1
|
Shen J, Ye D, Jin H, Wu Y, Peng L, Liang Y. Porcine nasal septum cartilage-derived decellularized matrix promotes chondrogenic differentiation of human umbilical mesenchymal stem cells without exogenous growth factors. J Mater Chem B 2024; 12:5513-5524. [PMID: 38745541 DOI: 10.1039/d3tb03077f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
BACKGROUND In the domain of plastic surgery, nasal cartilage regeneration is of significant importance. The extracellular matrix (ECM) from porcine nasal septum cartilage has shown potential for promoting human cartilage regeneration. Nonetheless, the specific biological inductive factors and their pathways in cartilage tissue engineering remain undefined. METHODS The decellularized matrix derived from porcine nasal septum cartilage (PN-DCM) was prepared using a grinding method. Human umbilical cord mesenchymal stem cells (HuMSCs) were cultured on these PN-DCM scaffolds for 4 weeks without exogenous growth factors to evaluate their chondroinductive potential. Subsequently, proteomic analysis was employed to identify potential biological inductive factors within the PN-DCM scaffolds. RESULTS Compared to the TGF-β3-cultured pellet model serving as a positive control, the PN-DCM scaffolds promoted significant deposition of a Safranin-O positive matrix and Type II collagen by HuMSCs. Gene expression profiling revealed upregulation of ACAN, COL2A1, and SOX9. Proteomic analysis identified potential chondroinductive factors in the PN-DCM scaffolds, including CYTL1, CTGF, MGP, ITGB1, BMP7, and GDF5, which influence HuMSC differentiation. CONCLUSION Our findings have demonstrated that the PN-DCM scaffolds promoted HuMSC differentiation towards a nasal chondrocyte phenotype without the supplementation of exogenous growth factors. This outcome is associated with the chondroinductive factors present within the PN-DCM scaffolds.
Collapse
Affiliation(s)
- Jinpeng Shen
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou, P. R. China.
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, P. R. China.
- Department of Plastic Surgery, Taizhou Enze Medical Center, Zhejiang, P. R. China
| | - Danyan Ye
- Research Center for Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P. R. China
| | - Hao Jin
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P. R. China
| | - Yongxuan Wu
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, P. R. China.
| | - Lihong Peng
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, P. R. China.
| | - Yan Liang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou, P. R. China.
| |
Collapse
|
2
|
Zhang Y, Sheng R, Chen J, Wang H, Zhu Y, Cao Z, Zhao X, Wang Z, Liu C, Chen Z, Zhang P, Kuang B, Zheng H, Shen C, Yao Q, Zhang W. Silk Fibroin and Sericin Differentially Potentiate the Paracrine and Regenerative Functions of Stem Cells Through Multiomics Analysis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210517. [PMID: 36915982 DOI: 10.1002/adma.202210517] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/08/2023] [Indexed: 05/19/2023]
Abstract
Silk fibroin (SF) and sericin (SS), the two major proteins of silk, are attractive biomaterials with great potential in tissue engineering and regenerative medicine. However, their biochemical interactions with stem cells remain unclear. In this study, multiomics are employed to obtain a global view of the cellular processes and pathways of mesenchymal stem cells (MSCs) triggered by SF and SS to discern cell-biomaterial interactions at an in-depth, high-throughput molecular level. Integrated RNA sequencing and proteomic analysis confirm that SF and SS initiate widespread but distinct cellular responses and potentiate the paracrine functions of MSCs that regulate extracellular matrix deposition, angiogenesis, and immunomodulation through differentially activating the integrin/PI3K/Akt and glycolysis signaling pathways. These paracrine signals of MSCs stimulated by SF and SS effectively improve skin regeneration by regulating the behavior of multiple resident cells (fibroblasts, endothelial cells, and macrophages) in the skin wound microenvironment. Compared to SS, SF exhibits better immunomodulatory effects in vitro and in vivo, indicating its greater potential as a carrier material of MSCs for skin regeneration. This study provides comprehensive and reliable insights into the cellular interactions with SF and SS, enabling the future development of silk-based therapeutics for tissue engineering and stem cell therapy.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Renwang Sheng
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing, 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Hongmei Wang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yue Zhu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhicheng Cao
- School of Medicine, Southeast University, Nanjing, 210009, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Xinyi Zhao
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhimei Wang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Chuanquan Liu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Po Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Baian Kuang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Haotian Zheng
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chuanlai Shen
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Qingqiang Yao
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
3
|
Eom YS, Ko BS, Shah FH, Kim SJ. E3 Ubiquitin Ligase Constitutive Photomorphogenic 1 Regulates Differentiation and Inflammation via MAPK Signaling Pathway in Rabbit Articular Chondrocytes. DNA Cell Biol 2023; 42:239-247. [PMID: 36940307 DOI: 10.1089/dna.2022.0664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
Abstract
Constitutive photomorphogenic 1 (COP1), is an E3 ubiquitin ligase that plays a role in the regulation of various cellular processes including cell growth, differentiation, and survival in mammals. In certain conditions such as overexpression or loss of function, COP1 acts either as an oncogenic protein or as a tumor suppressor by targeting specific proteins for ubiquitination-mediated degradation. However, the precise role of COP1 has not been well studied in primary articular chondrocytes. In this study, we investigated the role of COP1 in chondrocyte differentiation. Western blotting and reverse transcription-polymerase chain reaction analysis demonstrated that COP1 overexpression reduced type II collagen expression, promoted cyclooxygenase 2 (COX-2) expression, and reduced sulfated proteoglycan synthesis, as detected by Alcian blue staining. Upon siRNA treatment, revived type II collagen, sulfated proteoglycan production, and decreased COX-2 expression. Phosphorylation of p38 kinase and ERK-1/-2 signaling pathways was regulated by COP1 upon cDNA and siRNA transfection in chondrocytes. The inhibition of the p38 kinase and ERK-1/-2 signaling pathways with SB203580 and PD98059 ameliorated the expression of type II collagen and COX-2 in transfected chondrocytes, thus suggesting that COP1 regulates differentiation and inflammation in rabbit articular chondrocytes via the p38 kinase and ERK-1/-2 signaling pathway.
Collapse
Affiliation(s)
- Young Seok Eom
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| | - Byung Su Ko
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| | - Fahad Hassan Shah
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| |
Collapse
|
4
|
Khalid S, Ekram S, Ramzan F, Salim A, Khan I. Co-regulation of Sox9 and TGFβ1 transcription factors in mesenchymal stem cells regenerated the intervertebral disc degeneration. Front Med (Lausanne) 2023; 10:1127303. [PMID: 37007782 PMCID: PMC10063891 DOI: 10.3389/fmed.2023.1127303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
BackgroundIntervertebral disc (IVD) shows aging and degenerative changes earlier than any other body connective tissue. Its repair and regeneration provide a considerable challenge in regenerative medicine due to its high degree of infrastructure and mechanical complexity. Mesenchymal stem cells, due to their tissue resurfacing potential, represent many explanatory pathways to regenerate a tissue breakdown.MethodsThis study was undertaken to evaluate the co-regulation of Sox9 and TGFβ1 in differentiating human umbilical cord mesenchymal stem cells (hUC-MSC) into chondrocytes. The combinatorial impact of Sox9 and TGFβ1 on hUC-MSCs was examined in vitro by gene expression and immunocytochemical staining. In in vivo, an animal model of IVD degeneration was established under a fluoroscopic guided system through needle puncture of the caudal disc. Normal and transfected MSCs were transplanted. Oxidative stress, pain, and inflammatory markers were evaluated by qPCR. Disc height index (DHI), water content, and gag content were analyzed. Histological examinations were performed to evaluate the degree of regeneration.ResultshUC-MSC transfected with Sox9+TGFβ1 showed a noticeable morphological appearance of a chondrocyte, and highly expressed chondrogenic markers (aggrecan, Sox9, TGFβ1, TGFβ2, and type II collagens) after transfection. Histological observation demonstrated that cartilage regeneration, extracellular matrix synthesis, and collagen remodeling were significant upon staining with H&E, Alcian blue, and Masson's trichrome stain on day 14. Additionally, oxidative stress, pain, and inflammatory markers were positively downregulated in the animals transplanted with Sox9 and TGFβ1 transfected MSCs.ConclusionThese findings indicate that the combinatorial effect of Sox9 and TGFβ1 substantially accelerates the chondrogenesis in hUC-MSCs. Cartilage regeneration and matrix synthesis were significantly enhanced. Therefore, a synergistic effect of Sox9 and TGFβ1 could be an immense therapeutic combination in the tissue engineering of cartilaginous joint bio-prostheses and a novel candidate for cartilage stabilization.
Collapse
|
5
|
Kitahashi T, Kogawa R, Nakamura K, Sekiya I. Integrin β1, PDGFRβ, and type II collagen are essential for meniscus regeneration by synovial mesenchymal stem cells in rats. Sci Rep 2022; 12:14148. [PMID: 35986079 PMCID: PMC9391488 DOI: 10.1038/s41598-022-18476-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractSynovial mesenchymal stem cells (MSCs) injected into the knee promote meniscus regeneration in several animal models; however, the mode of action is unknown. Our purpose was to identify the molecules responsible for this meniscus regeneration. Rat synovial MSCs were treated with neutralizing antibodies for integrin β1, PDGFRβ, or CD44 or with the CRISPR/Cas9 system to delete Vcam1, Tnfr1, or Col2a1 genes. After partial meniscectomy, rat knees were injected with MSCs, and the regenerated meniscus area was quantified three weeks later. The in vivo and in vitro functions were compared between the treated and control MSCs. Anti-integrin β1 neutralizing antibody inhibited in vitro MSC adhesion to collagen-coated chambers, anti-PDGFRβ neutralizing antibody inhibited proliferation in culture dishes, and Col2a1 deletion inhibited in vitro chondrogenesis. In vivo, the regenerated meniscus area was significantly smaller after injection of MSCs treated with integrin β1 and PDGFRβ neutralizing antibodies or lacking type II collagen gene than after control MSC injection. By contrast, the regenerated areas were similar after injection of control, CD44-, Vcam1-, or Tnfr1 treated MSCs (n = 12–16) MSCs. Synovial MSCs injected into the knee joint promoted meniscus regeneration by adhesion to integrin β1 in the meniscectomized region, proliferation by PDGFRβ, and cartilage matrix production from type II collagen.
Collapse
|
6
|
Multi-pin contact drawing enables production of anisotropic collagen fiber substrates for alignment of fibroblasts and monocytes. Colloids Surf B Biointerfaces 2022; 215:112525. [PMID: 35500531 DOI: 10.1016/j.colsurfb.2022.112525] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022]
Abstract
Type I collagen is the most abundant protein in the human body and is known to play important roles in numerous biological processes including tissue morphogenesis and wound healing. As such, it is one of the most frequently used substrates for cell culture, and there have been considerable efforts to develop collagen-based cell culture substrates that mimic the structural organization of collagen as it is found in native tissues, i.e., collagen fibers. However, producing collagen fibers from extracted collagen has been notoriously difficult, with existing methods providing only low throughput production of collagen fibers. In this study, we prepared collagen fibers using a highly efficient, bio-friendly, and cost-effective approach termed contact drawing, which uses an entangled polymer fluid to aid in fiber formation. Contact drawing technology has been demonstrated previously for collagen using highly concentrated dextran solutions with low concentrations of collagen. Here, we show that by replacing dextran with polyethylene oxide (PEO), high collagen content fibers may be readily formed from mixtures of soluble collagen and PEO, a polymer that readily forms fibers by contact drawing at concentrations as low as 0.5%wt. The presence of collagen and the formation of well-ordered collagen structures in the resulting fibers were characterized by attenuated total reflectance Fourier-transform infrared spectromicroscopy, Raman spectromicroscopy, and fluorescence microscopy. Corresponding to well-ordered collagen, the mechanical properties of the PEO-collagen fibers approximated those observed for native collagen fibers. Growth of cells on aligned PEO-collagen fibers attached to a polydimethyl siloxane support was examined for human dermal fibroblast (WS1) and human peripheral leukemia blood monocyte (THP-1) cell lines. WS1 and THP-1 cells readily attached, displayed alignment through migration and spreading, and proliferated on the collagen fiber substrate over the course of several days. We also demonstrated the retrieval of viable cells from the PEO-collagen fiber substrates through enzymatic digestion of the collagen substrate with collagenase IV.
