serrated injuries on the cartilage surface. BAC removal attenuated the degenerative changes in the condylar cartilage and subchondral bone. Notably, the rescue effect was more pronounced in the younger animals. Our findings demonstrate the impacts of aging on both TMJ degenerative changes in response to BAC and regenerative changes following BAC removal. The reduced number of chondro-progenitor cells in aged TMJ cartilage provides an explanation for this age-related decline in TMJ rehabilitative behaviors.
Collapse
Affiliation(s)
- Yuejiao Zhang
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| | - Xiaojie Xu
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China.,The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Peng Zhou
- School of Stomatology, Jiamusi University, Jiamusi, China
| | - Qian Liu
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| | - Mian Zhang
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| | - Hongxu Yang
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| | - Shibin Yu
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| | - Jing Zhang
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| | - Wanqiu Huo
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yali Zhao
- First Center of Hepatobiliary Surgery, Fifth Medical Center of the PLA General Hospital, Beijing, China
| | - Meiqing Wang
- The Key Laboratory of Military Stomatology of State, Clinic of Temporomandibular Joint Disorders and Oral and Maxillofacial Pain, Department of Oral Anatomy and Physiology, School of Stomatology, The National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Enhancing the chondrogenic potential of chondrogenic progenitor cells by deleting RAB5C. iScience 2021; 24:102464. [PMID: 34013174 PMCID: PMC8113995 DOI: 10.1016/j.isci.2021.102464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 04/21/2021] [Indexed: 11/21/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease that affects a large proportion of the elderly population. Chondrogenic progenitor cells (CPCs) reside in late-stage OA cartilage tissue, producing a fibrocartilaginous extracellular matrix; these cells can be manipulated in vitro to deposit proteins of healthy articular cartilage. CPCs are under the control of SOX9 and RUNX2. In our earlier studies, we showed that a knockdown of RUNX2 enhanced the chondrogenic potential of CPCs. Here we demonstrate that CPCs carrying a knockout of RAB5C, a protein involved in endosomal trafficking, exhibited elevated expression of multiple chondrogenic markers, including the SOX trio, and increased COL2 deposition, whereas no changes in COL1 deposition were observed. We report RAB5C as an attractive target for future therapeutic approaches designed to increase the COL2 content in the diseased joint.
Collapse
|
9
|
Mou TC, Feng JY. Research advances in cartilage stem cells markers and induced differentiation. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:108-114. [PMID: 33723946 DOI: 10.7518/hxkq.2021.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cartilage stem cells (CSCs) are cells that self-proliferate, have surface antigen expression, and have multidirectional differentiation potential in the articular cartilage. CSCs, as an ideal source of stem cells, has a good application prospect in stem cell therapy. This article reviews the CSCs markers, cartilage differentiation signaling pathway, and clinical treatment of osteoarthritis.
Collapse
Affiliation(s)
- Ting-Chen Mou
- Dept. of Stomatological, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Jian-Ying Feng
- College of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
10
|
Arias C, Saavedra N, Leal K, Vásquez B, Abdalla DSP, Salazar LA. Histological Evaluation and Gene Expression Profiling of Autophagy-Related Genes for Cartilage of Young and Senescent Rats. Int J Mol Sci 2020; 21:ijms21228607. [PMID: 33203108 PMCID: PMC7697851 DOI: 10.3390/ijms21228607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 01/06/2023] Open
Abstract
Autophagy is a cellular mechanism that protects cells from stress by digesting non-functional cellular components. In the cartilage, chondrocytes depend on autophagy as a principal mechanism to maintain cellular homeostasis. This protective role diminishes prior to the structural damage that normally occurs during aging. Considering that aging is the main risk factor for osteoarthritis, evaluating the expression of genes associated with autophagy in senescent cartilage might allow for the identification of potential therapeutic targets for treatment. Thus, we studied two groups of young and senescent rats. A histological analysis of cartilage and gene expression quantification for autophagy-related genes were performed. In aged cartilage, morphological changes were observed, such as an increase in cartilage degeneration as measured by the modified Mankin score, a decrease in the number of chondrocytes and collagen II (Col2a1), and an increase in matrix metalloproteinase 13 (Mmp13). Moreover, 84 genes associated with autophagy were evaluated by a PCR array analysis, and 15 of them were found to be significantly decreased with aging. Furthermore, an in silico analysis based on by two different bioinformatics software tools revealed that several processes including cellular homeostasis, autophagosome assembly, and aging—as well as several biological pathways such as autophagy, insulin-like growth factor 1 (IGF-1) signaling, PI3K (phosphoinositide 3-kinase)/AKT (serine/threonine kinase) signaling, and mammalian target of rapamycin (mTOR) signaling—were enriched. In conclusion, the analysis identified some potential targets for osteoarthritis treatment that would allow for the development of new therapeutic strategies for this chronic disease.
Collapse
Affiliation(s)
- Consuelo Arias
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
- Carrera de Kinesiología, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Av. Alemania 1090, Temuco 4810101, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
| | - Karla Leal
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
| | - Bélgica Vásquez
- Facultad de Ciencias de la Salud, Universidad de Tarapacá, Av. General Velásquez 1775, Arica 1000007, Chile;
| | - Dulcineia S. P. Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, Universidade de São Paulo, Avenida Professor Lineu Prestes 580, São Paulo CEP 05508-000, SP, Brazil;
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Av. Francisco Salazar 01145, Temuco 4811230, Chile; (C.A.); (N.S.); (K.L.)
