1
|
Schumacher A, Mucha P, Puchalska I, Deptuła M, Wardowska A, Tymińska A, Filipowicz N, Mieczkowska A, Sachadyn P, Piotrowski A, Pikuła M, Cichorek M. Angiopoietin-like growth factor-derived peptides as biological activators of adipose-derived mesenchymal stromal cells. Biomed Pharmacother 2024; 177:117052. [PMID: 38943988 DOI: 10.1016/j.biopha.2024.117052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024] Open
Abstract
Adipose-derived mesenchymal stromal cells (AD-MSCs) are an essential issue in modern medicine. Extensive preclinical and clinical studies have shown that mesenchymal stromal/stem cells, including AD-MSCs, have specific properties (ability to differentiate into other cells, recruitment to the site of injury) of particular importance in the regenerative process. Ongoing research aims to elucidate factors supporting AD-MSC culture and differentiation in vitro. Angiopoietin-like proteins (ANGPTLs), known for their pleiotropic effects in lipid and glucose metabolism, may play a significant role in this context. Regeneration is a complex and dynamic process controlled by many factors. ANGPTL6 (Angiopoietin-related growth factor, AGF), among many activities modulated the biological activity of stem cells. This study examined the influence of synthesized AGF-derived peptides, designated as AGF9 and AGF27, on AD-MSCs. AGF9 and AGF27 enhanced the viability and migration of AD-MSCs and acted as a chemotactic factor for these cells. AGF9 stimulated chondrogenesis and lipid synthesis during AD-MSCs differentiation, influenced AD-MSCs cytokine secretion and modulated transcriptome for such basic cell activities as migration, transport of molecules, and apoptosis. The ability of AGF9 to modulate the biological activity of AD-MSCs warrants the consideration of this peptide a noteworthy therapeutic agent that deserves further investigation for applications in regenerative medicine.
Collapse
Affiliation(s)
- Adriana Schumacher
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Piotr Mucha
- Department of Molecular Biochemistry, University of Gdansk, Wita Stwosza 63 St, Gdansk 80-308, Poland
| | - Izabela Puchalska
- Department of Molecular Biochemistry, University of Gdansk, Wita Stwosza 63 St, Gdansk 80-308, Poland
| | - Milena Deptuła
- Division of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Anna Wardowska
- Department of Physiopathology, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Agata Tymińska
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Natalia Filipowicz
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Alina Mieczkowska
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Gdansk University of Technology, Narutowicza 11/12 St, Gdansk 80-233, Poland
| | - Arkadiusz Piotrowski
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Michał Pikuła
- Division of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Miroslawa Cichorek
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland.
| |
Collapse
|
2
|
Yang Y, Koga H, Nakagawa Y, Nakamura T, Katagiri H, Takada R, Katakura M, Tsuji K, Sekiya I, Miyatake K. Characteristics of the synovial microenvironment and synovial mesenchymal stem cells with hip osteoarthritis of different bone morphologies. Arthritis Res Ther 2024; 26:17. [PMID: 38200556 PMCID: PMC10777653 DOI: 10.1186/s13075-023-03252-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Variations in bone morphology in patients with hip osteoarthritis (HOA) can be broadly categorized into three types: atrophic, normotrophic, and hypertrophic. Despite the investigations examining clinical elements, such as bone morphology, pain, and range of motion, our understanding of the pathogenesis of HOA remains limited. Previous studies have suggested that osteophytes typically originate at the interface of the joint cartilage, periosteum, and synovium, potentially implicating synovial mesenchymal stem cells (SMSCs) in the process. This study aimed to investigate the potential factors that drive the development of bone morphological features in HOA by investigating the characteristics of the synovium, differentiation potential of SMSCs, and composition of synovial fluid in different types of HOA. METHODS Synovial tissue and fluid were collected from 30 patients who underwent total hip arthroplasty (THA) with the variable bone morphology of HOA patients. RNA sequencing analysis and quantitative reverse transcription-polymerase chain reaction (RT-qPCR) were