Collapse
|
7
|
Shang J, Li H, Wu B, Jiang N, Wang B, Wang D, Zhong J, Chen Y, Xu X, Lu H. CircHIPK3 prevents chondrocyte apoptosis and cartilage degradation by sponging miR-30a-3p and promoting PON2. Cell Prolif 2022; 55:e13285. [PMID: 35716032 PMCID: PMC9436899 DOI: 10.1111/cpr.13285] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/26/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA) is a common joint disease featured by the deterioration of articular cartilage and chondrocyte death. Emerging evidence has indicated that circular RNAs (circRNAs) play an essential role in OA progress. Here, we found that the expression of circHIPK3 was significantly decreased in human and mouse OA cartilage. Knocking down circHIPK3 increased apoptosis and intracellular ROS level in HC‐a chondrocytes. We performed proteomic studies and identified that circHIPK3 regulated chondrocyte apoptosis through the mitochondrial pathway. Results of JC‐1 staining and western blot further confirmed that mitochondrial outer membrane permeabilization was promoted in HC‐a chondrocytes transfected by circHIPK3 siRNA. In terms of mechanism, we showed that PON2 functioned as a potential target of circHIPK3 to regulate chondrocyte apoptosis. Moreover, we revealed that circHIPK3 interacted with miR‐30a‐3p to regulate PON2 expression in chondrocytes. Taken together, our findings suggested that circHIPK3 regulated chondrocyte apoptosis by mitochondrial pathway, and targeting the circHIPK3/miR‐30a‐3p/PON2 axis might be a potential strategy for OA treatment.
Collapse
Affiliation(s)
- Jie Shang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Huizi Li
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Biao Wu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Ning Jiang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Bin Wang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dawei Wang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Junlong Zhong
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yufeng Chen
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Xianghe Xu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Huading Lu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| |
Collapse
|
8
|
Das P, Rajesh K, Lalzawmliana V, Bavya Devi K, Basak P, Lahiri D, Kundu B, Roy M, Nandi SK. Development and Characterization of Acellular Caprine Choncal Cartilage Matrix for Tissue Engineering Applications. Cartilage 2021; 13:1292S-1308S. [PMID: 31215790 PMCID: PMC8804783 DOI: 10.1177/1947603519855769] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Because of poor regenerative capabilities of cartilage, reconstruction of similar rigidity and flexibility is difficult, challenging, and restricted. The aim of the present investigation was to develop cost-effective acellular xenogeneic biomaterial as cartilage substitution. Two novel biometrics have been developed using different chemical processes (Na-deoxycholate + SDS and GndHCl + NaOH) to decellularize caprine (goat) ear cartilage and further extensively characterized before preclinical investigation. Complete cell removal was ascertained by hematoxylin and eosin staining followed by DNA estimation. No adverse effect on extracellular matrix (ECM) was found by quantifying collagen and sulfated glycosaminoglycans (sGAG) content as well as collagen, sGAG and elastin staining. Results showed no drastic changes in ECM structure apart from desired sGAG loss. Scanning electron microscopy images confirmed cellular loss and unaltered orientation. Nano-indentation study on cartilage matrices indicated interesting output showing better results among decellularized groups. Increased elastic modulus and hardness indicated better stiffness and more active energy dissipation mechanism due to decellularization. Fluid uptake and retention property remained unchanged after decellularization as analyzed by swelling behavior study. Additionally, acellular materials were confirmed to be nonreactive and nonhemolytic as assessed by in vitro hemocompatibility study. In vivo study (up to 3 months) on rabbits showed no symptoms of graft rejection/ tissue necrosis, established through postoperative histology and biochemical analyses of tissue explants. With regard to size, shape, biomechanics, source of origin and nonimmunogenic properties, these developed materials can play versatile role in biomedical/ clinical applications and pave a new insight as alternatives in cartilage reconstruction.
Collapse
Affiliation(s)
- Piyali Das
- School of Biosciences and Engineering,
Jadavpur University, Kolkata, West Bengal, India
| | - Kanike Rajesh
- Department of Metallurgical and
Materials Engineering, Indian Institute of Technology, Roorkee, Uttarakhand,
India
| | - V. Lalzawmliana
- Department of Veterinary Surgery and
Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West
Bengal, India
| | - K. Bavya Devi
- Department of Metallurgical and
Materials Engineering, Indian Institute of Technology–Kharagpur, Kharagpur, West
Bengal, India
| | - Piyali Basak
- School of Biosciences and Engineering,
Jadavpur University, Kolkata, West Bengal, India
| | - Debrupa Lahiri
- Department of Metallurgical and
Materials Engineering, Indian Institute of Technology, Roorkee, Uttarakhand,
India
| | - Biswanath Kundu
- Bioceramics and Coating Division,
CSIR–Central Glass and Ceramic Research Institute, Kolkata, West Bengal, India
| | - Mangal Roy
- Department of Metallurgical and
Materials Engineering, Indian Institute of Technology–Kharagpur, Kharagpur, West
Bengal, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and
Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West
Bengal, India,Samit Kumar Nandi, Department of Veterinary
Surgery and Radiology, West Bengal University of Animal and Fishery Sciences,
37, K. B. Sarani, Kolkata 700037, West Bengal, India.
| |
Collapse
|
9
|
Das P, Mishra R, Devi B, Rajesh K, Basak P, Roy M, Roy P, Lahiri D, Nandi SK. Decellularized xenogenic cartilage extracellular matrix (ECM) scaffolds for the reconstruction of osteochondral defects in rabbits. J Mater Chem B 2021; 9:4873-4894. [PMID: 34095925 DOI: 10.1039/d1tb00314c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of decellularized native allogenic or xenogenic cartilaginous extracellular matrix (ECM) biomaterials is widely expanding in the fields of tissue engineering and regenerative medicine. In this study, we aimed to develop an acellular, affordable, biodegradable, easily available goat conchal cartilaginous ECM derived scaffolding biomaterial for repair and regeneration of osteochondral defects in rabbits. Cartilages harvested from freshly collected goat ears were decellularized using chemical agents, namely, hypotonic-hypertonic (HH) buffer and Triton X-100 solution, separately. The morphologies and ultrastructure orientations of the decellularized cartilages remained unaltered in spite of complete cellular loss. Furthermore, when the acellular cartilaginous ECMs were cultured with murine mesenchymal stem cells (MSCs) (C3H10T1/2 cells), cellular infiltration and proliferation were thoroughly monitored using SEM, DAPI and FDA stained images, whereas the MTT assay proved the biocompatibility of the matrices. The increasing amounts of secreted ECM proteins (collagen and sGAG) indicated successful chondrogenic differentiation of the MSCs in the presence of the treated cartilage samples. In vivo biocompatibility studies showed no significant immune response or tissue rejection in the treated samples but tissue necrosis in control samples after 3 months. Upon implantation of the constructs in rabbits' osteochondral defects for 3 months, the histological and micro-CT evaluation revealed significant enhancement and regeneration of neocartilage and subchondral bony tissues. The IGF-1 loaded cartilaginous constructs showed comparatively better healing response after 3 months. Our results showed that decellularized xenogenic cartilaginous biomaterials preserved the bioactivity and integrity of the matrices that also favored in vitro stem cell proliferation and chondrogenic differentiation and enabled osteochondral regeneration, thus paving a new way for articular cartilage reconstruction.
Collapse
Affiliation(s)
- Piyali Das
- School of Bioscience and Engineering, Jadavpur University, Kolkata, India
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Sasaki JI, Abe GL, Li A, Matsumoto T, Imazato S. Large three-dimensional cell constructs for tissue engineering. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2021; 22:571-582. [PMID: 34408551 PMCID: PMC8366663 DOI: 10.1080/14686996.2021.1945899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Much research has been conducted on fabricating biomimetic biomaterials in vitro. Tissue engineering approaches are often conducted by combining cells, scaffolds, and growth factors. However, the degradation rate of scaffolds is difficult to control and the degradation byproducts occasionally limit tissue regeneration. To overcome these issues, we have developed a novel system using a thermo-responsive hydrogel that forms scaffold-free, three-dimensional (3D) cell constructs with arbitrary size and morphology. 3D cell constructs prepared using bone marrow-derived stromal stem cells (BMSCs) exhibited self-organizing ability and formed bone-like tissue with endochondral ossification. Endothelial cells were then introduced into the BMSC construct and a vessel-like structure was formed within the constructs. Additionally, the bone formation ability was promoted by endothelial cells and cell constructs could be freeze-dried to improve their clinical application. A pre-treatment with specific protein protectant allowed for the fabrication of novel bone substitutes composed only of cells. This 3D cell construct technology using thermo-responsive hydrogels was then applied to other cell species. Cell constructs composed of dental pulp stem cells were fabricated, and the resulting construct regenerated pulp-like tissue within a human pulpless tooth. In this review, we demonstrate the approaches for the in vitro fabrication of bone and dental pulp-like tissue using thermo-responsive hydrogels and their potential applications.
Collapse
Affiliation(s)
- Jun-Ichi Sasaki
- Department of Biomaterials Science, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Gabriela L Abe
- Department of Biomaterials Science, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Aonan Li
- Department of Biomaterials Science, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Imazato
- Department of Biomaterials Science, Osaka University Graduate School of Dentistry, Suita, Japan
- Department of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita, Japan
- CONTACT Satoshi Imazato Department of Biomaterials Science, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
11
|
Madsen SD, Giler MK, Bunnell BA, O'Connor KC. Illuminating the Regenerative Properties of Stem Cells In Vivo with Bioluminescence Imaging. Biotechnol J 2020; 16:e2000248. [PMID: 33089922 DOI: 10.1002/biot.202000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/17/2020] [Indexed: 11/10/2022]
Abstract
Preclinical animal studies are essential to the development of safe and effective stem cell therapies. Bioluminescence imaging (BLI) is a powerful tool in animal studies that enables the real-time longitudinal monitoring of stem cells in vivo to elucidate their regenerative properties. This review describes the application of BLI in preclinical stem cell research to address critical challenges in producing successful stem cell therapeutics. These challenges include stem cell survival, proliferation, homing, stress response, and differentiation. The applications presented here utilize bioluminescence to investigate a variety of stem and progenitor cells in several different in vivo models of disease and implantation. An overview of luciferase reporters is provided, along with the advantages and disadvantages of BLI. Additionally, BLI is compared to other preclinical imaging modalities and potential future applications of this technology are discussed in emerging areas of stem cell research.
Collapse
Affiliation(s)
- Sean D Madsen
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Margaret K Giler
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Kim C O'Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
12
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
13
|
Kilmer CE, Battistoni CM, Cox A, Breur GJ, Panitch A, Liu JC. Collagen Type I and II Blend Hydrogel with Autologous Mesenchymal Stem Cells as a Scaffold for Articular Cartilage Defect Repair. ACS Biomater Sci Eng 2020; 6:3464-3476. [PMID: 33463160 PMCID: PMC8287628 DOI: 10.1021/acsbiomaterials.9b01939] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Collagen type II is a promising material to repair cartilage defects since it is a major component of articular cartilage and plays a key role in chondrocyte function. This study investigated the chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (MSCs) embedded within a 3:1 collagen type I to II blend (Col I/II) hydrogel or an all collagen type I (Col I) hydrogel. Glycosaminoglycan (GAG) production in Col I/II hydrogels was statistically higher than that in Col I hydrogels or pellet culture, and these results suggested that adding collagen type II promoted GAG production. Col I/II hydrogels had statistically lower alkaline phosphatase (AP) activity than pellets cultured in a chondrogenic medium. The ability of MSCs encapsulated in Col I/II hydrogels to repair cartilage defects was investigated by creating two cartilage defects in the femurs of rabbits. After 13 weeks, histochemical staining suggested that Col I/II blend hydrogels provided favorable conditions for cartilage repair. Histological scoring revealed a statistically higher cartilage repair score for the Col I/II hydrogels compared to either the Col I hydrogels or empty defect controls. Results from this study suggest that there is clinical value in the cartilage repair capabilities of our Col I/II hydrogel with encapsulated MSCs.