- Correspondence: ; Tel.: +56-45-259-6724
| |
Collapse
|
11
|
Sanjurjo-Rodriguez C, Altaie A, Mastbergen S, Baboolal T, Welting T, Lafeber F, Pandit H, McGonagle D, Jones E. Gene Expression Signatures of Synovial Fluid Multipotent Stromal Cells in Advanced Knee Osteoarthritis and Following Knee Joint Distraction. Front Bioeng Biotechnol 2020; 8:579751. [PMID: 33178674 PMCID: PMC7591809 DOI: 10.3389/fbioe.2020.579751] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disorder. Although joint replacement remains the standard of care for knee OA patients, knee joint distraction (KJD), which works by temporarily off-loading the joint for 6–8 weeks, is becoming a novel joint-sparing alternative for younger OA sufferers. The biological mechanisms behind KJD structural improvements remain poorly understood but likely involve joint-resident regenerative cells including multipotent stromal cells (MSCs). In this study, we hypothesized that KJD leads to beneficial cartilage-anabolic and anti-catabolic changes in joint-resident MSCs and investigated gene expression profiles of synovial fluid (SF) MSCs following KJD as compared with baseline. To obtain further insights into the effects of local biomechanics on MSCs present in late OA joints, SF MSC gene expression was studied in a separate OA arthroplasty cohort and compared with subchondral bone (SB) MSCs from medial (more loaded) and lateral (less loaded) femoral condyles from the same joints. In OA arthroplasty cohort (n = 12 patients), SF MSCs expressed lower levels of ossification- and hypotrophy-related genes [bone sialoprotein (IBSP), parathyroid hormone 1 receptor (PTH1R), and runt-related transcription factor 2 (RUNX2)] than did SB MSCs. Interestingly, SF MSCs expressed 5- to 50-fold higher levels of transcripts for classical extracellular matrix turnover molecules matrix metalloproteinase 1 (MMP1), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5), and tissue inhibitor of metalloproteinase-3 (TIMP3), all (p < 0.05) potentially indicating greater cartilage remodeling ability of OA SF MSCs, compared with SB MSCs. In KJD cohort (n = 9 patients), joint off-loading resulted in sustained, significant increase in SF MSC colonies’ sizes and densities and a notable transcript upregulation of key cartilage core protein aggrecan (ACAN) (weeks 3 and 6), as well as reduction in pro-inflammatory C–C motif chemokine ligand 2 (CCL2) expression (weeks 3 and 6). Additionally, early KJD changes (week 3) were marked by significant increases in MSC chondrogenic commitment markers gremlin 1 (GREM1) and growth differentiation factor 5 (GDF5). In combination, our results reveal distinct transcriptomes on joint-resident MSCs from different biomechanical environments and show that 6-week joint off-loading leads to transcriptional changes in SF MSCs that may be beneficial for cartilage regeneration. Biomechanical factors should be certainly considered in the development of novel MSC-based therapies for OA.
Collapse
Affiliation(s)
- Clara Sanjurjo-Rodriguez
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Physiotherapy, Medicine and Biomedical Sciences department, CIBER-BBN, Institute of Biomedical Research of A Coruña (INIBIC)-Centre of Advanced Scientific Researches (CICA), University of A Coruña, A Coruña, Spain
| | - Ala Altaie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Simon Mastbergen
- University Medical Center Utrecht, Rheumatology & Clinical Immunology, Regenerative Medicine Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Thomas Baboolal
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tim Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Floris Lafeber
- University Medical Center Utrecht, Rheumatology & Clinical Immunology, Regenerative Medicine Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
12
|
Biological potential alterations of migratory chondrogenic progenitor cells during knee osteoarthritic progression. Arthritis Res Ther 2020; 22:62. [PMID: 32216831 PMCID: PMC7099802 DOI: 10.1186/s13075-020-2144-z] [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: 12/10/2019] [Accepted: 03/05/2020] [Indexed: 12/22/2022] Open
Abstract
Background Although increasing studies have demonstrated that chondrogenic progenitor cells (CPCs) remain present in human osteoarthritic cartilage, the biological alterations of the CPCs from the less diseased lateral tibial condyle and the more diseased medial condyle of same patient remain to be investigated. Methods CPCs were isolated from paired grade 1–2 and grade 3–4 osteoarthritic cartilage by virtue of cell migratory capacities. The cell morphology, immunophenotype, self-renewal, multi-differentiation, and cell migration of these CPCs were evaluated. Additionally, the distributions of CD105+/CD271+ cells in OA osteochondral specimen were determined. Furthermore, a high-throughput mRNA sequencing was performed. Results Migratory CPCs (mCPCs) robustly outgrew from mildly collagenases-digested osteoarthritic cartilages. The mCPCs from grade 3–4 cartilages (mCPCs, grades 3–4) harbored morphological characteristics, cell proliferation, and colony formation capacity that were similar to those of the mCPCs from the grade 1–2 OA cartilages (mCPCs, grades 1–2). However, the mCPCs (grades 3–4) highly expressed CD271. In addition, the mCPCs (grades 3–4) showed enhanced osteo-adipogenic activities and decreased chondrogenic capacity. Furthermore, the mCPCs (grades 3–4) exhibited stronger cell migration in response to osteoarthritis synovial fluids. More CD105+/CD271+ cells resided in grade 3–4 articular cartilages. Moreover, the results of mRNA sequencing showed that mCPCs (grades 3–4) expressed higher migratory molecules. Conclusions Our data suggest that more mCPCs (grades 3–4) migrate to injured articular cartilages but with enhanced osteo-adipogenic and decreased chondrogenic capacity, which might explain the pathological changes of mCPCs during the progression of OA from early to late stages. Thus, these dysfunctional mCPCs might be optional cell targets for OA therapies.
Collapse
|
13
|
Olate-Moya F, Arens L, Wilhelm M, Mateos-Timoneda MA, Engel E, Palza H. Chondroinductive Alginate-Based Hydrogels Having Graphene Oxide for 3D Printed Scaffold Fabrication. ACS APPLIED MATERIALS & INTERFACES 2020; 12:4343-4357. [PMID: 31909967 DOI: 10.1021/acsami.9b22062] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Scaffolds based on bioconjugated hydrogels are attractive for tissue engineering because they can partly mimic human tissue characteristics. For example, they can further increase their bioactivity with cells. However, most of the hydrogels present problems related to their processability, consequently limiting their use in 3D printing to produce tailor-made scaffolds. The goal of this work is to develop bioconjugated hydrogel nanocomposite inks for 3D printed scaffold fabrication through a micro-extrusion process having improved both biocompatibility and processability. The hydrogel is based on a photocrosslinkable alginate bioconjugated with both gelatin and chondroitin sulfate in order to mimic the cartilage extracellular matrix, while the nanofiller is based on graphene oxide to enhance the printability and cell proliferation. Our results show that the incorporation of graphene oxide into the hydrogel inks considerably improved the shape fidelity and resolution of 3D printed scaffolds because of a faster viscosity recovery post extrusion of the ink. Moreover, the nanocomposite inks produce anisotropic threads after the 3D printing process because of the templating of the graphene oxide liquid crystal. The in vitro proliferation assay of human adipose tissue-derived mesenchymal stem cells (hADMSCs) shows that bioconjugated scaffolds present higher cell proliferation than pure alginate, with the nanocomposites presenting the highest values at long times. Live/Dead assay otherwise displays full viability of hADMSCs adhered on the different scaffolds at day 7. Notably, the scaffolds produced with nanocomposite hydrogel inks were able to guide the cell proliferation following the direction of the 3D printed threads. In addition, the bioconjugated alginate hydrogel matrix induced chondrogenic differentiation without exogenous pro-chondrogenesis factors as concluded from immunostaining after 28 days of culture. This high cytocompatibility and chondroinductive effect toward hADMSCs, together with the improved printability and anisotropic structures, makes these nanocomposite hydrogel inks a promising candidate for cartilage tissue engineering based on 3D printing.