performed to analyse the genes in the normotrophic and hypertrophic synovial tissue. SMSCs were isolated and cultured from the normotrophic and hypertrophic synovial tissues of each hip joint in accordance with the variable bone morphology of HOA patients. Cell differentiation potential was compared using differentiation and colony-forming unit assays. Cytokine array was performed to analyse the protein expression in the synovial fluid. RESULTS In the RNA sequencing analysis, 103 differentially expressed genes (DEGs) were identified, predominantly related to the interleukin 17 (IL-17) signalling pathway. Using a protein-protein interaction (PPI) network, 20 hub genes were identified, including MYC, CXCL8, ATF3, NR4A1, ZC3H12A, NR4A2, FOSB, and FOSL1. Among these hub genes, four belonged to the AP-1 family. There were no significant differences in the tri-lineage differentiation potential and colony-forming capacity of SMSCs. However, RT-qPCR revealed elevated SOX9 expression levels in synovial tissues from the hypertrophic group. The cytokine array demonstrated significantly higher levels of CXCL8, MMP9, and VEGF in the synovial fluid of the hypertrophic group than in the normotrophic group, with CXCL8 and MMP9 being significantly expressed in the hypertrophic synovium. CONCLUSION Upregulation of AP-1 family genes in the synovium and increased concentrations of CXCL8, MMP9, and VEGF were detected in the synovial fluid of the hypertrophic group of HOA patients, potentially stimulating the differentiation of SMSCs towards the cartilage and thereby contributing to severe osteophyte formation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Nakagawa
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomasa Nakamura
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroki Katagiri
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Orthopaedic Surgery, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Ryohei Takada
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mai Katakura
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazumasa Miyatake
- Department of Joint Surgery and Sports Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
3
|
Chen X, Liang XM, Zheng J, Dong YH. Stromal cell-derived factor-1α regulates chondrogenic differentiation via activation of the Wnt/β-catenin pathway in mesenchymal stem cells. World J Stem Cells 2023; 15:490-501. [PMID: 37342217 PMCID: PMC10277961 DOI: 10.4252/wjsc.v15.i5.490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/21/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been applied to treat degenerative articular diseases, and stromal cell-derived factor-1α (SDF-1α) may enhance their therapeutic efficacy. However, the regulatory effects of SDF-1α on cartilage differentiation remain largely unknown. Identifying the specific regulatory effects of SDF-1α on MSCs will provide a useful target for the treatment of degenerative articular diseases.
AIM To explore the role and mechanism of SDF-1α in cartilage differentiation of MSCs and primary chondrocytes.
METHODS The expression level of C-X-C chemokine receptor 4 (CXCR4) in MSCs was assessed by immunofluorescence. MSCs treated with SDF-1α were stained for alkaline phosphatase (ALP) and with Alcian blue to observe differentiation. Western blot analysis was used to examine the expression of SRY-box transcription factor 9, aggrecan, collagen II, runt-related transcription factor 2, collagen X, and matrix metalloproteinase (MMP)13 in untreated MSCs, of aggrecan, collagen II, collagen X, and MMP13 in SDF-1α-treated primary chondrocytes, of glycogen synthase kinase 3β (GSK3β) p-GSK3β and β-catenin expression in SDF-1α-treated MSCs, and of aggrecan, collagen X, and MMP13 in SDF-1α-treated MSCs in the presence or absence of ICG-001 (SDF-1α inhibitor).
RESULTS Immunofluorescence showed CXCR4 expression in the membranes of MSCs. ALP stain was intensified in MSCs treated with SDF-1α for 14 d. The SDF-1α treatment promoted expression of collagen X and MMP13 during cartilage differentiation, whereas it had no effect on the expression of collagen II or aggrecan nor on the formation of cartilage matrix in MSCs. Further, those SDF-1α-mediated effects on MSCs were validated in primary chondrocytes. SDF-1α promoted the expression of p-GSK3β and β-catenin in MSCs. And, finally, inhibition of this pathway by ICG-001 (5 µmol/L) neutralized the SDF-1α-mediated up-regulation of collagen X and MMP13 expression in MSCs.