Collapse
Affiliation(s)
- Claire E. Kilmer
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
| | - Carly M. Battistoni
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
| | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University,
West Lafayette, IN, 47907, USA
| | - Gert J. Breur
- Department of Veterinary Clinical Sciences, Purdue
University, West Lafayette, IN, 47907, USA
| | - Alyssa Panitch
- Weldon School of Biomedical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
- School of Biomedical Engineering, University of California
Davis, Davis, CA, 95616, USA
| | - Julie C. Liu
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
| |
Collapse
|
14
|
Zahiri S, Masaeli E, Poorazizi E, Nasr-Esfahani MH. Chondrogenic response in presence of cartilage extracellular matrix nanoparticles. J Biomed Mater Res A 2019; 106:2463-2471. [PMID: 29664223 DOI: 10.1002/jbm.a.36440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/26/2018] [Accepted: 03/28/2018] [Indexed: 12/19/2022]
Abstract
Current studies based on regenerative medicine suggest, decellularized extracellular matrix (DC-ECM) components can regulate cell phenotype. In this regard, it is believed, presence of cartilage extracellular matrix particles in culture condition could produce physical and biochemical supportive cues for chondrogenesis. In this study, DC-ECM nanoparticles with average size of 61.5± 22.4 nm were produced by decellularization and mechanical processing. Homogenous distribution and nanoscale size of yield particles were observed by scanning electron microscopy (SEM) and transmission electron microscopy (TEM) microscopy imaging. Chemical structure preservation of cartilage ECM after decellularization was also confirmed by typical Fourier transform infrared (FTIR) spectrum mapping. The influence these nanoparticles on chondrogenic response of chondrocyte cells was investigated by direct and indirect addition of nanoparticles to culture medium. A clinical devitalized cartilage powder (DV-ECM) was also used as a positive control. Totally, MTS results showed that direct and indirect presence of both DC-ECM and DV-ECM particles in culture medium enhanced cellular metabolic activity except on day one of culture. Furthermore, on day 21, SOX9 and COL2 expression of cultured chondrocytes in the medium containing DC-ECM nanoparticles were up-regulated in comparison to negative control, which was further confirmed by presence more frequent number of larger size lacunae in micromass spheroids. Our findings support the use of ECM nanoparticles as condition supplement in culture medium and injectable biomaterials, especially for cell-based therapies for cartilage regeneration. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A:2463-2471, 2018.
Collapse
Affiliation(s)
- Saeed Zahiri
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Department of Tissue Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Elahe Masaeli
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Elahe Poorazizi
- Department of Biochemistry, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
15
|
Irawan V, Sung TC, Higuchi A, Ikoma T. Collagen Scaffolds in Cartilage Tissue Engineering and Relevant Approaches for Future Development. Tissue Eng Regen Med 2018; 15:673-697. [PMID: 30603588 PMCID: PMC6250655 DOI: 10.1007/s13770-018-0135-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cartilage tissue engineering (CTE) aims to obtain a structure mimicking native cartilage tissue through the combination of relevant cells, three-dimensional scaffolds, and extraneous signals. Implantation of 'matured' constructs is thus expected to provide solution for treating large injury of articular cartilage. Type I collagen is widely used as scaffolds for CTE products undergoing clinical trial, owing to its ubiquitous biocompatibility and vast clinical approval. However, the long-term performance of pure type I collagen scaffolds would suffer from its limited chondrogenic capacity and inferior mechanical properties. This paper aims to provide insights necessary for advancing type I collagen scaffolds in the CTE applications. METHODS Initially, the interactions of type I/II collagen with CTE-relevant cells [i.e., articular chondrocytes (ACs) and mesenchymal stem cells (MSCs)] are discussed. Next, the physical features and chemical composition of the scaffolds crucial to support chondrogenic activities of AC and MSC are highlighted. Attempts to optimize the collagen scaffolds by blending with natural/synthetic polymers are described. Hybrid strategy in which collagen and structural polymers are combined in non-blending manner is detailed. RESULTS Type I collagen is sufficient to support cellular activities of ACs and MSCs; however it shows limited chondrogenic performance than type II collagen. Nonetheless, type I collagen is the clinically feasible option since type II collagen shows arthritogenic potency. Physical features of scaffolds such as internal structure, pore size, stiffness, etc. are shown to be crucial in influencing the differentiation fate and secreting extracellular matrixes from ACs and MSCs. Collagen can be blended with native or synthetic polymer to improve the mechanical and bioactivities of final composites. However, the versatility of blending strategy is limited due to denaturation of type I collagen at harsh processing condition. Hybrid strategy is successful in maximizing bioactivity of collagen scaffolds and mechanical robustness of structural polymer. CONCLUSION Considering the previous improvements of physical and compositional properties of collagen scaffolds and recent manufacturing developments of structural polymer, it is concluded that hybrid strategy is a promising approach to advance further collagen-based scaffolds in CTE.
Collapse
Affiliation(s)
- Vincent Irawan
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| | - Tzu-Cheng Sung
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Toshiyuki Ikoma
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| |
Collapse
|
16
|
Wang KH, Wan R, Chiu LH, Tsai YH, Fang CL, Bowley JF, Chen KC, Shih HN, Lai WFT. Effects of collagen matrix and bioreactor cultivation on cartilage regeneration of a full-thickness critical-size knee joint cartilage defects with subchondral bone damage in a rabbit model. PLoS One 2018; 13:e0196779. [PMID: 29746554 PMCID: PMC5945026 DOI: 10.1371/journal.pone.0196779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 04/19/2018] [Indexed: 12/23/2022] Open
Abstract
Cartilage has limited self-repair ability. The purpose of this study was to investigate the effects of different species of collagen-engineered neocartilage for the treatment of critical-size defects in the articular joint in a rabbit model. Type II and I collagen obtained from rabbits and rats was mixed to form a scaffold. The type II/I collagen scaffold was then mixed with rabbit chondrocytes to biofabricate neocartilage constructs using a rotating cell culture system [three-dimensional (3D)-bioreactor]. The rabbit chondrocytes were mixed with rabbit collagen scaffold and rat collagen scaffold to form neoRBT (neo-rabbit cartilage) and neoRAT (neo-rat cartilage) constructs, respectively. The neocartilage matrix constructs were implanted into surgically created defects in rabbit knee chondyles, and histological examinations were performed after 2 and 3 months. Cartilage-like lacunae formation surrounding the chondrocytes was noted in the cell cultures. After 3 months, both the neoRBT and neoRAT groups showed cartilage-like repair tissue covering the 5-mm circular, 4-mm-deep defects that were created in the rabbit condyle and filled with neocartilage plugs. Reparative chondrocytes were aligned as apparent clusters in both the neoRAT and neoRBT groups. Both neoRBT and neoRAT cartilage repair demonstrated integration with healthy adjacent tissue; however, more integration was obtained using the neoRAT cartilage. Our data indicate that different species of type II/I collagen matrix and 3D bioreactor cultivation can facilitate cartilage engineering in vitro for the repair of critical-size defect.
Collapse
Affiliation(s)
- Kuo-Hwa Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
- Department of Obstetrics and Gynecology, Chung Kang branch, Cheng Ching Hospital, Taichung, Taiwan, ROC
| | - Richard Wan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
| | - Li-Hsuan Chiu
- Department of Research, Taipei Medical University-Shaung-Ho Hospital, Taipei, Taiwan, ROC
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States of America
| | - Yu-Hui Tsai
- Department of Research, Taipei Medical University-Shaung-Ho Hospital, Taipei, Taiwan, ROC
| | - Chia-Lang Fang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - John F. Bowley
- Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Kuan-Chou Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
| | - Hsin-Nung Shih
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Chang Gung University, Linkou Taoyuan, Taiwan, ROC
- * E-mail: (HNS); (WFTL)
| | - Wen-Fu Thomas Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
- Department of Research, Taipei Medical University-Shaung-Ho Hospital, Taipei, Taiwan, ROC
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States of America
- * E-mail: (HNS); (WFTL)
| |
Collapse
|
17
|
Wong CC, Chen CH, Chiu LH, Tsuang YH, Bai MY, Chung RJ, Lin YH, Hsieh FJ, Chen YT, Yang TL. Facilitating In Vivo Articular Cartilage Repair by Tissue-Engineered Cartilage Grafts Produced From Auricular Chondrocytes. Am J Sports Med 2018; 46:713-727. [PMID: 29211970 DOI: 10.1177/0363546517741306] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Insufficient cell numbers still present a challenge for articular cartilage repair. Converting heterotopic auricular chondrocytes by extracellular matrix may be the solution. HYPOTHESIS Specific extracellular matrix may convert the phenotype of auricular chondrocytes toward articular cartilage for repair. STUDY DESIGN Controlled laboratory study. METHODS For in vitro study, rabbit auricular chondrocytes were cultured in monolayer for several passages until reaching status of dedifferentiation. Later, they were transferred to chondrogenic type II collagen (Col II)-coated plates for further cell conversion. Articular chondrogenic profiles, such as glycosaminoglycan deposition, articular chondrogenic gene, and protein expression, were evaluated after 14-day cultivation. Furthermore, 3-dimensional constructs were fabricated using Col II hydrogel-associated auricular chondrocytes, and their histological and biomechanical properties were analyzed. For in vivo study, focal osteochondral defects were created in the rabbit knee joints, and auricular Col II constructs were implanted for repair. RESULTS The auricular chondrocytes converted by a 2-step protocol expressed specific profiles of chondrogenic molecules associated with articular chondrocytes. The histological and biomechanical features of converted auricular chondrocytes became similar to those of articular chondrocytes when cultivated with Col II 3-dimensional scaffolds. In an in vivo animal model of osteochondral defects, the treated group (auricular Col II) showed better cartilage repair than did the control groups (sham, auricular cells, and Col II). Histological analyses revealed that cartilage repair was achieved in the treated groups with abundant type II collagen and glycosaminoglycans syntheses rather than elastin expression. CONCLUSION The study confirmed the feasibility of applying heterotopic chondrocytes for cartilage repair via extracellular matrix-induced cell conversion. CLINICAL RELEVANCE This study proposes a feasible methodology to convert heterotopic auricular chondrocytes for articular cartilage repair, which may serve as potential alternative sources for cartilage repair.
Collapse
Affiliation(s)
- Chin-Chean Wong
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hwa Chen
- Bone and Joint Research Center, Department of Orthopedics, Taipei Medical University Hospital, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Li-Hsuan Chiu
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, USA.,Center for Nano Tissue Engineering and Image Research, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yang-Hwei Tsuang
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Yi Bai
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan.,Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan
| | - Yun-Ho Lin
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Fon-Jou Hsieh
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - You-Tzung Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Lin Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,a Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
18
|
Rahman S, Griffin M, Naik A, Szarko M, Butler PEM. Optimising the decellularization of human elastic cartilage with trypsin for future use in ear reconstruction. Sci Rep 2018; 8:3097. [PMID: 29449572 PMCID: PMC5814427 DOI: 10.1038/s41598-018-20592-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 01/18/2018] [Indexed: 01/16/2023] Open
Abstract
Decellularized scaffolds can induce chondrogenic differentiation of stem cells. This study compares different methods to optimise the decellularization of auricular cartilage. The process consisted of an initial 12 hour dry freeze thaw which froze the cartilage specimens in an empty tube at -20 °C. Samples were allowed to thaw at room temperature followed by submersion in phosphate buffer solution in which they were frozen at -20 °C for a 12 hour period. They were then allowed to thaw at room temperature as before. Protocol A subsequently involved subjecting specimens to both deoxyribonuclease and sodium deoxycholate. Protocol B and C were adaptations of this using 0.25% trypsin (7 cycles) and a 0.5 molar solution of ethylenediaminetetraacetic acid (3 hours for each cycle) respectively as additional steps. Trypsin accelerated the decellularization process with a reduction in DNA content from 55.4 ng/μL (native) to 17.3 ng/μL (P-value < 0.05) after 14 days. Protocol B showed a faster reduction in DNA content when compared with protocol A. In comparison to protocol C after 14 days, trypsin also showed greater decellularization with a mean difference of 11.7 ng/μL (P-value < 0.05). Histological analysis with H&E and DAPI confirmed depletion of cells at 14 days with trypsin.