Collapse
Affiliation(s)
- Felipe Olate-Moya
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas , Universidad de Chile , Beauchef 851 , 8370456 Santiago , Chile
| | - Lukas Arens
- Institute for Technical Chemistry and Polymer Chemistry (ITCP) , Karlsruhe Institute of Technology (KIT) , Engesserstr. 18 , 76131 Karlsruhe , Germany
| | - Manfred Wilhelm
- Institute for Technical Chemistry and Polymer Chemistry (ITCP) , Karlsruhe Institute of Technology (KIT) , Engesserstr. 18 , 76131 Karlsruhe , Germany
| | - Miguel Angel Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC) , The Barcelona Institute of Science and Technology , Baldiri Reixac 10-12 , 08028 Barcelona , Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Monforte de Lemos, 3-5 , 28029 Madrid , Spain
- Department of Materials Science, EEBE , Technical University of Catalonia (UPC) , d'Eduard Maristany 16 , 08019 Barcelona , Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC) , The Barcelona Institute of Science and Technology , Baldiri Reixac 10-12 , 08028 Barcelona , Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Monforte de Lemos, 3-5 , 28029 Madrid , Spain
- Department of Materials Science, EEBE , Technical University of Catalonia (UPC) , d'Eduard Maristany 16 , 08019 Barcelona , Spain
| | - Humberto Palza
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas , Universidad de Chile , Beauchef 851 , 8370456 Santiago , Chile
- Millennium Nuclei in Soft Smart Mechanical Metamaterials , Beauchef 851 , 8370456 Santiago , Chile
| |
Collapse
|
14
|
Liu X, Wang L, Ma C, Wang G, Zhang Y, Sun S. Exosomes derived from platelet-rich plasma present a novel potential in alleviating knee osteoarthritis by promoting proliferation and inhibiting apoptosis of chondrocyte via Wnt/β-catenin signaling pathway. J Orthop Surg Res 2019; 14:470. [PMID: 31888697 PMCID: PMC6936129 DOI: 10.1186/s13018-019-1529-7] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/19/2019] [Indexed: 03/20/2023] Open
Abstract
Background Platelet-rich plasma (PRP) provides a nonsurgical approach for treating osteoarthritis (OA). Exosomes that play vital roles in intercellular communication have been studied extensively. Here, we investigated the therapeutic potential and molecular mechanism of exosomes derived from PRP (PRP-Exos) in alleviating OA. Methods Exosomes derived from PRP(PRP-Exos) were isolated and purified using the exoEasy Maxi Kit and then identified and analyzed. Primary rabbit chondrocytes were isolated and treated with interleukin 1 beta (IL-1β) to establish the OA model in vitro. Proliferation, migration, and apoptosis assays were measured and compared between PRP-Exos and activated PRP (PRP-As) to evaluate the therapeutic effects on OA. The mechanism involving the Wnt/β-catenin signaling pathway was investigated by Western blot analysis. In vivo, we established animal knee OA model by surgery to compare the therapeutic effect of PRP-Exos and PRP-As. Results We successfully isolated and purified exosomes from PRP using the exoEasy Maxi Kit. We also isolated and identified chondrocytes from the New Zealand white rabbit and established the IL-1β-induced OA model; meanwhile, PRP-Exos and PRP-As both inhibited the release of tumor necrosis factor-α(TNF-α) and there was no statistically significant difference between the two. In proliferation, migration, scratch assay, the promoting effect of PRP-Exos was significantly more better than PRP-As. Furthermore, PRP-Exos could significantly decreased apoptotic rate of OA chondrocyte compared with PRP-As. In Western blot analysis, the expression of β-catenin, and RUNX2, Wnt5a were increased in IL-1β-treated chondrocytes, but PRP-Exos and PRP-As could both reverse these changes, and the reversal effect of the former was better than the latter. In vivo, we found that both PRP-Exos and PRP-As displayed the progression of OA, and the effect of PRP-Exos was obviously better than PRP-As by chondrocyte count and Osteoarthritis Research Society International (OARSI) scoring system. Conclusion The therapeutic effects of PRP-Exos on OA were similar or better compared with those of PRP-As in vitro or in vivo. PRP-Exos acting as carriers containing growth factors derived from PRP present a novel therapy for OA by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xuchang Liu
- Department of Emergency Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.,School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Taian, 271016, Shandong, China
| | - Lubo Wang
- Department of Trauma Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Chengshan Ma
- Department of Emergency Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Guozong Wang
- School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China
| | - Yuanji Zhang
- School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
15
|
Propolis Reduces the Expression of Autophagy-Related Proteins in Chondrocytes under Interleukin-1β Stimulus. Int J Mol Sci 2019; 20:ijms20153768. [PMID: 31374866 PMCID: PMC6695581 DOI: 10.3390/ijms20153768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Osteoarthritis (OA) is a progressive and multifactorial disease that is associated with aging. A number of changes occur in aged cartilage, such as increased oxidative stress, decreased markers of healthy cartilage, and alterations in the autophagy pathway. Propolis extracts contain a mixture of polyphenols and it has been proved that they have high antioxidant capacity and could regulate the autophagic pathway. Our objective was to evaluate the effect of ethanolic extract of propolis (EEP) on chondrocytes that were stimulated with IL-1β. Methods: Rabbit chondrocytes were isolated and stimulated with IL-1β and treated with EEP. We evaluated cell viability, nitric oxide production, healthy cartilage, and OA markers, and the expression of three proteins associated with the autophagy pathway LC3, ATG5, and AKT1. Results: The EEP treatment reduces the expression of LC3, ATG5, and AKT1, reduces the production of nitric oxide, increases the expression of healthy markers, and reduces OA markers. Conclusions: These results suggest that treatment with EEP in chondrocytes that were stimulated with IL-1β has beneficial effects, such as a decrease in the expression of proteins associated with autophagy, MMP13, and production of nitric oxide, and also increased collagen II.