CONCLUSION SDF-1α may promote hypertrophic cartilage differentiation in MSCs by activating the Wnt/β-catenin pathway. These findings provide further evidence for the use of MSCs and SDF-1α in the treatment of cartilage degeneration and osteoarthritis.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Xia-Ming Liang
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Yong-Hui Dong
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
4
|
Du X, Cai L, Xie J, Zhou X. The role of TGF-beta3 in cartilage development and osteoarthritis. Bone Res 2023; 11:2. [PMID: 36588106 PMCID: PMC9806111 DOI: 10.1038/s41413-022-00239-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/25/2022] [Accepted: 11/03/2022] [Indexed: 01/03/2023] Open
Abstract
Articular cartilage serves as a low-friction, load-bearing tissue without the support with blood vessels, lymphatics and nerves, making its repair a big challenge. Transforming growth factor-beta 3 (TGF-β3), a vital member of the highly conserved TGF-β superfamily, plays a versatile role in cartilage physiology and pathology. TGF-β3 influences the whole life cycle of chondrocytes and mediates a series of cellular responses, including cell survival, proliferation, migration, and differentiation. Since TGF-β3 is involved in maintaining the balance between chondrogenic differentiation and chondrocyte hypertrophy, its regulatory role is especially important to cartilage development. Increased TGF-β3 plays a dual role: in healthy tissues, it can facilitate chondrocyte viability, but in osteoarthritic chondrocytes, it can accelerate the progression of disease. Recently, TGF-β3 has been recognized as a potential therapeutic target for osteoarthritis (OA) owing to its protective effect, which it confers by enhancing the recruitment of autologous mesenchymal stem cells (MSCs) to damaged cartilage. However, the biological mechanism of TGF-β3 action in cartilage development and OA is not well understood. In this review, we systematically summarize recent progress in the research on TGF-β3 in cartilage physiology and pathology, providing up-to-date strategies for cartilage repair and preventive treatment.
Collapse
Affiliation(s)
- Xinmei Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
5
|
Oct4 facilitates chondrogenic differentiation of mesenchymal stem cells by mediating CIP2A expression. Cell Tissue Res 2022; 389:11-21. [PMID: 35435493 DOI: 10.1007/s00441-022-03619-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Bone development and cartilage formation require strict modulation of gene expression for mesenchymal stem cells (MSCs) to progress through their differentiation stages. Octamer-binding transcription factor 4 (Oct4) expression is generally restricted to developing embryonic pluripotent cells, but its role in chondrogenic differentiation (CD) of MSCs remains unclear. We therefore investigated the role of Oct4 in CD using a microarray, quantitative real-time polymerase chain reaction, and western blotting. The expression of Oct4 was elevated when the CD of cultured MSCs was induced. Silencing Oct4 damaged MSC growth and proliferation and decreased CD, indicated by decreased cartilage matrix formation and the expression of Col2a1, Col10a1, Acan, and Sox9. We found a positive correlation between the expression of CIP2A, a natural inhibitor of protein phosphatase 2A (PP2A) and that of Oct4. Cellular inhibitor of PP2A (CIP2A) expression gradually increased after CD. Overexpression of CIP2A in MSCs with Oct4 depletion promoted cartilage matrix deposition as well as Col2a1, Col10a1, Acan, and Sox9 expression. The chondrogenic induction triggered c-Myc, Akt, ERK, and MEK phosphorylation and upregulated c-Myc and mTOR expression, which was downregulated upon Oct4 knockdown and restored by CIP2A overexpression. These findings indicated that Oct4 functions as an essential chondrogenesis regulator, partly via the CIP2A/PP2A pathway.