Collapse
Affiliation(s)
- Shafiq Rahman
- Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom.
| | - Michelle Griffin
- Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom. .,Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom. .,Plastic and Reconstructive Surgery Department, Royal Free Hospital, London, United Kingdom.
| | - Anish Naik
- Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom
| | - Matthew Szarko
- Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom.,Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom.,Anatomy Department, St George's University, London, United Kingdom
| | - Peter E M Butler
- Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom.,Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom.,Plastic and Reconstructive Surgery Department, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
19
|
Chen G, Kawazoe N. Biomimetic Extracellular Matrices and Scaffolds Prepared from Cultured Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:465-474. [DOI: 10.1007/978-981-13-0950-2_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Ewa-Choy YW, Pingguan-Murphy B, Abdul-Ghani NA, Jahendran J, Chua KH. Effect of alginate concentration on chondrogenesis of co-cultured human adipose-derived stem cells and nasal chondrocytes: a biological study. Biomater Res 2017; 21:19. [PMID: 29075508 PMCID: PMC5646124 DOI: 10.1186/s40824-017-0105-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/28/2017] [Indexed: 02/07/2023] Open
Abstract
Background The three-dimensional (3D) system is one of the important factors to engineer a biocompatible and functional scaffold for the applications of cell-based therapies for cartilage repair. The 3D alginate hydrogels system has previously been shown to potentially promote chondrogenesis. The chondrocytic differentiation of co-cultured adipose-derived stem cells (ADSCs) and nasal chondrocytes (NCs) within alginate constructs are hypothesized to be influenced by concentration of alginate hydrogel. In this study, we evaluated the effects of alginate concentration on chondrogenic differentiation of ADSCs and NCs co-cultured in a biological approach. Method The co-cultured cells of 2:1 ADSCs-to-NCs ratio were encapsulated in alginate constructs in one of three concentrations (1.0%, 1.2% and 1.5%) and cultured under serum free conditions for 7 days. Cell viability, cell proliferation, immunohistochemical, gycosaminogylycans (GAG) synthesis, and gene expression were examined. Results Overall, the 1.2% alginate concentration group was relatively effective in chondrocytic differentiation in comparable to other groups. The cell morphology, cell viability, and cell proliferation revealed initial chondrogenic differentiation by the formation of cell clusters as well as the high permeability for exchange of solutes. The formation of newly synthesis cartilage-specific extracellular matrix in 1.2% group was demonstrated by positive immunohistochemical staining of collagen type II. The co-cultured cells in 1.2% group highly expressed COL II, ACP and SOX-9, compared to 1.0% and 1.5% groups, denote the retention of cartilaginous-specific phenotype by suppressing the undifferentiation stem cell markers of SOX-2 and OCT-4. The study showed 1.2% group was less likely to differentiate towards osteogenesis by downregulating hyperthrophy chondrocytic gene of COL X and osseous marker genes of OSC and OSP. Conclusion This study suggests that variations in the alginate concentration of co-cultured ADSCs and NCs influenced the chondrogenesis. The remarkable biological performance on chondrogenic differentiation in regulating the concentration of alginate 3D culture provides new insights into the cell cross-talk and demonstrates the effectiveness in regenerative therapies of cartilage defects in tissue engineering.
Collapse
Affiliation(s)
- Y W Ewa-Choy
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - B Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - N A Abdul-Ghani
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - J Jahendran
- Suite 16, Pantai Cheras Medical Centre, Kuala Lumpur, Malaysia
| | - K H Chua
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Jin GZ, Kim HW. Effects of Type I Collagen Concentration in Hydrogel on the Growth and Phenotypic Expression of Rat Chondrocytes. Tissue Eng Regen Med 2017; 14:383-391. [PMID: 30603494 PMCID: PMC6171609 DOI: 10.1007/s13770-017-0060-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 11/25/2022] Open
Abstract
It is controversial whether type I collagen itself can maintain and improve chondrogenic phenotype of chondrocytes in a three-dimensional (3D) environment. In this study, we examined the effect of type I collagen concentration in hydrogel (0.5, 1, and 2 mg/ml) on the growth and phenotype expression of rat chondrocytes in vitro. All collagen hydrogels showed substantial contractions during culture, in a concentration-dependent manner, which was due to the cell proliferation. The cell viability was shown to be the highest in 2 mg/ml collagen gel. The mRNA expression of chondrogenic phenotypes, including SOX9, type II collagen, and aggrecan, was significantly up-regulated, particularly in 1 mg/ml collagen gel. Furthermore, the production of type II collagen and glycosaminoglycan (GAG) content was also enhanced. The results suggest that type I collagen hydrogel is not detrimental to, but may be useful for, the chondrocyte culture for cartilage tissue engineering.
Collapse
Affiliation(s)
- Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116 Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116 Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116 Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116 Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116 Korea
| |
Collapse
|
22
|
Zayed M, Caniglia C, Misk N, Dhar MS. Donor-Matched Comparison of Chondrogenic Potential of Equine Bone Marrow- and Synovial Fluid-Derived Mesenchymal Stem Cells: Implications for Cartilage Tissue Regeneration. Front Vet Sci 2017; 3:121. [PMID: 28149840 PMCID: PMC5241318 DOI: 10.3389/fvets.2016.00121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 12/19/2016] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been demonstrated to be useful for cartilage tissue regeneration. Bone marrow (BM) and synovial fluid (SF) are promising sources for MSCs to be used in cartilage regeneration. In order to improve the clinical outcomes, it is recommended that prior to clinical use, the cellular properties and, specifically, their chondrogenic potential must be investigated. The purpose of this study is to compare and better understand the in vitro chondrogenic potential of equine bone marrow-derived mesenchymal stem cells (BMMSCs) and synovial fluid-derived mesenchymal stem cells (SFMSCs) populated from the same equine donor. BM- and SF-derived MSCs cultures were generated from five equine donors, and the MSCs were evaluated in vitro for their morphology, proliferation, trilineage differentiation, and immunophenotyping. Differences in their chondrogenic potentials were further evaluated quantitatively using glycosaminoglycan (GAG) content and via immunofluorescence of chondrogenic differentiation protein markers, SRY-type HMG box9, Aggrecan, and collagen II. The BMMSCs and SFMSCs were similar in cellular morphology, viability, and immunophenotype, but, varied in their chondrogenic potential, and expression of the key chondrogenic proteins. The SFMSCs exhibited a significant increase in GAG content compared to the BMMSCs (P < 0.0001) in three donors, suggesting increased levels of chondrogenesis. The expression of the key chondrogenic proteins correlated positively with the GAG content, suggesting that the differentiation process is dependent on the expression of the target proteins in these three donors. Our findings suggest that even though SFMSCs were hypothesized to be more chondrogenic relative to BMMSCs, there was considerable donor-to-donor variation in the primary cultures of MSCs which can significantly affect their downstream application.
Collapse
Affiliation(s)
- Mohammed Zayed
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA; Department of Animal Surgery, College of Veterinary Medicine, South Valley University, Qena, Egypt
| | | | - Nabil Misk
- Department of Animal Surgery, College of Veterinary Medicine, Assiut University , Asyut , Egypt
| | - Madhu S Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee , Knoxville, TN , USA
| |
Collapse
|
23
|
Mahapatra C, Jin GZ, Kim HW. Alginate-hyaluronic acid-collagen composite hydrogel favorable for the culture of chondrocytes and their phenotype maintenance. Tissue Eng Regen Med 2016; 13:538-546. [PMID: 30603434 PMCID: PMC6170835 DOI: 10.1007/s13770-016-0059-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/15/2016] [Accepted: 06/01/2016] [Indexed: 12/21/2022] Open
Abstract
Articular cartilage has limited regeneration capacity, thus significant challenge has been made to restore the functions. The development of hydrogels that can encapsulate and multiply cells, and then effectively maintain the chondrocyte phenotype is a meaningful strategy to this cartilage repair. In this study, we prepared alginate-hyaluronic acid based hydrogel with type I collagen being incorporated, namely Alg-HA-Col composite hydrogel. The incorporation of Col enhanced the chemical interaction of molecules, and the thermal stability and dynamic mechanical properties of the resultant hydrogels. The primary chondrocytes isolated from rat cartilage were cultured within the composite hydrogel and the cell viability recorded revealed active proliferation over a period of 21 days. The mRNA levels of chondrocyte phenotypes, including SOX9, collagen type II, and aggrecan, were significantly up-regulated when the cells were cultured within the Alg-HA-Col gel than those cultured within the Alg-HA. Furthermore, the secretion of sulphated glycosaminoglycan, a cartilage-specific matrix molecule, was recorded higher in the collagen-added composite hydrogel. Although more in-depth studies are required such as the in vivo functions, the currently-prepared Alg-HA-Col composite hydrogel is considered to provide favorable 3-dimensional matrix conditions for the cultivation of chondrocytes. Moreover, the cell-cultured constructs may be useful for the cartilage repair and tissue engineering.
Collapse
Affiliation(s)
- Chinmaya Mahapatra
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116 Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116 Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116 Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Korea
| |
Collapse
|
24
|
Arai Y, Park S, Choi B, Ko KW, Choi WC, Lee JM, Han DW, Park HK, Han I, Lee JH, Lee SH. Enhancement of Matrix Metalloproteinase-2 (MMP-2) as a Potential Chondrogenic Marker during Chondrogenic Differentiation of Human Adipose-Derived Stem Cells. Int J Mol Sci 2016; 17:ijms17060963. [PMID: 27322256 PMCID: PMC4926495 DOI: 10.3390/ijms17060963] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 01/09/2023] Open
Abstract
Human adipose-derived stem cells (hASCs) have a capacity to undergo adipogenic, chondrogenic, and osteogenic differentiation. Recently, hASCs were applied to various fields including cell therapy for tissue regeneration. However, it is hard to predict the direction of differentiation of hASCs in real-time. Matrix metalloproteinases (MMPs) are one family of proteolytic enzymes that plays a pivotal role in regulating the biology of stem cells. MMPs secreted by hASCs are expected to show different expression patterns depending on the differentiation state of hASCs because biological functions exhibit different patterns during the differentiation of stem cells. Here, we investigated proteolytic enzyme activity, especially MMP-2 activity, in hASCs during their differentiation. The activities of proteolytic enzymes and MMP-2 were higher during chondrogenic differentiation than during adipogenic and osteogenic differentiation. During chondrogenic differentiation, mRNA expression of MMP-2 and the level of the active form of MMP-2 were increased, which also correlated with Col II. It is concluded that proteolytic enzyme activity and the level of the active form of MMP-2 were increased during chondrogenic differentiation, which was accelerated in the presence of Col II protein. According to our findings, MMP-2 could be a candidate maker for real-time detection of chondrogenic differentiation of hASCs.