Collapse
|
16
|
Sahu N, Viljoen HJ, Subramanian A. Continuous low-intensity ultrasound attenuates IL-6 and TNFα-induced catabolic effects and repairs chondral fissures in bovine osteochondral explants. BMC Musculoskelet Disord 2019; 20:193. [PMID: 31054572 PMCID: PMC6499975 DOI: 10.1186/s12891-019-2566-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/11/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cartilage repair outcomes are compromised in a pro-inflammatory environment; therefore, the mitigation of pro-inflammatory responses is beneficial. Treatment with continuous low-intensity ultrasound (cLIUS) at the resonant frequency of 5 MHz is proposed for the repair of chondral fissures under pro-inflammatory conditions. METHODS Bovine osteochondral explants, concentrically incised to create chondral fissures, were maintained under cLIUS (14 kPa (5 MHz, 2.5 Vpp), 20 min, 4 times/day) for a period of 28 days in the presence or absence of cytokines, interleukin-6 (IL-6) or tumor necrosis factor (TNF)α. Outcome assessments included histological and immunohistochemical staining of the explants; and the expression of catabolic and anabolic genes by qRT-PCR in bovine chondrocytes. Cell migration was assessed by scratch assays, and by visualizing migrating cells into the hydrogel core of cartilage-hydrogel constructs. RESULTS Both in the presence and absence of cytokines, higher percent apposition along with closure of fissures were noted in cLIUS-stimulated explants as compared to non-cLIUS-stimulated explants on day 14. On day 28, the percent apposition was not significantly different between unstimulated and cLIUS-stimulated explants exposed to cytokines. As compared to non-cLIUS-stimulated controls, on day 28, cLIUS preserved the distribution of proteoglycans and collagen II in explants despite exposure to cytokines. cLIUS enhanced the cell migration irrespective of cytokine treatment. IL-6 or TNFα-induced increases in MMP13 and ADAMTS4 gene expression was rescued by cLIUS stimulation in chondrocytes. Under cLIUS, TNFα-induced increase in NF-κB expression was suppressed, and the expression of collagen II and TIMP1 genes were upregulated. CONCLUSION cLIUS repaired chondral fissures, and elicited pro-anabolic and anti-catabolic effects, thus demonstrating the potential of cLIUS in improving cartilage repair outcomes.
Collapse
Affiliation(s)
- Neety Sahu
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588-0643, USA
| | - Hendrik J Viljoen
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588-0643, USA
| | - Anuradha Subramanian
- Department of Chemical and Materials Engineering, University of Alabama at Huntsville, Huntsville, Alabama, 35899, USA.
| |
Collapse
|
17
|
Janssen JN, Batschkus S, Schimmel S, Bode C, Schminke B, Miosge N. The Influence of TGF-β3, EGF, and BGN on SOX9 and RUNX2 Expression in Human Chondrogenic Progenitor Cells. J Histochem Cytochem 2018; 67:117-127. [PMID: 30431382 DOI: 10.1369/0022155418811645] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease and leads to the degradation of the extracellular matrix by an imbalance between anabolic and catabolic processes. TGF-β3 (transforming growth factor beta-3) and epidermal growth factor (EGF) influence the osteochondrogenic potential of chondrocytes. In this study, we compared the expression of mediators and receptors in the TGF-β3 and EGF pathways, as well as biglycan (BGN), in healthy and diseased chondrocytes. Furthermore, we used chondrogenic progenitor cells (CPCs) for in vitro stimulation and knockdown experiments to elucidate the effects of TGF-β3 and EGF on the chondrogenic potential. Our results demonstrate that the expression of TGF-beta receptor type-1 (TGFBRI) and epidermal growth factor receptor (EGFR) is altered in diseased chondrocytes as well as in CPCs. Moreover, TGF-β3 and EGF stimulation influenced the expression levels of BGN, SRY (sex determining region Y)-box 9 (SOX9), and Runt-related transcription factor 2 (RUNX2) in CPCs. Therefore, changes in TGFBRI and EGFR expression likely contribute to the degenerative and regenerative effects seen in late stages of OA.
Collapse
Affiliation(s)
| | | | - Stefan Schimmel
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Christa Bode
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medical Center, Göttingen, Germany
| | - Nicolai Miosge
- Tissue Regeneration Work Group, Department of Prosthodontics
| |
Collapse
|
18
|
Vinod E, Boopalan PRJVC, Sathishkumar S. Reserve or Resident Progenitors in Cartilage? Comparative Analysis of Chondrocytes versus Chondroprogenitors and Their Role in Cartilage Repair. Cartilage 2018; 9:171-182. [PMID: 29047310 PMCID: PMC5871122 DOI: 10.1177/1947603517736108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction Articular cartilage is made up of hyaline tissue embodying chondrocytes, which arise from mesenchymal stromal cells (MSCs) and specialized extracellular matrix. Despite possessing resident progenitors in and around the joint primed for chondrogenesis, cartilage has limited intrinsic capacity of repair and cell turnover. Advances in isolation, culture, and characterization of these progenitors have raised the possibility for their use in cell-based cartilage repair. Chondroprogenitors (CPCs) have been classified as MSCs and have been postulated to play a vital role in injury response and are identified by their colony forming ability, proliferative potential, telomere dynamics, multipotency, and expression of stem cell markers. The combined presence of CPCs and chondrocytes within the same tissue compartments and the ability of chondrocytes to dedifferentiate and acquire stemness during culture expansion has obscured our ability to define and provide clear-cut differences between these 2 cell populations. Objective This review aims to evaluate and summarize the available literature on CPCs in terms of their origin, growth kinetics, molecular characteristics, and differential and therapeutic potential with emphasis on their difference from daughter chondrocytes. Design For this systematic review, a comprehensive electronic search was performed on PubMed and Google Scholar using relevant terms such as chondrocytes, chondroprogenitors, and surface marker expression. Results and Conclusion Our comparative analysis shows that there is an ill-defined distinction between CPCs and chondrocytes with respect to their cell surface expression (MSC markers and CPC-specific markers) and differentiation potential. Accumulating evidence indicates that the 2 subpopulations may be distinguished based on their growth kinetics and chondrogenic marker.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India
| | - P. R. J. V. C. Boopalan
- Department of Orthopaedics, Christian Medical College/Center for Stem Cell Research, Vellore, India,P. R. J. V. C. Boopalan, Department of Orthopaedics, Centre for Stem Cell Research, Christian Medical College & Hospital, Vellore 632002, India.