Collapse
|
6
|
Costela Ruiz VJ, Melguizo Rodríguez L, Illescas Montes R, García Recio E, Arias Santiago S, Ruiz C, De Luna Bertos E. Human adipose tissue-derived mesenchymal stromal cells and their phagocytic capacity. J Cell Mol Med 2021; 26:178-185. [PMID: 34854223 PMCID: PMC8742185 DOI: 10.1111/jcmm.17070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have evidenced considerable therapeutic potential in numerous clinical fields, especially in tissue regeneration. The immunological characteristics of this cell population include the expression of Toll‐like receptors and mannose receptors, among others. The study objective was to determine whether MSCs have phagocytic capacity against different target particles. We isolated and characterized three human adipose tissue MSC (HAT‐MSC) lines from three patients and analysed their phagocytic capacity by flow cytometry, using fluorescent latex beads, and by transmission electron microscopy, using Escherichia coli, Staphylococcus aureus and Candida albicans as biological materials and latex beads as non‐biological material. The results demonstrate that HAT‐MSCs can phagocyte particles of different nature and size. The percentage of phagocytic cells ranged between 33.8% and 56.2% (mean of 44.37% ± 11.253) according to the cell line, and a high phagocytic index was observed. The high phagocytic capacity observed in MSCs, which have known regenerative potential, may offer an advance in the approach to certain local and systemic infections.
Collapse
Affiliation(s)
- Víctor J Costela Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Lucía Melguizo Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Rebeca Illescas Montes
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Enrique García Recio
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Salvador Arias Santiago
- Biosanitary Research Institute, ibs Granada, Granada, Spain.,Surgical Medical Dermatology and Venereology Service, Department of Medicine, Virgen de las Nieves Hospital, Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain.,Institute of Neuroscience, Centre for Medical Research (CIBM), Health Technology Park (PTS), University of Granada, Granada, Spain
| | - Elvira De Luna Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| |
Collapse
|
7
|
Comparative evaluation of pathways and gene expression profile similarity in differentiated stem cells versus normal adult cells in seven human tissues. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
8
|
Robert AW, Marcon BH, Dallagiovanna B, Shigunov P. Adipogenesis, Osteogenesis, and Chondrogenesis of Human Mesenchymal Stem/Stromal Cells: A Comparative Transcriptome Approach. Front Cell Dev Biol 2020; 8:561. [PMID: 32733882 PMCID: PMC7362937 DOI: 10.3389/fcell.2020.00561] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022] Open
Abstract
Adipogenesis, osteogenesis and chondrogenesis of human mesenchymal stem/stromal cells (MSC) are complex and highly regulated processes. Over the years, several studies have focused on understanding the mechanisms involved in the MSC commitment to the osteogenic, adipogenic and/or chondrogenic phenotypes. High-throughput methodologies have been used to investigate the gene expression profile during differentiation. Association of data analysis of mRNAs, microRNAs, circular RNAs and long non-coding RNAs, obtained at different time points over these processes, are important to depict the complexity of differentiation. This review will discuss the results that were highlighted in transcriptome analyses of MSC undergoing adipogenic, osteogenic and chondrogenic differentiation. The focus is to shed light on key molecules, main signaling pathways and biological processes related to different time points of adipogenesis, osteogenesis and chondrogenesis.
Collapse
Affiliation(s)
- Anny W Robert
- Instituto Carlos Chagas - Fiocruz Paraná, Curitiba, Brazil
| | - Bruna H Marcon
- Instituto Carlos Chagas - Fiocruz Paraná, Curitiba, Brazil
| | | | | |
Collapse
|
9
|
Zaffaroni G, Okawa S, Morales-Ruiz M, del Sol A. An integrative method to predict signalling perturbations for cellular transitions. Nucleic Acids Res 2020; 47:e72. [PMID: 30949696 PMCID: PMC6614844 DOI: 10.1093/nar/gkz232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/22/2019] [Accepted: 03/22/2019] [Indexed: 12/19/2022] Open
Abstract
Induction of specific cellular transitions is of clinical importance, as it allows to revert disease cellular phenotype, or induce cellular reprogramming and differentiation for regenerative medicine. Signalling is a convenient way to accomplish such transitions without transfer of genetic material. Here we present the first general computational method that systematically predicts signalling molecules, whose perturbations induce desired cellular transitions. This probabilistic method integrates gene regulatory networks (GRNs) with manually-curated signalling pathways obtained from MetaCore from Clarivate Analytics, to model how signalling cues are received and processed in the GRN. The method was applied to 219 cellular transition examples, including cell type transitions, and overall correctly predicted experimentally validated signalling molecules, consistently outperforming other well-established approaches, such as differential gene expression and pathway enrichment analyses. Further, we validated our method predictions in the case of rat cirrhotic liver, and identified the activation of angiopoietins receptor Tie2 as a potential target for reverting the disease phenotype. Experimental results indicated that this perturbation induced desired changes in the gene expression of key TFs involved in fibrosis and angiogenesis. Importantly, this method only requires gene expression data of the initial and desired cell states, and therefore is suited for the discovery of signalling interventions for disease treatments and cellular therapies.