Collapse
Affiliation(s)
- Yoshie Arai
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Sunghyun Park
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Bogyu Choi
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Kyoung-Won Ko
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Won Chul Choi
- Department of Orthopedic Surgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Joong-Myung Lee
- Department of Orthopedic Surgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Dong-Wook Han
- Department of Optics and Mechatronics Engineering, BK21+ Nano-Integrated Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan 619-961, Korea.
| | - Hun-Kuk Park
- Department of Biomedical Engineering, Collage of Medicine, Kyung Hee University, Seoul 151-742, Korea.
| | - Inbo Han
- Department of Neurosurgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Jong Hun Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Gyeonggi-do 443-742, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| |
Collapse
|
25
|
Jhala D, Vasita R. A Review on Extracellular Matrix Mimicking Strategies for an Artificial Stem Cell Niche. POLYM REV 2015. [DOI: 10.1080/15583724.2015.1040552] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
26
|
Tsai HL, Chiu WT, Fang CL, Hwang SM, Renshaw PF, Lai WFT. Different forms of tenascin-C with tenascin-R regulate neural differentiation in bone marrow-derived human mesenchymal stem cells. Tissue Eng Part A 2015; 20:1908-21. [PMID: 24829055 DOI: 10.1089/ten.tea.2013.0188] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are currently thought to transdifferentiate into neural lineages under specific microenvironments. Studies have reported that the tenascin family members, tenascin-C (TnC) and tenascin-R (TnR), regulate differentiation and migration, in addition to neurite outgrowth and survival in numerous types of neurons and mesenchymal progenitor cells. However, the mechanisms by which TnC and TnR affect neuronal differentiation are not well understood. In this study, we hypothesized that different forms of tenascin might regulate the neural transdifferentiation of human bone marrow-derived mesenchymal stem cells. Human MSCs were cultured in media incorporated with soluble tenascins, or on precoated tenascins. In a qualitative polymerase chain reaction analysis, adding a soluble TnC and TnR mixture to the medium significantly enhanced the expression of neuronal and glial markers, whereas no synaptic markers were expressed. Conversely, in groups of cells treated with coated TnC, hMSCs showed neurite outgrowth and synaptic marker expression. After being treated with coated TnR, hMSCs exhibited neuronal differentiation; however, it inhibited neurite outgrowth and synaptic marker expression. A combination of TnC and TnR significantly promoted hMSC differentiation in neurons or oligodendrocytes, induced neurite formation, and inhibited differentiation into astrocytes. Furthermore, the effect of the tenascin mixture showed dose-dependent effects, and a mixture ratio of 1:1 to 1:2 (TnC:TnR) provided the most obvious differentiation of neurons and oligodendrocytes. In a functional blocking study, integrin α7 and α9β1-blocking antibodies inhibited, respectively, 80% and 20% of mRNA expression by hMSCs in the coated tenascin mixture. In summary, the coated combination of TnC and TnR appeared to regulate neural differentiation signaling through integrin α7 and α9β1 in bone marrow-derived hMSCs. Our findings demonstrate novel mechanisms by which tenascin regulates neural differentiation, and enable the use of cell therapy to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Hung-Li Tsai
- 1 Graduate Institute of Medical Sciences, Taipei Medical University , Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
27
|
Utomo L, Pleumeekers MM, Nimeskern L, Nürnberger S, Stok KS, Hildner F, van Osch GJVM. Preparation and characterization of a decellularized cartilage scaffold for ear cartilage reconstruction. ACTA ACUST UNITED AC 2015; 10:015010. [PMID: 25586138 DOI: 10.1088/1748-6041/10/1/015010] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Scaffolds are widely used to reconstruct cartilage. Yet, the fabrication of a scaffold with a highly organized microenvironment that closely resembles native cartilage remains a major challenge. Scaffolds derived from acellular extracellular matrices are able to provide such a microenvironment. Currently, no report specifically on decellularization of full thickness ear cartilage has been published. In this study, decellularized ear cartilage scaffolds were prepared and extensively characterized. Cartilage decellularization was optimized to remove cells and cell remnants from elastic cartilage. Following removal of nuclear material, the obtained scaffolds retained their native collagen and elastin contents as well as their architecture and shape. High magnification scanning electron microscopy showed no obvious difference in matrix density after decellularization. However, glycosaminoglycan content was significantly reduced, resulting in a loss of viscoelastic properties. Additionally, in contact with the scaffolds, human bone-marrow-derived mesenchymal stem cells remained viable and are able to differentiate toward the chondrogenic lineage when cultured in vitro. These results, including the ability to decellularize whole human ears, highlight the clinical potential of decellularization as an improved cartilage reconstruction strategy.
Collapse
Affiliation(s)
- Lizette Utomo
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands. Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands. Faculty of Science and Technology, University of Twente, PO Box 217, 7500 AE Enschede, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
28
|
Choi B, Kim S, Lin B, Wu BM, Lee M. Cartilaginous extracellular matrix-modified chitosan hydrogels for cartilage tissue engineering. ACS APPLIED MATERIALS & INTERFACES 2014; 6:20110-21. [PMID: 25361212 DOI: 10.1021/am505723k] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Cartilaginous extracellular matrix (ECM) components such as type-II collagen (Col II) and chondroitin sulfate (CS) play a crucial role in chondrogenesis. However, direct clinical use of natural Col II or CS as scaffolds for cartilage tissue engineering is limited by their instability and rapid enzymatic degradation. Here, we investigate the incorporation of Col II and CS into injectable chitosan hydrogels designed to gel upon initiation by exposure to visible blue light (VBL) in the presence of riboflavin. Unmodified chitosan hydrogel supported proliferation and deposition of cartilaginous ECM by encapsulated chondrocytes and mesenchymal stem cells. The incorporation of native Col II or CS into chitosan hydrogels further increased chondrogenesis. The incorporation of Col II, in particular, was found to be responsible for the enhanced cellular condensation and chondrogenesis observed in modified hydrogels. This was mediated by integrin α10 binding to Col II, increasing cell-matrix adhesion. These findings demonstrate the potential of cartilage ECM-modified chitosan hydrogels as biomaterials to promote cartilage regeneration.
Collapse
Affiliation(s)
- Bogyu Choi
- Division of Advanced Prosthodontics, ‡Department of Bioengineering, University of California, Los Angeles , Los Angeles, California 90095, United States
| | | | | | | | | |
Collapse
|
29
|
Deng WP, Yang CC, Yang LY, Chen CWD, Chen WH, Yang CB, Chen YH, Lai WFT, Renshaw PF. Extracellular matrix-regulated neural differentiation of human multipotent marrow progenitor cells enhances functional recovery after spinal cord injury. Spine J 2014; 14:2488-99. [PMID: 24792783 PMCID: PMC4692164 DOI: 10.1016/j.spinee.2014.04.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 04/01/2014] [Accepted: 04/15/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Recent advanced studies have demonstrated that cytokines and extracellular matrix (ECM) could trigger various types of neural differentiation. However, the efficacy of differentiation and in vivo transplantation has not yet thoroughly been investigated. PURPOSE To highlight the current understanding of the effects of ECM on neural differentiation of human bone marrow-derived multipotent progenitor cells (MPCs), regarding state-of-art cure for the animal with acute spinal cord injury (SCI), and explore future treatments aimed at neural repair. STUDY DESIGN A selective overview of the literature pertaining to the neural differentiation of the MSCs and experimental animals aimed at improved repair of SCI. METHODS Extracellular matrix proteins, tenascin-cytotactin (TN-C), tenascin-restrictin (TN-R), and chondroitin sulfate (CS), with the cytokines, nerve growth factor (NGF)/brain-derived neurotrophic factor (BDNF)/retinoic acid (RA) (NBR), were incorporated to induce transdifferentiation of human MPCs. Cells were treated with NBR for 7 days, and then TN-C, TN-R, or CS was added for 2 days. The medium was changed every 2 days. Twenty-four animals were randomly assigned to four groups with six animals in each group: one experimental and three controls. Animals received two (bilateral) injections of vehicle, MPCs, NBR-induced MPCs, or NBR/TN-C-induced MPCs into the lesion sites after SCI. Functional assessment was measured using the Basso, Beattie, and Bresnahan locomotor rating score. Data were analyzed using analysis of variance followed by Student-Newman-Keuls (SNK) post hoc tests. RESULTS Results showed that MPCs with the transdifferentiation of human MPCs to neurons were associated with increased messenger-RNA (mRNA) expression of neuronal markers including nestin, microtubule-associated protein (MAP) 2, glial fibrillary acidic protein, βIII tubulin, and NGF. Greater amounts of neuronal morphology appeared in cultures incorporated with TN-C and TN-R than those with CS. The addition of TN-C enhanced mRNA expressions of MAP2, βIII tubulin, and NGF, whereas TN-R did not significantly change. Conversely, CS exposure decreased MAP2, βIII tubulin, and NGF expressions. The TN-C-treated MSCs significantly and functionally repaired SCI-induced rats at Day 42. Present results indicate that ECM components, such as tenascins and CS in addition to cytokines, may play functional roles in regulating neurogenesis by human MPCs. CONCLUSIONS These findings suggest that the combined use of TN-C, NBR, and human MPCs offers a new feasible method for nerve repair.
Collapse
Affiliation(s)
- Win-Ping Deng
- Graduate Institute of Biomedical Materials and Engineering, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Chi-Chiang Yang
- Department of Neurology, Tungs’ Taichung Metroharbor Hospital, 699 Taiwan Blvd. 8 Sec., Taitung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Chun-Wei D. Chen
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 415 E. 68th Street, New York 10065, NY, USA
| | - Wei-Hong Chen
- Graduate Institute of Biomedical Materials and Engineering, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Charn-Bing Yang
- Orthopedic Section Department, New Taipei City Hospital, 198 Yin-His Rd., Banquiao District, New Taipei City, Taiwan
| | - Yu-Hsin Chen
- Department of Physiology, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Wen-Fu T. Lai
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 415 E. 68th Street, New York 10065, NY, USA,International Center of Nano Biomedicine Research, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan,Brain McLean Imaging Center, McLean Hospital/Harvard Medical School, 115 Mill Strret, Belmont 02115, MA, USA,Corresponding author. Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan. Tel.: (886)2-23916632; fax: (886)2-23967262. (W.-F.T. Lai)
| | - Perry F. Renshaw
- The Brain Institute, The University of Utah, 201 Presidents Cir, Salt Lake City 84112, UT, USA
| |
Collapse
|
30
|
Wang YG, Xie P, Wang YG, Li XD, Zhang TG, Liu ZY, Hong Q, Du SX. All-trans-retinoid acid (ATRA) suppresses chondrogenesis of rat primary hind limb bud mesenchymal cells by downregulating p63 and cartilage-specific molecules. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:460-8. [PMID: 25136779 DOI: 10.1016/j.etap.2014.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 02/05/2023]
Abstract
P63 null mice have no or truncated limbs and mutations in human p63 cause several skeletal syndromes that also show limb and digit abnormalities, suggesting its essential role in bone development. In the current study, we investigated the effect of ATRA on chondrogenesis using mesenchymal cells from rat hind limb bud and further examined the mRNA and protein expression of Sox9 and Col2a1 and p63 in rat hind limb bud cells. Limb buds were isolated from embryos from euthanized female rats. Growth of hind limb bud mesenchymal cells was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assays. Formation of cartilage nodules was examined by Alcian blue-nuclear fast red staining. The expression of Sox9, Col2al and p63 was determined by Real-time RT-PCR and immunoblotting assays, respectively. Our MTT assays revealed that ATRA at 1 and 10μM significantly suppressed the growth of mesenchymal cells from rat hind limb bud at 24 and 48h (P<0.01 vs. controls). Alcian blue staining further showed that ATRA caused a significant dose-dependent reduction in the area of cartilage nodules (P<0.05 in all vs. controls). At 1μM ATRA, the area of cartilage nodules from hind limb bud cells was reduced to 0.05±0.03mm from 0.15±0.01mm in controls. Real-time RT-PCR assays further indicated that 1 and 10μM ATRA markedly reduced the mRNA expression of Sox9, Col2al and p63 in hind limb bud cells (P<0.05 in all vs. controls). In addition, ATRA time-dependently inhibits the mRNA expression of p63, Sox9 and Col2al. Western blotting assays additionally showed that ATRA dose-dependently reduced the expression of Sox9, Col2al and p63 (P<0.05 in all vs. controls). Together, our results suggest that ATRA suppresses chondrogenesis by modulating the expression of Sox9, Col2al and p63 in primary hind limb bud mesenchymal cells.
Collapse
Affiliation(s)
- Yun-Guo Wang
- Department of Orthopedics, the Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Peng Xie
- Department of Orthopedics, the Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yun-Gong Wang
- Wang Chuanshan College, University of South China, Hengyang, Hunan, 421001, China
| | - Xue-Dong Li
- Department of Orthopedics, the First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Tao-Gen Zhang
- Department of Orthopedics, the First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Zhao-Yong Liu
- Department of Orthopedics, the First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Quan Hong
- Department of Orthopedics, the First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Shi-Xin Du
- Department of Orthopedics, the Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| |
Collapse
|
31
|
Kim HJ, Yeom JS, Koh YG, Yeo JE, Kang KT, Kang YM, Chang BS, Lee CK. Anti-inflammatory effect of platelet-rich plasma on nucleus pulposus cells with response of TNF-α and IL-1. J Orthop Res 2014; 32:551-6. [PMID: 24338609 DOI: 10.1002/jor.22532] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 11/09/2013] [Indexed: 02/04/2023]
Abstract
The purpose of this study was to investigate the anti-inflammatory effect of platelet-rich plasma (PRP) with collagen matrix on human nucleus pulposus (NP) cell in response to pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1). NP cells from human disks were cultured in a monolayer and maintained in the collagen matrix prior to the addition of recombinant human IL-1 and TNF-α. After applying IL-1 and TNF-α, PRP prepared by using a commercially available platelet concentration system was added. The response was investigated using real-time PCR for mRNA expression of type II collagen, aggrecan, matrix metalloproteinase-3 (MMP-3), and cyclooxygenase-2 (COX-2). The combination of IL-1β and TNF-α led to decrease of matrix synthesis gene expression such as collagen type II and aggrecan and increase of the degradation gene expression of COX-2 and MMP-3, compared to the control. Consecutive PRP exposure significantly recovered the down-regulated gene expression of collagen type II and aggrecan and significantly reduced the increased MMP-3 and COX-2 gene expression, compared to that of control groups with pro-inflammatory cytokines. The administration of PRP with collagen matrix markedly suppressed cytokine-induced pro-inflammatory degrading enzymes and mediators in the NP cell. It also rescued gene expression concerning matrix synthesis, thereby stabilizing NP cell differentiation.