| | | |
Collapse
|
19
|
Zellner J, Pattappa G, Koch M, Lang S, Weber J, Pfeifer CG, Mueller MB, Kujat R, Nerlich M, Angele P. Autologous mesenchymal stem cells or meniscal cells: what is the best cell source for regenerative meniscus treatment in an early osteoarthritis situation? Stem Cell Res Ther 2017; 8:225. [PMID: 29017608 PMCID: PMC5634903 DOI: 10.1186/s13287-017-0678-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/11/2017] [Accepted: 09/21/2017] [Indexed: 01/05/2023] Open
Abstract
Background Treatment of meniscus tears within the avascular region represents a significant challenge, particularly in a situation of early osteoarthritis. Cell-based tissue engineering approaches have shown promising results. However, studies have not found a consensus on the appropriate autologous cell source in a clinical situation, specifically in a challenging degenerative environment. The present study sought to evaluate the appropriate cell source for autologous meniscal repair in a demanding setting of early osteoarthritis. Methods A rabbit model was used to test autologous meniscal repair. Bone marrow and medial menisci were harvested 4 weeks prior to surgery. Bone marrow-derived mesenchymal stem cells (MSCs) and meniscal cells were isolated, expanded, and seeded onto collagen-hyaluronan scaffolds before implantation. A punch defect model was performed on the lateral meniscus and then a cell-seeded scaffold was press-fit into the defect. Following 6 or 12 weeks, gross joint morphology and OARSI grade were assessed, and menisci were harvested for macroscopic, histological, and immunohistochemical evaluation using a validated meniscus scoring system. In conjunction, human meniscal cells isolated from non-repairable bucket handle tears and human MSCs were expanded and, using the pellet culture model, assessed for their meniscus-like potential in a translational setting through collagen type I and II immunostaining, collagen type II enzyme-linked immunosorbent assay (ELISA), and gene expression analysis. Results After resections of the medial menisci, all knees showed early osteoarthritic changes (average OARSI grade 3.1). However, successful repair of meniscus punch defects was performed using either meniscal cells or MSCs. Gross joint assessment demonstrated donor site morbidity for meniscal cell treatment. Furthermore, human MSCs had significantly increased collagen type II gene expression and production compared to meniscal cells (p < 0.05). Conclusions The regenerative potential of the meniscus by an autologous cell-based tissue engineering approach was shown even in a challenging setting of early osteoarthritis. Autologous MSCs and meniscal cells were found to have improved meniscal healing in an animal model, thus demonstrating their feasibility in a clinical setting. However, donor site morbidity, reduced availability, and reduced chondrogenic differentiation of human meniscal cells from debris of meniscal tears favors autologous MSCs for clinical use for cell-based meniscus regeneration.
Collapse
Affiliation(s)
- Johannes Zellner
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany.
| | - Girish Pattappa
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Matthias Koch
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Siegmund Lang
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Johannes Weber
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Christian G Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Michael B Mueller
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Richard Kujat
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Michael Nerlich
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany
| | - Peter Angele
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93042, Regensburg, Germany.,Sporthopaedicum Regensburg, Hildegard von Bingen Strasse 1, 93053, Regensburg, Germany
| |
Collapse
|
20
|
Zhang M, Yang H, Lu L, Wan X, Zhang J, Zhang H, Liu X, Huang X, Xiao G, Wang M. Matrix replenishing by BMSCs is beneficial for osteoarthritic temporomandibular joint cartilage. Osteoarthritis Cartilage 2017; 25:1551-1562. [PMID: 28532603 DOI: 10.1016/j.joca.2017.05.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 04/24/2017] [Accepted: 05/03/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVES The present goal was to explore whether matrix replenishment is the primary requirement for osteoarthritic (OA) cartilage. METHODS Cells isolated from the superficial and deep zone cartilage of a pig temporomandibular joint (TMJ) were exposed to fluid flow shear stress (FFSS). Differences in matrix production and cellular differentiation were detected. Unilateral anterior crossbite (UAC) was applied to C57BL/6J female mice. Green fluorescent protein-labeled exogenous bone marrow stromal cells (GFP-BMSCs) were injected weekly into TMJs, starting from 3 weeks of UAC stimulation and continuing for 4-, 8- and 12-weeks. Another GFP-BMSCs injection UAC group stopped receiving injections for 4-weeks after 8-weeks of injections. Assessments were focused on morphological alterations in UAC mouse TMJ cartilage, the expression levels of DAP3, an anoikis marker, CD163, a scavenger receptor family member, and ki67, a proliferation indicator. RESULTS FFSS down-regulated type-II collagen expression but stimulated terminal differentiation in cells isolated from deep zone cartilage. It down-regulated aggrecan expression but up-regulated type I collagen in cells isolated from both superficial and deep zones. UAC caused matrix loss and anoikis and enhanced scavenging activity in deep zone chondrocytes without affecting cell proliferation. Superficial fibrillation was obvious in the late stage. Weekly injections of BMSCs largely restored these changes. The implanted BMSCs expressed a high level of CD163 protein but did not show remarkable cell proliferation. Terminating the supply of exogenous BMSCs reversed the restorative effects. CONCLUSIONS Scavenging the degraded matrix and replenishing the fibrosis-developmental matrix are the primary requirements for the repair of OA cartilage.