Collapse
Affiliation(s)
- Gaia Zaffaroni
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
| | - Satoshi Okawa
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
- Integrated BioBank of Luxembourg, Dudelange L-3555, Luxembourg
| | - Manuel Morales-Ruiz
- Biochemistry and Molecular Genetics Department-Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Working group for the biochemical assessment of hepatic disease-SEQC, Barcelona 08036, Spain
- Department of Biomedicine-Biochemistry Unit, School of Medicine-University of Barcelona, Barcelona 08036, Spain
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
- CIC bioGUNE, Bizkaia Technology Park, Derio 48160, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
- To whom correspondence should be addressed. Tel: +352 46 66 44 6982; Fax: +352 46 66 44 6949;
| |
Collapse
|
10
|
Cheng BF, Lian JJ, Yang HJ, Wang L, Yu HH, Bi JJ, Gao YX, Chen SJ, Wang M, Feng ZW. Neural cell adhesion molecule regulates chondrocyte hypertrophy in chondrogenic differentiation and experimental osteoarthritis. Stem Cells Transl Med 2019; 9:273-283. [PMID: 31742919 PMCID: PMC6988767 DOI: 10.1002/sctm.19-0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022] Open
Abstract
Chondrocyte hypertrophy-like change is an important pathological process of osteoarthritis (OA), but the mechanism remains largely unknown. Neural cell adhesion molecule (NCAM) is highly expressed and involved in the chondrocyte differentiation of mesenchymal stem cells (MSCs). In this study, we found that NCAM deficiency accelerates chondrocyte hypertrophy in articular cartilage and growth plate of OA mice. NCAM deficiency leads to hypertrophic chondrocyte differentiation in both murine MSCs and chondrogenic cells, in which extracellular signal-regulated kinase (ERK) signaling plays an important role. Moreover, NCAM expression is downregulated in an interleukin-1β-stimulated OA cellular model and monosodium iodoacetate-induced OA rats. Overexpression of NCAM substantially inhibits hypertrophic differentiation in the OA cellular model. In conclusion, NCAM could inhibit hypertrophic chondrocyte differentiation of MSCs by inhibiting ERK signaling and reduce chondrocyte hypertrophy in experimental OA model, suggesting the potential utility of NCAM as a novel therapeutic target for alleviating chondrocyte hypertrophy of OA.
Collapse
Affiliation(s)
- Bin-Feng Cheng
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jun-Jiang Lian
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China.,Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Hai-Jie Yang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Lei Wang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Hao-Heng Yu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jia-Jia Bi
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Yao-Xin Gao
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Su-Juan Chen
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Mian Wang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Zhi-Wei Feng
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| |
Collapse
|
11
|
Ma C, Wang K, Ji H, Wang H, Guo L, Wang Z, Ren H, Wang X, Guan W. Multilineage potential research of Beijing duck amniotic mesenchymal stem cells. Cell Tissue Bank 2018; 19:519-529. [PMID: 29858719 PMCID: PMC6280870 DOI: 10.1007/s10561-018-9701-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 05/14/2018] [Indexed: 12/18/2022]
Abstract
Amnion, which is usually discarded as medical waste, is considered as abundant sources for mesenchymal stem cells. In human and veterinary medicine, the multipotency of mesenchymal stem cells derived from amnion (AMSCs) together with their plasticity, self-renewal, low immunogenicity and nontumorigenicity characteristics make AMSCs a promising candidate cell for cell-based therapies and tissue engineering. However, up till now, the multipotential characteristics and therapeutic potential of AMSCs on preclinical studies remain uncertain. In this work, we successfully obtained AMSCs from Beijing duck embryos in vitro, and also attempted to detect their biological characteristics. The isolated AMSCs were phenotypically identified, the growth kinetics and karyotype were tested. Also, the cells were positive for MSCs-related markers (CD29, CD71, CD105, CD166, Vimentin and Fibronection), while the expression of CD34 and CD45 were undetectable. Additionally, AMSCs also expressed the pluripotent marker gene OCT4. Particularly, when appropriately induced, AMSCs could be induced to trans-differentiate into adipocytes, osteoblasts, chondrocytes and neurocytes in vitro. Together, these results demonstrated that the isolated AMSCs maintained their stemness and proliferation in vitro, which may be useful for future cell therapy in regenerative medicine.