Collapse
Affiliation(s)
- Ho-Joong Kim
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, 166 Gumiro, Bundang-gu, Sungnam, 463-707, Republic of Korea; Spine Center, Seoul National University Bundang Hospital, 166 Gumiro, Bundang-gu, Sungnam, 463-707, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wang X, Schröder HC, Grebenjuk V, Diehl-Seifert B, Mailänder V, Steffen R, Schloßmacher U, Müller WEG. The marine sponge-derived inorganic polymers, biosilica and polyphosphate, as morphogenetically active matrices/scaffolds for the differentiation of human multipotent stromal cells: potential application in 3D printing and distraction osteogenesis. Mar Drugs 2014; 12:1131-47. [PMID: 24566262 PMCID: PMC3944534 DOI: 10.3390/md12021131] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/10/2014] [Accepted: 02/17/2014] [Indexed: 01/03/2023] Open
Abstract
The two marine inorganic polymers, biosilica (BS), enzymatically synthesized from ortho-silicate, and polyphosphate (polyP), a likewise enzymatically synthesized polymer consisting of 10 to >100 phosphate residues linked by high-energy phosphoanhydride bonds, have previously been shown to display a morphogenetic effect on osteoblasts. In the present study, the effect of these polymers on the differential differentiation of human multipotent stromal cells (hMSC), mesenchymal stem cells, that had been encapsulated into beads of the biocompatible plant polymer alginate, was studied. The differentiation of the hMSCs in the alginate beads was directed either to the osteogenic cell lineage by exposure to an osteogenic medium (mineralization activation cocktail; differentiation into osteoblasts) or to the chondrogenic cell lineage by incubating in chondrocyte differentiation medium (triggering chondrocyte maturation). Both biosilica and polyP, applied as Ca²⁺ salts, were found to induce an increased mineralization in osteogenic cells; these inorganic polymers display also morphogenetic potential. The effects were substantiated by gene expression studies, which revealed that biosilica and polyP strongly and significantly increase the expression of bone morphogenetic protein 2 (BMP-2) and alkaline phosphatase (ALP) in osteogenic cells, which was significantly more pronounced in osteogenic versus chondrogenic cells. A differential effect of the two polymers was seen on the expression of the two collagen types, I and II. While collagen Type I is highly expressed in osteogenic cells, but not in chondrogenic cells after exposure to biosilica or polyP, the upregulation of the steady-state level of collagen Type II transcripts in chondrogenic cells is comparably stronger than in osteogenic cells. It is concluded that the two polymers, biosilica and polyP, are morphogenetically active additives for the otherwise biologically inert alginate polymer. It is proposed that alginate, supplemented with polyP and/or biosilica, is a suitable biomaterial that promotes the growth and differentiation of hMSCs and might be beneficial for application in 3D tissue printing of hMSCs and for the delivery of hMSCs in fractures, surgically created during distraction osteogenesis.
Collapse
Affiliation(s)
- Xiaohong Wang
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Vladislav Grebenjuk
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | | | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany.
| | - Renate Steffen
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Ute Schloßmacher
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Werner E G Müller
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| |
Collapse
|
33
|
Liu MC, Chen WH, Wu LC, Hsu WC, Lo WC, Yeh SD, Wang MF, Zeng R, Deng WP. Establishment of a Promising Human Nucleus Pulposus Cell Line for Intervertebral Disc Tissue Engineering. Tissue Eng Part C Methods 2014; 20:1-10. [PMID: 23675702 DOI: 10.1089/ten.tec.2013.0048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Ming-Che Liu
- College of Oral Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
- Department of Urology, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China
| | - Wei-Hong Chen
- Stem Cell Research Center, Taipei Medical University, Taipei, Taiwan, Republic of China
- Graduate Institute of Biomedical Materials and Engineering, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Ling-Chiao Wu
- Graduate Institute of Biomedical Materials and Engineering, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Wei-Che Hsu
- Graduate Institute of Biomedical Materials and Engineering, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Wen-Cheng Lo
- School of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China
| | - Shauh-Der Yeh
- Department of Urology, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China
| | - Ming-Fu Wang
- Department of Food Science, Yuanpei University, Taipei, Taiwan, Republic of China
| | - Rong Zeng
- Department of Orthopedic Surgery, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Win-Ping Deng
- Stem Cell Research Center, Taipei Medical University, Taipei, Taiwan, Republic of China
- Graduate Institute of Biomedical Materials and Engineering, Taipei Medical University, Taipei, Taiwan, Republic of China
- Cancer Center, Taipei Medical University and Hospital, Taipei, Taiwan, Republic of China
| |
Collapse
|
34
|
Grogan SP, Chen X, Sovani S, Taniguchi N, Colwell CW, Lotz MK, D'Lima DD. Influence of cartilage extracellular matrix molecules on cell phenotype and neocartilage formation. Tissue Eng Part A 2013; 20:264-74. [PMID: 23962090 DOI: 10.1089/ten.tea.2012.0618] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interaction between chondrocytes and the cartilage extracellular matrix (ECM) is essential for maintaining the cartilage's role as a low-friction and load-bearing tissue. In this study, we examined the influence of cartilage zone-specific ECM on human articular chondrocytes (HAC) in two-dimensional and three-dimensional (3D) environments. Two culture systems were used. SYSTEM 1: HAC were cultured on cell-culture plates that had been precoated with the following ECM molecules for 7 days: decorin, biglycan, tenascin C (superficial zone), collagen type II, hyaluronan (HA) (middle and deep zones), and osteopontin (deep zone). Uncoated standard culture plates were used as controls. Expanded cells were examined for phenotypic changes using real-time polymerase chain reaction. In addition, expanded cells were placed into high-density pellet cultures for 14 days. Neocartilage formation was assessed via gene expression and histology evaluations. SYSTEM 2: HAC that were cultured on untreated plates and encapsulated in a 3D alginate scaffold were mixed with one of the zone-specific ECM molecules. Cell viability, gene expression, and histology assessments were conducted on 14-day-old tissues. In HAC monolayer culture, exposure to decorin, HA, and osteopontin increased COL2A1 and aggrecan messenger RNA (mRNA) levels compared with controls. Biglycan up-regulated aggrecan without a significant impact on COL2A1 expression; Tenascin C reduced COL2A1 expression. Neocartilage formed after preculture on tenascin C and collagen type II expressed higher COL2A1 mRNA compared with control pellets. Preculture of HAC on HA decreased both COL2A1 and aggrecan expression levels compared with controls, which was consistent with histology. Reduced proteoglycan 4 (PRG4) mRNA levels were observed in HAC pellets that had been precultured with biglycan and collagen type II. Exposing HAC to HA directly in 3D-alginate culture most effectively induced neocartilage formation, showing increased COL2A1 and aggrecan, and reduced COL1A1 compared with controls. Decorin treatments increased HAC COL2A1 mRNA levels. These data indicate that an appropriate exposure to cartilage-specific ECM proteins could be used to enhance cartilage formation and to even induce the formation of zone-specific phenotypes to improve cartilage regeneration.
Collapse
Affiliation(s)
- Shawn P Grogan
- 1 Shiley Center for Orthopaedic Research and Education , Scripps Clinic, La Jolla, California
| | | | | | | | | | | | | |
Collapse
|
35
|
Heard BJ, Fritzler MJ, Wiley JP, McAllister J, Martin L, El-Gabalawy H, Hart DA, Frank CB, Krawetz R. Intraarticular and Systemic Inflammatory Profiles May Identify Patients with Osteoarthritis. J Rheumatol 2013; 40:1379-87. [DOI: 10.3899/jrheum.121204] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective.To determine whether cytokine/chemokine profiles from synovial fluid and sera discriminate mild/moderate osteoarthritis (OA) from normal and severe OA cohorts.Methods.Multiplex technology was used to quantify expression levels for 42 cytokines in the synovial fluid of patients diagnosed with severe OA (n = 20) and mild/moderate OA (n = 12), as well as normal controls (n = 34). The same 42 cytokines were examined in serum samples of patients with severe OA (n = 26) and mild/moderate OA (n = 74) and normal individuals (n = 100). Treatment group comparisons followed by principal component analysis (PCA) and K-means clustering of the significantly different cytokines/chemokines revealed groupings of patients by physician diagnosis.Results.Differences in cytokine/chemokine levels were found between control, mild/moderate OA, and severe OA synovial fluid samples, as well as between normal and mild/moderate OA serum samples, and between control and severe OA serum samples. No differences were observed between mild/moderate and severe OA serum samples. Visual groupings based on PCA were validated by K-means analysis, with the best results obtained from the comparison of normal and mild/moderate OA serum samples with 96% of normal and 93% of mild/moderate OA samples accurately identified.Conclusion.Our study suggests that comparing the expression levels of cytokines/chemokines in synovial fluid and/or serum of patients with OA may have promise as a diagnostic platform to identify patients early in their disease course. This high-throughput low-cost assay may be able to provide clinicians with a diagnostic test to complement existing clinical and imaging modalities currently used to diagnose OA.
Collapse
|
36
|
Espinosa M, Vaisman A, Nazal N, Figueroa D, Gallegos M, Conget P. Intraarticular Administration of Dexamethasone after Mesenchymal Stem Cells Implantation Does Not Improve Significantly the Treatment of Preestablished Full-Thickness Chondral Defect in a Rabbit Model. Cartilage 2013; 4:144-52. [PMID: 26069657 PMCID: PMC4297101 DOI: 10.1177/1947603512472696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the contribution to hyaline cartilage regeneration of dexamethasone intraarticular administration after autologous mesenchymal stem cells (MSCs) implantation into a preestablished knee full-thickness chondral defect. DESIGN Full-thickness chondral defects of 4.5 × 4.5 mm(2) were surgically made in both medial femoral condyles of adult male New Zealand rabbits. Two weeks later, autologous ex vivo expanded bone marrow-derived MSCs were embedded in hyaluronic acid and implanted into the chondral defects. Immediately and every week after the intervention, dexamethasone 0.25 mg/kg was intraarticularly administered (MSC/dexa-treated group). Six weeks after MSC transplantation, the animals were euthanized and condyles were characterized molecularly according to aggrecan, collagen type II, and collagen type I gene expression (quantitative reverse transcriptase-polymerase chain reaction) and histologically (hematoxylin-eosin staining). Data of MSC/dexa-treated condyles were compared with untreated, dexa-treated, MSC-treated, or normal unlesioned condyles. RESULTS The ratio between collagen type II expression versus collagen type I expression in MSC/dexa-treated condyles was higher than one, even though the group mean value was not statistically different from that of untreated defects. Histological changes were observed between MSC/dexa-treated and untreated defects mainly in surface regularity and in hyaline matrix abundance. However, International Cartilage Repair Society score analysis did not support robust differences between those groups. CONCLUSION Intraarticular administration of dexamethasone after autologous MSC implantation into a preestablished full-thickness chondral defect does not contribute significantly to the regeneration of a tissue with molecular and histological characteristics identical to hyaline cartilage.