Collapse
Affiliation(s)
- M Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - H Yang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - L Lu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - X Wan
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - J Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - H Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - X Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China
| | - X Huang
- Department of Biology, The Fourth Military Medical University, 17 Changle West Road, Xi'an, China
| | - G Xiao
- Department of Biology, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China; Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - M Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xi'an, China.
| |
Collapse
|
21
|
Phull AR, Eo SH, Abbas Q, Ahmed M, Kim SJ. Applications of Chondrocyte-Based Cartilage Engineering: An Overview. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1879837. [PMID: 27631002 PMCID: PMC5007317 DOI: 10.1155/2016/1879837] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 06/24/2016] [Accepted: 06/26/2016] [Indexed: 12/31/2022]
Abstract
Chondrocytes are the exclusive cells residing in cartilage and maintain the functionality of cartilage tissue. Series of biocomponents such as different growth factors, cytokines, and transcriptional factors regulate the mesenchymal stem cells (MSCs) differentiation to chondrocytes. The number of chondrocytes and dedifferentiation are the key limitations in subsequent clinical application of the chondrocytes. Different culture methods are being developed to overcome such issues. Using tissue engineering and cell based approaches, chondrocytes offer prominent therapeutic option specifically in orthopedics for cartilage repair and to treat ailments such as tracheal defects, facial reconstruction, and urinary incontinence. Matrix-assisted autologous chondrocyte transplantation/implantation is an improved version of traditional autologous chondrocyte transplantation (ACT) method. An increasing number of studies show the clinical significance of this technique for the chondral lesions treatment. Literature survey was carried out to address clinical and functional findings by using various ACT procedures. The current study was conducted to study the pharmacological significance and biomedical application of chondrocytes. Furthermore, it is inferred from the present study that long term follow-up studies are required to evaluate the potential of these methods and specific positive outcomes.
Collapse
Affiliation(s)
- Abdul-Rehman Phull
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| | - Seong-Hui Eo
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| | - Qamar Abbas
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| | - Madiha Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| |
Collapse
|
22
|
Verdonk R, Madry H, Shabshin N, Dirisamer F, Peretti GM, Pujol N, Spalding T, Verdonk P, Seil R, Condello V, Di Matteo B, Zellner J, Angele P. The role of meniscal tissue in joint protection in early osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2016; 24:1763-74. [PMID: 27085362 DOI: 10.1007/s00167-016-4069-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/23/2016] [Indexed: 01/05/2023]
Abstract
It is widely accepted that partial meniscectomy leads to early onset of osteoarthritis (OA). A strong correlation exists between the amount and location of the resected meniscus and the development of degenerative changes in the knee. On the other hand, osteoarthritic changes of the joint alter the structural and functional integrity of meniscal tissue. These alterations might additionally compromise the limited healing capacity of the meniscus. In young, active patients without cartilage damage, meniscus therapy including partial meniscectomy, meniscus suture, and meniscus replacement has proven beneficial effects in long-term studies. Even in an early osteoarthritic milieu, there is a relevant regenerative potential of the meniscus and the surrounding cartilage. This potential should be taken into account, and meniscal surgery can be performed with the correct timing and the proper indication even in the presence of early OA.
Collapse
Affiliation(s)
- Rene Verdonk
- Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Henning Madry
- Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrberger Strasse 100, Building 37-38, 66421, Homburg, Saarland, Germany
| | - Nogah Shabshin
- Department of Radiology, Carmel Medical Center, Haifa, Israel.,Department of Radiology, Hospital of University of Pennsylvania, Philadelphia, PA, USA
| | - Florian Dirisamer
- Orthopädie und Sportchirurgie, Schloss Puchenau, Karl-Leitl-Str. 1, 4048, Linz-Puchenau, Austria
| | - Giuseppe M Peretti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Nicolas Pujol
- Centre hospitalier de Versailles, 177, rue de Versailles, 78150, Le Chesnay, France
| | - Tim Spalding
- Department of Orthopaedics, University Hospital of Coventry and Warwickshire, Rugby, UK
| | - Peter Verdonk
- Antwerp Orthopedic Center, Monica Hospitals, Antwerp, Belgium
| | - Romain Seil
- Clinique d'Eich and Sports Medicine Research Laboratory, Department of Orthopaedic Surgery, Centre Hospitalier Luxembourg, Luxembourg Institute of Health, 78 rue d'Eich, 1460, Luxembourg, Luxembourg
| | - Vincenzo Condello
- Dipartimento di Ortopedia - Responsabile di Struttura Semplice di Traumatologia dello, Sport Knee Surgery and Sports Traumatology Ospedale Sacro Cuore - Don Calabria Via Don, Sempreboni, 5, 37024, Negrar Verona, Italy
| | - Berardo Di Matteo
- II Orthopaedic Clinic and Biomechanics Lab, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Johannes Zellner
- Department of Trauma Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany
| | - Peter Angele
- Department of Trauma Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany. .,Sporthopaedicum Regensburg, Hildegard von Bingen Strasse 1, 93053, Regensburg, Germany.
| |
Collapse
|
23
|
Cheng A, Gustafson AR, Schaner Tooley CE, Zhang M. BMP-9 dependent pathways required for the chondrogenic differentiation of pluripotent stem cells. Differentiation 2016; 92:298-305. [PMID: 27056281 DOI: 10.1016/j.diff.2016.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/17/2016] [Accepted: 03/29/2016] [Indexed: 01/20/2023]
Abstract
Current cartilage repair therapies focus on the delivery of chondrocytes differentiated from mesenchymal stem cells, and thus understanding the factors that promote chondrogenesis may lead to improved therapies. Several bone morphogenetic proteins (BMPs) have been implicated in chondrogenic differentiation and/or chondrocyte function. Although the signaling pathways downstream of BMPs have been studied in other systems, their role in chondrogenesis is less well characterized. Here, we investigated the effects of BMP-9 in chondroprogenitor cells. Compared to BMP-2 and BMP-6, we showed that BMP-9 was significantly more potent in inducing chondrogenic differentiation in mouse C3H10T1/2 and ATDC5 cells. Moreover, we demonstrated that BMP-9 induces the phosphorylation of SMAD1/5 in a dose and time dependent manner. Confocal immunofluorescence microscopy further demonstrated an accumulation of phosphorylated SMAD1/5 in the nuclei of BMP-9 treated cells. Consistent with activation of the SMAD signaling pathway, we also observed an up-regulation of Id1 and PAI-I expression. Importantly, we demonstrated that the simultaneous knockdown of SMAD1 and SMAD5 was able to inhibit chondrogenesis. Additionally, we also observed activation of p38 by BMP-9, and pharmacological inhibition of this pathway blocked chondrogenesis. In contrast, inhibition of p44/42 ERK had no effect. Finally, we tested the ability of Noggin to block the actions of BMP-9. While Noggin potently inhibited the ability of BMP-2 to mediate differentiation, it had no significant effect on BMP-9. Our findings provide a clearer understanding of the cellular pathways utilized by BMP-9 for chondrogenesis that may help improve current therapies for regenerative cartilage repair.