Collapse
Affiliation(s)
- Caiyun Ma
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Kunfu Wang
- College of Wildlife Resources, Northeast Forestry University, Harbin, China
| | - Hongda Ji
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hongliang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Liangcai Guo
- Mudanjiang Normal University, Mudanjiang, 157011, China
| | - Zhiyong Wang
- Mudanjiang Normal University, Mudanjiang, 157011, China
| | - Han Ren
- Mudanjiang Normal University, Mudanjiang, 157011, China
| | - Xishuai Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Weijun Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
12
|
Zhang Z, Duan Y, Wu Z, Zhang H, Ren J, Huang L. PPARD is an Inhibitor of Cartilage Growth in External Ears. Int J Biol Sci 2017; 13:669-681. [PMID: 28539839 PMCID: PMC5441183 DOI: 10.7150/ijbs.19714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 03/29/2017] [Indexed: 01/16/2023] Open
Abstract
Peroxisome proliferator-activated receptor beta/delta (PPARD) is an important determinant of multiple biological processes. Our previous studies identified a missense mutation in the PPARD gene that significantly reduces its transcription activity, and consequently causes enlarged external ears in pigs. However, the mechanisms underlying the causality has remained largely unknown. Here, we show that PPARD retards the development of auricular cartilage by accelerating the apoptosis of cartilage stem/progenitor cells (CSPCs), the terminal differentiation of cartilage cells and the degradation of cartilage extracellular matrix in the auricle. At the transcription level, PPARD upregulates a set of genes that are associated with CSPCs apoptosis and chondrogenic differentiation, chondroblast differentiation and extracellular matrix degradation. ChIP-seq identified direct target genes of PPARD, including a well-documented gene for cartilage development: PPARG. We further show that compared to wild-type PPARD, the G32E mutant up-regulates the expression of PPARG and subsequently leads to the downregulation of critical genes that inhibit cartilage growth. These findings allow us to conclude that PPARD is an inhibitor of auricular cartilage growth in pigs. The causative mutation (G32E) in the PPARD gene attenuates the PPARD-mediated retardation of cartilage growth in the auricle, contributing to enlarged ears in pigs. The findings advance our understanding of the mechanisms underlying auricular development in mammals, and shed insight into the studies of innate pinna disorders and cartilage regeneration medicine in humans.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yanyu Duan
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhongping Wu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Hui Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jun Ren
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Lusheng Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| |
Collapse
|
13
|
Xu X, Jiang H, Li X, Wu P, Liu J, Wang T, Zhou X, Xiong J, Li W. Bioinformatics analysis on the differentiation of bone mesenchymal stem cells into osteoblasts and adipocytes. Mol Med Rep 2017; 15:1571-1576. [PMID: 28260060 PMCID: PMC5365017 DOI: 10.3892/mmr.2017.6178] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/15/2016] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to screen several differentially expressed genes (DEGs) and differentially expressed microRNAs (miRNAs) for two types of mesenchymal stem cell (MSC) differentiation. Bone morphogenetic protein 6 (BMP-6) and dexamethasone were used to induce MSCs towards osteoblastic differentiation or adipocytic differentiation. The t-test in the Bioconductor bioinformatics software tool was used to screen DEGs and differentially expressed miRNAs in the two samples. Subsequent gene ontology (GO) and pathway analyses on the DEGs were performed using the GO and Kyoto Encyclopedia of Genes and Genomes databases, respectively; potential target genes for the screened miRNAs were predicted using the TargetScan database. In addition, an interaction network between the DEGs and miRNAs was constructed. Numerous DEGs and miRNAs were screened during osteoblastic and adipocytic differentiation of MSCs. Important pathways, such as