Collapse
Affiliation(s)
- Maximiliano Espinosa
- Instituto de Ciencias, Facultad de Medicina Clinica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Alex Vaisman
- Instituto de Ciencias, Facultad de Medicina Clinica Alemana, Universidad del Desarrollo, Santiago, Chile,Clinica Alemana, Santiago, Chile
| | - Nicolas Nazal
- Instituto de Ciencias, Facultad de Medicina Clinica Alemana, Universidad del Desarrollo, Santiago, Chile
| | | | | | - Paulette Conget
- Instituto de Ciencias, Facultad de Medicina Clinica Alemana, Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
37
|
Lo WC, Chen WH, Lin TC, Hwang SM, Zeng R, Hsu WC, Chiang YM, Liu MC, Williams DF, Deng WP. Preferential therapy for osteoarthritis by cord blood MSCs through regulation of chondrogenic cytokines. Biomaterials 2013; 34:4739-48. [PMID: 23557858 DOI: 10.1016/j.biomaterials.2013.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 03/06/2013] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) is a common rheumatic disease associated with imbalanced cartilage homeostasis which could be corrected by mesenchymal stem cells (MSCs) therapy. However, MSCs from different origins might exhibit distinct differentiation capacities. This study was undertaken to compare the therapeutic efficacies between MSCs from cord blood (CB-MSCs) and bone marrow (BM-MSCs) on OA treatment. The surface phenotypes and multipotent capacities of CB-MSCs and BM-MSCs were first characterized. The coculture commitment system was subsequently utilized for comparing the patterned molecules in stage-specific chondrogenesis of committed MSCs. For examining the therapeutic efficacies, committed CB-MSCs and BM-MSCs were encapsulated in neo-cartilage and subjected into pro-inflammatory cytokine environment. Finally, chondrogenic and inflammatory cytokine profiles in committed MSCs were evaluated. CB-MSCs and BM-MSCs were both negative for hematopoietic markers and positive for adhesion and mesenchymal cell markers. The CB-MSCs showed a markedly higher chondrogenic potential and relatively lower osteogenic and adipogenic capacities than BM-MSCs. During chondrogenesis, the committed CB-MSCs also showed significant increases in cell proliferation, adhesion molecules, signaling molecules, and chondrogenic-specific gene expressions in a coculture system. For the therapeutic efficacies, the committed CB-MSCs could strongly recover the pro-inflammatory cytokines diminished-Col II and proteoglycan expressions in a 3D arthritic model. The IL-10, ICAM-1 and TGF-β1 were also up-regulated in committed CB-MSCs analyzed by using cytokine profiling. Our data demonstrate that CB-MSCs possess specific advantages in cartilage regeneration over BM-MSCs. The CB-MSCs showed a better therapeutic potential that can contribute to advanced cell-based transplantation for clinical OA therapy.
Collapse
Affiliation(s)
- Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wu X, Cai ZD, Lou LM, Chen ZR. The effects of inhibiting hedgehog signaling pathways by using specific antagonist cyclopamine on the chondrogenic differentiation of mesenchymal stem cells. Int J Mol Sci 2013; 14:5966-77. [PMID: 23493060 PMCID: PMC3634433 DOI: 10.3390/ijms14035966] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/05/2013] [Accepted: 03/07/2013] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate the effects of cyclopamine, a specific inhibitor of Hedgehog signaling pathways, on the chondrogenic differentiation of mesenchymal stem cells (MSCs). During culture, the experimental groups were treated with cyclopamine and their cell proliferation status was assessed using the MTT test. The extra-bone cellular matrix (ECM) and Collagen II (Col II) was detected by toluidine blue staining and immunohistochemistry of cells. The concentrations of Col II and aggrecan in the culture solution and cytosol were detected using ELISA on the 7th, 14th, and 21st days of cyclopamine induction. Gene and protein expression of Col II and aggrecan were analyzed on the 14th day of cyclopamine induction using real-time PCR and western blot analyses. No significant differences in proliferation of mesenchymal stem cells were found between the control group and the group treated with cyclopamine. Compared to the blank control group, the ECM level was low and the protein and mRNA concentrations of Collagen II (Col II) and aggrecan in the culture solution and cytosol, respectively, were significantly reduced in the experimental group. The Smo acted as a key point in the regulations of Hedgehog signaling pathway on the chondrogenic differentiation of rabbit MSCs.
Collapse
Affiliation(s)
- Xing Wu
- Department of Orthopaedics, Shanghai tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China; E-Mails: (Z.-D.C.); (L.-M.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-21-6630-0588; Fax: +86-21-6630-1051
| | - Zheng-Dong Cai
- Department of Orthopaedics, Shanghai tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China; E-Mails: (Z.-D.C.); (L.-M.L.)
| | - Lei-Ming Lou
- Department of Orthopaedics, Shanghai tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China; E-Mails: (Z.-D.C.); (L.-M.L.)
| | - Zheng-Rong Chen
- Department of Orthopaedics, Shanghai Zhongshan Hospital, Fudan University School of Medicine, Shanghai 200032, China; E-Mail:
| |
Collapse
|
39
|
Jiang J, Papoutsakis ET. Stem-cell niche based comparative analysis of chemical and nano-mechanical material properties impacting ex vivo expansion and differentiation of hematopoietic and mesenchymal stem cells. Adv Healthc Mater 2013. [PMID: 23184458 DOI: 10.1002/adhm.201200169] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The ability of stem cells to self-renew with minimal or no differentiation and, when appropriately cued, to give rise to many types of progenitor and mature cells, is the basis for applications in regenerative and transfusion medicine, but also in drug discovery and in vitro toxicology. Inspired by the complex interactions between stem cells and their microenvironment, the so-called stem-cell niche, the properties of supporting biomaterials, including surface biochemistry, topography (type, size, organization, and geometry of nanostructures), and mechanical properties, have been identified as important determinants of stem-cell fate in vitro. 3D culture environments that could recapitulate the complexity of the in vivo stem-cell microenvironment could further expand the complexity and repertoire of engineered environments with exciting translational applications. Herein, the material aspects that affect the expansion and differentiation fate of adult hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stem cells (MSCs), two powerful cell types that co-reside in the bone-marrow niche, but with distinct, sometime complementary, differentiation fates, properties, and translational applications, are examined. Although MSCs are adherent cells and, in contrast, HSPCs are non- or weakly adherent cells, both can sense and respond to material properties, including surface (bio)chemistry, ECM composition, topography, and matrix elasticity, possibly through similar molecular mechanisms.
Collapse
Affiliation(s)
- Jinlin Jiang
- Dept. of Chemical & Biomolecular Engineering, University of Delaware, Newark, DE 19711, USA
| | | |
Collapse
|
40
|
The role of the ERK1/2 pathway as an alternative to the aging-diminished cyclic AMP pathway in calcitonin-mediated chondrogenesis in human nucleus pulposus. Biomaterials 2012; 33:8256-64. [PMID: 22938762 DOI: 10.1016/j.biomaterials.2012.06.088] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 06/28/2012] [Indexed: 01/08/2023]
Abstract
Human disc degeneration initiated by aging in the central nucleus pulposus (hNP) is an irreversible process and the recovery has become seriously emerging. In this study, the related mechanisms of calcitonin on the regeneration of hNP and the effects of calcitonin on the age-related alterations were examined. The harvested hNP population was designated as YhNP (from young donor, age <50) and OhNP (from old donor, age >50). Primary OhNP cells showed more hypertrophic phenotypes than YhNP. However, calcitonin (10(-8)-10(-6) M) was able to induce the same chondrogenesis in both YhNP and OhNP by elevating chondrogenic specific-mRNA and protein expressions. Their cell viabilities were increased with calcitonin treatment. No significant differences of calcitonin receptor (CTR) were expressed between YhNP and OhNP cells. Interestingly, in calcitonin-induced pathways for chondrogenesis, highly increased cyclic AMP (cAMP) was detected in YhNP but was strongly diminished by aging in OhNP after calcitonin treatment. However, to maintain the chondrogenesis, calcitonin-induced an alterative phosphorylated ERK1/2 (p-ERK) in both cells. After inhibiting ERK1/2 by PD98059, calcitonin-induced chondrogenesis in OhNP was almost restrained while YhNP cells were not affected. Our results demonstrated that the regeneration of calcitonin on hNP was maintained with aging which was satisfied by an alternative signaling pathway. Therefore, calcitonin shows great potential for clinical therapy for disc regeneration without aging considerations.
Collapse
|
41
|
Chiu LH, Yeh TS, Huang HM, Leu SJ, Yang CB, Tsai YH. Diverse effects of type II collagen on osteogenic and adipogenic differentiation of mesenchymal stem cells. J Cell Physiol 2012; 227:2412-20. [PMID: 21826655 DOI: 10.1002/jcp.22976] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Type II collagen is known to modulate chondrogenesis of mesenchymal stem cells (MSCs). In this study, MSCs from human bone marrow aspirates were used to study the modulating effects of type II collagen on MSC differentiation during the early stages of osteogenesis and adipogenesis. With osteogenic induction, MSCs cultured on the type II collagen-coated surface showed an enhanced calcium deposition level with increasing mRNA expressions of RUNX2, osteocalcin, and alkaline phosphatase. A synthetic integrin binding peptide, which specifically interacts with the I-domain of α(1)β(1)/α(2)β(1) integrins significantly blocks the mineralization-enhancing effect of type II collagen. MSCs attached on the type II collagen-coated plates exhibited expanded cell morphology with increasing spreading area, and the pretreatment of cells with integrin α(1)β(1) or α(2)β(1)-blocking antibody reduced the effect. The phosphorylation levels of FAK, ERK, and JNK significantly increased in the MSCs that attached on the type II collagen-coated plates. On the contrary, the mineralization-enhancing effect of type II collagen was diminished by JNK and MEK inhibitors. Furthermore, type II collagen blocked the adipogenic differentiation of MSCs, and this effect is rescued by JNK and MEK inhibitors. In conclusion, type II collagen facilitates osteogenesis and suppresses adipogenesis during early stage MSC differentiation. Such effects are integrin binding-mediated and conducted through FAK-JNK and/or FAK-ERK signaling cascades. These results inspire a novel strategy encompassing type II collagen in bone tissue engineering.
Collapse
Affiliation(s)
- Li-Hsuan Chiu
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
42
|
Ge Z, Li C, Heng BC, Cao G, Yang Z. Functional biomaterials for cartilage regeneration. J Biomed Mater Res A 2012; 100:2526-36. [PMID: 22492677 DOI: 10.1002/jbm.a.34147] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 01/30/2012] [Accepted: 02/19/2012] [Indexed: 12/22/2022]
Abstract
The injury and degeneration of articular cartilage and associated arthritis are leading causes of disability worldwide. Cartilage tissue engineering as a treatment modality for cartilage defects has been investigated for over 20 years. Various scaffold materials have been developed for this purpose, but has yet to achieve feasibility and effectiveness for widespread clinical use. Currently, the regeneration of articular cartilage remains a formidable challenge, due to the complex physiology of cartilage tissue and its poor healing capacity. Although intensive research has been focused on the developmental biology and regeneration of cartilage tissue and a diverse plethora of biomaterials have been developed for this purpose, cartilage regeneration is still suboptimal, such as lacking a layered structure, mechanical mismatch with native cartilage and inadequate integration between native tissue and implanted scaffold. The ideal scaffold material should have versatile properties that actively contribute to cartilage regeneration. Functional scaffold materials may overcome the various challenges faced in cartilage tissue engineering by providing essential biological, mechanical, and physical/chemical signaling cues through innovative design. This review thus focuses on the complex structure of native articular cartilage, the critical properties of scaffolds required for cartilage regeneration, present strategies for scaffold design, and future directions for cartilage regeneration with functional scaffold materials.