Collapse
Affiliation(s)
- Alan Cheng
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | - Amber Renee Gustafson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | - Mei Zhang
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| |
Collapse
|
24
|
Lowenstine LJ, McManamon R, Terio KA. Comparative Pathology of Aging Great Apes: Bonobos, Chimpanzees, Gorillas, and Orangutans. Vet Pathol 2015; 53:250-76. [PMID: 26721908 DOI: 10.1177/0300985815612154] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The great apes (chimpanzees, bonobos, gorillas, and orangutans) are our closest relatives. Despite the many similarities, there are significant differences in aging among apes, including the human ape. Common to all are dental attrition, periodontitis, tooth loss, osteopenia, and arthritis, although gout is uniquely human and spondyloarthropathy is more prevalent in apes than humans. Humans are more prone to frailty, sarcopenia, osteoporosis, longevity past reproductive senescence, loss of brain volume, and Alzheimer dementia. Cerebral vascular disease occurs in both humans and apes. Cardiovascular disease mortality increases in aging humans and apes, but coronary atherosclerosis is the most significant type in humans. In captive apes, idiopathic myocardial fibrosis and cardiomyopathy predominate, with arteriosclerosis of intramural coronary arteries. Similar cardiac lesions are occasionally seen in wild apes. Vascular changes in heart and kidneys and aortic dissections in gorillas and bonobos suggest that hypertension may be involved in pathogenesis. Chronic kidney disease is common in elderly humans and some aging apes and is linked with cardiovascular disease in orangutans. Neoplasms common to aging humans and apes include uterine leiomyomas in chimpanzees, but other tumors of elderly humans, such as breast, prostate, lung, and colorectal cancers, are uncommon in apes. Among the apes, chimpanzees have been best studied in laboratory settings, and more comparative research is needed into the pathology of geriatric zoo-housed and wild apes. Increasing longevity of humans and apes makes understanding aging processes and diseases imperative for optimizing quality of life in all the ape species.
Collapse
Affiliation(s)
- L J Lowenstine
- Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA Mountain Gorilla Veterinary Project-Gorilla Doctors, Karen C. Drayer Wildlife Health Center, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - R McManamon
- Zoo and Exotic Animal Pathology Service, Infectious Diseases Laboratory, Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - K A Terio
- Zoological Pathology Program, University of Illinois College of Veterinary Medicine, Maywood, IL, USA
| |
Collapse
|
25
|
Mesenchymal stem cells in regenerative medicine: Focus on articular cartilage and intervertebral disc regeneration. Methods 2015; 99:69-80. [PMID: 26384579 DOI: 10.1016/j.ymeth.2015.09.015] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 08/10/2015] [Accepted: 09/15/2015] [Indexed: 01/15/2023] Open
Abstract
Musculoskeletal disorders represent a major cause of disability and morbidity globally and result in enormous costs for health and social care systems. Development of cell-based therapies is rapidly proliferating in a number of disease areas, including musculoskeletal disorders. Novel biological therapies that can effectively treat joint and spine degeneration are high priorities in regenerative medicine. Mesenchymal stem cells (MSCs) isolated from bone marrow (BM-MSCs), adipose tissue (AD-MSCs) and umbilical cord (UC-MSCs) show considerable promise for use in cartilage and intervertebral disc (IVD) repair. This review article focuses on stem cell-based therapeutics for cartilage and IVD repair in the context of the rising global burden of musculoskeletal disorders. We discuss the biology MSCs and chondroprogenitor cells and specifically focus on umbilical cord/Wharton's jelly derived MSCs and examine their potential for regenerative applications. We also summarize key components of the molecular machinery and signaling pathways responsible for the control of chondrogenesis and explore biomimetic scaffolds and biomaterials for articular cartilage and IVD regeneration. This review explores the exciting opportunities afforded by MSCs and discusses the challenges associated with cartilage and IVD repair and regeneration. There are still many technical challenges associated with isolating, expanding, differentiating, and pre-conditioning MSCs for subsequent implantation into degenerate joints and the spine. However, the prospect of combining biomaterials and cell-based therapies that incorporate chondrocytes, chondroprogenitors and MSCs leads to the optimistic view that interdisciplinary approaches will lead to significant breakthroughs in regenerating musculoskeletal tissues, such as the joint and the spine in the near future.