glutathione metabolism, pathogenic Escherichia coli infection and Parkinson's disease, and GO terms, including cytoskeletal protein binding and phospholipase inhibitor activity, were enriched in the screened DEGs from MSCs undergoing osteogenic differentiation and adipocytic differentiation. miRNAs, including miRNA (miR)-382 and miR-203, and DEGs, including neuronal growth regulator 1 (NEGR1), phosphatidic acid phosphatase 2B (PPAP2B), platelet-derived growth factor receptor alpha (PDGFRA), interleukin 6 signal transducer (IL6ST) and sortilin 1 (SORT1), were demonstrated to be involved in osteoblastic differentiation. In addition, the downregulated miRNAs (including miR-495, miR-376a and miR-543), the upregulated miR-106a, the upregulated DEGs, including enabled homolog (ENAH), polypeptide N-acetylgalactosaminyltransferase 1 and acyl-CoA synthetase long-chain family member 1, and the downregulated repulsive guidance molecule family member B and semaphorin SEMA7A were demonstrated to be involved in adipocytic differentiation. The results of the present study suggested that miRNAs (miR-203 and miR-382) and DEGs (NEGR1, PPAP2B, PDGFRA, IL6ST and SORT1) may serve pivotal functions in the osteoblastic differentiation of MSCs, whereas miR-495, which is also involved in osteoblast differentiation and had four targets, including NEGR1, miR-376a, miR-543 and ENAH may have crucial roles in adipocytic differentiation of MSCs.
Collapse
Affiliation(s)
- Xiaoyuan Xu
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - He Jiang
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xingnuan Li
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Ping Wu
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianyun Liu
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Tao Wang
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaoou Zhou
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianjun Xiong
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Weidong Li
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
14
|
Study on the effects of gradient mechanical pressures on the proliferation, apoptosis, chondrogenesis and hypertrophy of mandibular condylar chondrocytes in vitro. Arch Oral Biol 2017; 73:186-192. [DOI: 10.1016/j.archoralbio.2016.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 09/03/2016] [Accepted: 10/15/2016] [Indexed: 11/23/2022]
|
15
|
Wu JG, Jia QW, Li Y, Cao FF, Zhang XS, Liu C. Investigating ego modules involved in TGFβ3-induced chondrogenesis in mesenchymal stem cells based on ego network. Comput Biol Chem 2016; 65:16-20. [PMID: 27694041 DOI: 10.1016/j.compbiolchem.2016.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVE This paper aimed to investigate ego modules for TGFβ3-induced chondrogenesis in mesenchymal stem cells (MSCs) using ego network algorithm. METHODS The ego network algorithm comprised three parts, extracting differential expression network (DEN) based on gene expression data and protein-protein interaction (PPI) data; exploring ego genes by reweighting DEN; and searching ego modules by ego gene expansions. Subsequently, permutation test was carried out to evaluate the statistical significance of the ego modules. Finally, pathway enrichment analysis was conducted to investigate ego pathways enriched by the ego modules. RESULTS A total of 15 ego genes were obtained from the DEN, such as PSMA4, HNRNPM and WDR77. Starting with each ego genes, 15 candidate modules were gained. When setting the thresholds of the area under the receiver operating characteristics curve (AUC) ≥0.9 and gene size ≥4, three ego modules (Module 3, Module 8 and Module 14) were identified, and all of them had statistical significances between normal and TGFβ3-induced chondrogenesis in MSCs. By mapping module genes to confirmed pathway database, their ego pathways were detected, Cdc20:Phospho-APC/C mediated degradation of Cyclin A for Module 3, Mitotic G1-G1/S phases for Module 8, and mRNA Splicing for Module 14. CONCLUSIONS We have successfully identified three ego modules, evaluated their statistical significances and investigated their functional enriched ego pathways. The findings might provide potential biomarkers and give great insights to reveal molecular mechanism underlying this process.