Collapse
Affiliation(s)
- Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, People's Republic of China.
| | | | | | | | | |
Collapse
|
43
|
Vaisman A, Figueroa D, Calvo R, Espinosa M, Melean P, Gallegos M, Conget P. Steroids and Platelet-Rich Plasma as Coadjuvants to Microfracture for the Treatment of Chondral Lesions in an Animal Model: Can the Healing Be Enhanced? Cartilage 2012; 3:118-27. [PMID: 26069625 PMCID: PMC4297125 DOI: 10.1177/1947603511422053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the contribution to hyaline cartilage regeneration of the microfracture (MFx) technique plus intraarticular betamethasone (BMS) or platelet-rich plasma (PRP). DESIGN Full-thickness chondral defects of 3 × 6 mm(2) were surgically performed in both femoral condyles of each knee in 13 New Zealand rabbits and then treated with MFx associated with intraarticular BMS or PRP. At 12 weeks postimplantation, the animals were killed and the condyles were characterized macroscopically, molecularly according to collagen type II and I gene expression (quantitative reverse transcriptase-polymerase chain reaction), and histologically (hematoxylin-eosin staining). For the latter, samples were scored using the International Cartilage Repair Society visual histological scale. Data of MFx/BMS-treated and MFx/PRP-treated condyles were compared against untreated, MFx-treated, or normal condyles without lesions. RESULTS Our macroscopic findings showed that in MFx/BMS-treated and MFx/PRP-treated groups, the defects were filled with an irregular, partially rough tissue similar to the MFx-treated group. No differences in the ratio between collagen type II versus collagen type I expression were observed among groups. Histological changes were observed between MFx/BMS-treated and MFx/PRP-treated groups versus untreated defects mainly in surface regularity and cell distribution. However, International Cartilage Repair Society score analysis did not support statistical differences between MFx/BMS-treated and MFx/PRP-treated groups versus MFx-treated group. CONCLUSIONS These results provide evidence that the use of intraarticular BMS or PRP as coadjuvants to the microfracture technique in the treatment of acute chondral lesions is not associated with a significant improvement of hyaline cartilage regeneration.
Collapse
Affiliation(s)
- Alex Vaisman
- Orthopedic Surgery Department, Clínica Alemana de Santiago, Chile
- Faculty of Medicine, Clínica Alemana–Universidad del Desarrollo
| | - David Figueroa
- Orthopedic Surgery Department, Clínica Alemana de Santiago, Chile
- Faculty of Medicine, Clínica Alemana–Universidad del Desarrollo
| | - Rafael Calvo
- Orthopedic Surgery Department, Clínica Alemana de Santiago, Chile
- Faculty of Medicine, Clínica Alemana–Universidad del Desarrollo
| | | | - Patricio Melean
- Orthopedic Surgery Department, Hospital Padre Hurtado, Santiago, Chile
| | | | - Paulette Conget
- Faculty of Medicine, Clínica Alemana–Universidad del Desarrollo
| |
Collapse
|
44
|
Rathore A, Cleary M, Naito Y, Rocco K, Breuer C. Development of tissue engineered vascular grafts and application of nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2012; 4:257-72. [DOI: 10.1002/wnan.1166] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
45
|
Chiu LH, Chen SC, Wu KC, Yang CB, Fang CL, Lai WFT, Tsai YH. Differential effect of ECM molecules on re-expression of cartilaginous markers in near quiescent human chondrocytes. J Cell Physiol 2011; 226:1981-8. [PMID: 21520049 DOI: 10.1002/jcp.22530] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The limited source of healthy primary chondrocytes restricts the clinical application of tissue engineering for cartilage repair. Therefore, method to maintain or restore the chondrocyte phenotype during in vitro expansion is essential. The objective of this study is to establish the beneficial effect of ECM molecules on restoring the re-expression of cartilaginous markers in primary human chondrocytes after extensive monolayer expansion. During the course of chondrocyte serial expansion, COL2A1, SOX9, and AGN mRNA expression levels, and GAG accumulation level were reduced significantly in serially passaged cells. Exogenous type II collagen dose-dependently elevated GAG level and induced the re-expression of cartilaginous marker mRNAs in P7 chondrocytes. Chondroitin sulfate did not show significant effect on P7 chondrocytes, while hyaluronic acid inhibited the expression of SOX9 and AGN mRNAs. Upon treatment with type II collagen, FAK, ERK1/2, and JNK were activated via phosphorylation in P7 chondrocytes within 15 min. Furthermore, GFOGER integrin blocking peptide, MEK inhibitor and JNK inhibitor, not p38 inhibitor, significantly reduced the type II collagen-induced GAG deposition level. Finally, in the presence of TGF-β1 and IGF-I, P7 chondrocytes cultured in 3D type II collagen matrix exhibited better cartilaginous features than those cells cultured in the type I collagen matrix. In conclusion, type II collagen alone can effectively restore cartilaginous features of expanded P7 human chondrocytes. It is probably mediated via the activation of FAK-ERK1/2 and FAK-JNK signaling pathways. The potential application of type II collagen in expanding a scarcity of healthy chondrocytes in vitro for further tissue engineering is implicated.
Collapse
Affiliation(s)
- Li-Hsuan Chiu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
46
|
Wu CC, Chen WH, Zao B, Lai PL, Lin TC, Lo HY, Shieh YH, Wu CH, Deng WP. Regenerative potentials of platelet-rich plasma enhanced by collagen in retrieving pro-inflammatory cytokine-inhibited chondrogenesis. Biomaterials 2011; 32:5847-54. [PMID: 21616530 DOI: 10.1016/j.biomaterials.2011.05.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 05/01/2011] [Indexed: 12/14/2022]
Abstract
This study was undertaken to evaluate the role of collagen matrix to enhance platelet-rich plasma (PRP) effects on pro-inflammatory cytokine-induced arthritic model. We have previously demonstrated the highly regenerative roles of PRP to restore disc degeneration and osteoporosis. In this study, PRP modulated by collagen matrix was used as a regenerative and anti-inflammatory mediator to rescue the chondrocyte degeneration induced by pro-inflammatory cytokines IL-1β (10 ng/ml)+TNF-α (20 ng/ml). First, the MTT result indicated that 1 ng/ml TGF-β1 in PRP showed an optimal dosage for chondrocytes proliferation. The chondrogenic-specific gene expressions were rescued by PRP from the inhibition of IL-1β+TNF-α, especially under the modulation of collagen matrix. The inflammatory molecules activated by IL-1β+TNF-α were also significantly diminished by PRP with collagen matrix. The membrane receptors integrin α1β1 and CD44 were strongly inhibited by IL-1β+TNF-α, while this inhibition was then recovered by PRP in collagen coating condition. In a 3D model encapsulated with collagen, PRP-induced chondrogenesis were highly enhanced, such as strong restoration of type II collagen and proteoglycan from the inhibition of IL-1β+TNF-α. The result indicated that collagen matrix enhances the effect of PRP on chondrogenesis in response to pro-inflammatory cytokines. The combination of PRP and collagen matrix might facilitate a physiological microenvironment beneficial for maintaining chondrocyte homeostasis and represents an advanced osteoarthritis therapy for clinical applications.
Collapse
Affiliation(s)
- Chia-Che Wu
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hoshiba T, Lu H, Yamada T, Kawazoe N, Tateishi T, Chen G. Effects of extracellular matrices derived from different cell sources on chondrocyte functions. Biotechnol Prog 2011; 27:788-95. [PMID: 21509950 DOI: 10.1002/btpr.592] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 02/01/2011] [Indexed: 12/28/2022]
Abstract
Cell-derived extracellular matrices (ECMs) are a key factor in regulating cell functions in tissue engineering and regenerative medicine. The fact that cells are surrounded by their specific ECM in vivo elicits the need to elucidate the effects of ECM derived from different cell sources on cell functions. Here, three types of ECM were prepared by decellularizing cultured chondrocytes, fibroblasts, and mesenchymal stem cells (MSC) and used for chondrocyte culture to compare their effects on chondrocyte adhesion, proliferation, and differentiation. Chondrocyte adhesion to the chondrocyte-derived ECM was greater than those to the fibroblast- and MSC-derived ECM. Chondrocyte proliferation on the chondrocyte-derived ECM was lower than those on the fibroblast- and MSC-derived ECM. The ECM showed no evident effect on chondrocyte differentiation. The effects of ECM on cell functions depended on the cell source used to prepare the ECM.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Biomaterials Center, National Institute for Materials Science, Tsukuba 305-0044, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Hwang NS, Varghese S, Li H, Elisseeff J. Regulation of osteogenic and chondrogenic differentiation of mesenchymal stem cells in PEG-ECM hydrogels. Cell Tissue Res 2011; 344:499-509. [PMID: 21503601 DOI: 10.1007/s00441-011-1153-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 02/25/2011] [Indexed: 12/13/2022]
Abstract
Bone-marrow-derived mesenchymal stem cells (MSCs) are candidates for regeneration applications in musculoskeletal tissue such as cartilage and bone. Various soluble factors in the form of growth factors and cytokines have been widely studied for directing the chondrogenic and osteogenic differentiation of MSCs, but little is known about the way that the composition of extracellular matrix (ECM) components in three-dimensional microenvironments plays a role in regulating the differentiation of MSCs. To define whether ECM components influence the regulation of osteogenic and chondrogenic differentiation by MSCs, we encapsulated MSCs in poly-(ethylene glycol)-based (PEG-based) hydrogels containing exogenous type I collagen, type II collagen, or hyaluronic acids (HA) and cultured them for up to 6 weeks in chondrogenic medium containing transforming growth factor-β1 (10 ng/ml) or osteogenic medium. Actin cytoskeleton organization and cellular morphology were strongly dependent on which ECM components were added to the PEG-based hydrogels. Additionally, chondrogenic differentiation of MSCs was marginally enhanced in collagen-matrix-based hydrogels, whereas osteogenic differentiation, as measured by calcium accumulation, was induced in HA-containing hydrogels. Thus, the microenvironments created by exogenous ECM components seem to modulate the fate of MSC differentiation.
Collapse
Affiliation(s)
- Nathaniel S Hwang
- Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
49
|
Hübner S, Efthymiadis A. Histochemistry and cell biology: the annual review 2010. Histochem Cell Biol 2011; 135:111-40. [PMID: 21279376 DOI: 10.1007/s00418-011-0781-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
Abstract
This review summarizes recent advances in histochemistry and cell biology which complement and extend our knowledge regarding various aspects of protein functions, cell and tissue biology, employing appropriate in vivo model systems in conjunction with established and novel approaches. In this context several non-expected results and discoveries were obtained which paved the way of research into new directions. Once the reader embarks on reading this review, it quickly becomes quite obvious that the studies contribute not only to a better understanding of fundamental biological processes but also provide use-oriented aspects that can be derived therefrom.
Collapse
Affiliation(s)
- Stefan Hübner
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstrasse 6, 97070 Würzburg, Germany.
| | | |
Collapse
|
50
|
Raabe O, Reich C, Wenisch S, Hild A, Burg-Roderfeld M, Siebert HC, Arnhold S. Hydrolyzed fish collagen induced chondrogenic differentiation of equine adipose tissue-derived stromal cells. Histochem Cell Biol 2010; 134:545-54. [PMID: 21076963 DOI: 10.1007/s00418-010-0760-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2010] [Indexed: 10/18/2022]
Abstract
Adipose-derived stromal cells (ADSCs) are multipotent cells which, in the presence of appropriate stimuli, can differentiate into various lineages such as the osteogenic, adipogenic and chondrogenic. In this study, we investigated the effect of transforming growth factor beta 1 (TGF-β1) in comparison to hydrolyzed fish collagen in terms of the chondrogenic differentiation potential of ADSCs. ADSCs were isolated from subcutaneous fat of horses by liposuction. Chondrogenesis was investigated using a pellet culture system. The differentiation medium was either supplemented with TGF-β1 (5 ng/ml) or fish collagen (0.5 mg/ml) for a 3 week period. After the 3 weeks in vitro differentiation, RT-PCR and histological staining for proteoglycan synthesis and type II collagen were performed to evaluate the degree of chondrogenic differentiation and the formation of cartilaginous extracellular matrix (ECM). The differentiation of ADSCs induced by TGF-β1 showed a high expression of glycosaminoglycan (GAG). Histological analysis of cultures stimulated by hydrolyzed fish collagen demonstrated an even higher GAG expression than cultures stimulated under standard conditions by TGF-β1. The expression of cartilage-specific type II collagen and Sox9 was about the same in both stimulated cultures. In this study, chondrogenesis was as effectively induced by hydrolyzed fish collagen as it was successfully induced by TGF-β1. These findings demonstrated that hydrolyzed fish collagen alone has the potential to induce and maintain ADSCs-derived chondrogenesis. These results support the application of ADSCs in equine veterinary tissue engineering, especially for cartilage repair.
Collapse
Affiliation(s)
- O Raabe
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig University of Giessen, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|