Collapse
|
26
|
Hopper N, Henson F, Brooks R, Ali E, Rushton N, Wardale J. Peripheral blood derived mononuclear cells enhance osteoarthritic human chondrocyte migration. Arthritis Res Ther 2015; 17:199. [PMID: 26249339 PMCID: PMC4528856 DOI: 10.1186/s13075-015-0709-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 07/07/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction A major problem in cartilage repair is the lack of chondrogenic cells migrating from healthy tissue into defects. Cartilage is essentially avascular and therefore its healing is not considered to involve mononuclear cells. Peripheral blood derived mononuclear cells (PBMC) offer a readily available autologous cell source for clinical use and therefore this study was designed to evaluate the effects of PBMCs on chondrocytes and cartilage. Methods Human primary chondrocytes and cartilage tissue explants were taken from patients undergoing total knee replacement (n = 17). Peripheral blood samples were obtained from healthy volunteers (n = 12) and mononuclear cells were isolated by density-gradient centrifugation. Cell migration and chemokinetic potential were measured using a scratch assay, xCELLigence and CyQuant assay. PCR array and quantitative PCR was used to evaluate mRNA expression of 87 cell motility and/or chondrogenic genes. Results The chondrocyte migration rate was 2.6 times higher at 3 hour time point (p < 0.0001) and total number of migrating chondrocytes was 9.7 times higher (p < 0.0001) after three day indirect PBMC stimulus and 8.2 times higher (p < 0.0001) after three day direct co-culture with PBMCs. A cartilage explant model confirmed that PBMCs also exert a chemokinetic role on ex vivo tissue. PBMC stimulation was found to significantly upregulate the mRNA levels of 2 chondrogenic genes; collagen type II (COL2A1 600–fold, p < 0.0001) and SRY box 9 (SOX9 30–fold, p < 0.0001) and the mRNA levels of 7 genes central in cell motility and migration were differentially regulated by 24h PBMC stimulation. Conclusion The results support the concept that PBMC treatment enhances chondrocyte migration without suppressing the chondrogenic phenotype possibly via mechanistic pathways involving MMP9 and IGF1. In the future, peripheral blood mononuclear cells could be used as an autologous point-ofcare treatment to attract native chondrocytes from the diseased tissue to aid in cartilage repair.
Collapse
Affiliation(s)
- Niina Hopper
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, BC2 0QQ, Cambridge, UK.
| | - Frances Henson
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK.
| | - Roger Brooks
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, BC2 0QQ, Cambridge, UK.
| | - Erden Ali
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, BC2 0QQ, Cambridge, UK.
| | - Neil Rushton
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, BC2 0QQ, Cambridge, UK.
| | - John Wardale
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, BC2 0QQ, Cambridge, UK.
| |
Collapse
|
27
|
Mardones R, Jofré CM, Minguell JJ. Cell Therapy and Tissue Engineering Approaches for Cartilage Repair and/or Regeneration. Int J Stem Cells 2015; 8:48-53. [PMID: 26019754 PMCID: PMC4445709 DOI: 10.15283/ijsc.2015.8.1.48] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 05/04/2015] [Indexed: 01/27/2023] Open
Abstract
Articular cartilage injuries caused by traumatic, mechanical and/or by progressive degeneration result in pain, swelling, subsequent loss of joint function and finally osteoarthritis. Due to the peculiar structure of the tissue (no blood supply), chondrocytes, the unique cellular phenotype in cartilage, receive their nutrition through diffusion from the synovial fluid and this limits their intrinsic capacity for healing. The first cellular avenue explored for cartilage repair involved the in situ transplantation of isolated chondrocytes. Latterly, an improved alternative for the above reparative strategy involved the infusion of mesenchymal stem cells (MSC), which in addition to a self-renewal capacity exhibit a differentiation potential to chondrocytes, as well as a capability to produce a vast array of growth factors, cytokines and extracellular matrix compounds involved in cartilage development. In addition to the above and foremost reparative options up till now in use, other therapeutic options have been developed, comprising the design of biomaterial substrates (scaffolds) capable of sustaining MSC attachment, proliferation and differentiation. The implantation of these engineered platforms, closely to the site of cartilage damage, may well facilitate the initiation of an ‘in situ’ cartilage reparation process. In this mini-review, we examined the timely and conceptual development of several cell-based methods, designed to repair/regenerate a damaged cartilage. In addition to the above described cartilage reparative options, other therapeutic alternatives still in progress are portrayed.
Collapse
Affiliation(s)
- Rodrigo Mardones
- Centro de Traumatología y Ortopedia, Laboratorio de Ingeniería de Tejidos, Clínica Las Condes, Santiago, Chile ; Centro de Terapia Regenerativa Celular, Laboratorio de Ingeniería de Tejidos, Clínica Las Condes, Santiago, Chile
| | - Claudio M Jofré
- Centro de Terapia Regenerativa Celular, Laboratorio de Ingeniería de Tejidos, Clínica Las Condes, Santiago, Chile
| | - José J Minguell
- Centro de Terapia Regenerativa Celular, Laboratorio de Ingeniería de Tejidos, Clínica Las Condes, Santiago, Chile
| |
Collapse
|
28
|
Zellner J, Mueller M, Xin Y, Krutsch W, Brandl A, Kujat R, Nerlich M, Angele P. Dynamic hydrostatic pressure enhances differentially the chondrogenesis of meniscal cells from the inner and outer zone. J Biomech 2015; 48:1479-84. [PMID: 25698240 DOI: 10.1016/j.jbiomech.2015.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 02/02/2015] [Indexed: 01/24/2023]
Abstract
This study analyses the influence of dynamic hydrostatic pressure on chondrogenesis of human meniscus-derived fibrochondrocytes and explores the differences in chondrogenic differentiation under loading conditions between cells derived from the avascular inner zone and vascularized outer region of the meniscus. Aggregates of human fibrochondrocytes with cell origin from the inner region or with cell origin from the outer region were generated. From the two groups of either cell origin, aggregates were treated with dynamic hydrostatic pressure (1Hz for 4h; 0.55-5.03MPa, cyclic sinusoidal) from day 1 to day 7. The other aggregates served as unloaded controls. At day 0, 7, 14 and 21 aggregates were harvested for evaluation including histology, immunostaining and ELISA analysis for glycosaminoglycan (GAG) and collagen II. Loaded aggregates were found to be macroscopically larger and revealed immunohistochemically enhanced chondrogenesis compared to the corresponding controls. Loaded or non-loaded meniscal cells from the outer zone showed a higher potential and earlier onset of chondrogenesis compared to the cells from the inner part of the meniscus. This study suggests that intrinsic factors like cell properties in the different areas of the meniscus and their reaction on mechanical load might play important roles in designing Tissue Engineering strategies for meniscal repair in vivo.
Collapse
Affiliation(s)
- J Zellner
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany.
| | - M Mueller
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| | - Y Xin
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| | - W Krutsch
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| | - A Brandl
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| | - R Kujat
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| | - M Nerlich
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| | - P Angele
- University Hospital of Regensburg, Department of Trauma Surgery, Regensburg, Germany
| |
Collapse
|