Collapse
Affiliation(s)
- Jing-Guo Wu
- Department of Orthopedics, Affiliated Hospital of Taishan Medical University, Tai'an, 271000, Shandong Province, China
| | - Qing-Wei Jia
- Department of Orthopedics, Affiliated Hospital of Taishan Medical University, Tai'an, 271000, Shandong Province, China
| | - Yong Li
- Department of Orthopedics, Tai'an Centre Hospital Branch, Tai'an, 271000, Shandong Province, China
| | - Fei-Fei Cao
- Department of Orthopedics, Tai'an Centre Hospital Branch, Tai'an, 271000, Shandong Province, China
| | - Xi-Shan Zhang
- Department of Orthopedics, Affiliated Hospital of Taishan Medical University, Tai'an, 271000, Shandong Province, China
| | - Cong Liu
- Department of Orthopedics, Tai'an Centre Hospital, Tai'an, 271000, No.29 on Longtan Road, Shandong Province, China.
| |
Collapse
|
16
|
Bone morphogenetic protein 2 stimulates chondrogenesis of equine synovial membrane-derived progenitor cells. Vet Comp Orthop Traumatol 2016; 29:378-85. [PMID: 27468832 DOI: 10.3415/vcot-16-02-0035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/18/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Bone morphogenetic protein 2 (BMP-2) is critical for skeletal and cartilage development, homeostasis and repair. This study was conducted to clone and characterize equine BMP-2, develop expression constructs for equine BMP-2, and to determine whether BMP-2 can stimulate chondrogenesis of equine synovial membrane-derived progenitor cells (SMPC). METHODS Equine BMP-2 cDNA was amplified from chondrocyte RNA, and then transferred into an expression plasmid and adenoviral vector. Effective expression of equine BMP-2 was confirmed using a BMP reporter cell line. SMPC were isolated from synovium, expanded through two passages and transferred to chondrogenic cultures, with recombinant human (rh) transforming growth factor beta 1 (TGF-β1) or rhBMP-2. Chondrogenesis was assessed by up-regulation of collagen types II and X, and aggrecan mRNA, secretion of collagen type II protein and sulfated glycosaminoglycans (sGAG), and by alkaline phosphatase induction. Chondrogenic stimulation of SMPC by the equine BMP-2 adenovirus was assessed by sGAG secretion and histology. RESULTS The mature equine BMP-2 peptide is identical to human and murine peptides. Recombinant human BMP-2 and TGF-β1 stimulated equivalent amounts of collagen type II protein in SMPC pellets, but sGAG secretion was doubled by BMP-2. Neither factor stimulated hypertrophic marker expression. The equine BMP-2 adenoviral vector induced chondrogenesis comparably to rhBMP-2 protein, with no indication of hypertrophy. CLINICAL SIGNIFICANCE Bone morphogenetic protein 2 is a potent inducer of SMPC non-hypertrophic chondrogenesis, supporting the use of this combination for articular cartilage repair applications.
Collapse
|
17
|
Qiao GH, Zhao F, Cheng S, Luo MH. Multipotent mesenchymal stromal cells are fully permissive for human cytomegalovirus infection. Virol Sin 2016; 31:219-28. [PMID: 27105639 DOI: 10.1007/s12250-016-3754-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/05/2016] [Indexed: 01/03/2023] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is a leading infectious cause of birth defects. Previous studies have reported birth defects with multiple organ maldevelopment in congenital HCMV-infected neonates. Multipotent mesenchymal stromal cells (MSCs) are a group of stem/progenitor cells that are multi-potent and can self-renew, and they play a vital role in multi-organ formation. Whether MSCs are susceptible to HCMV infection is unclear. In this study, MSCs were isolated from Wharton's jelly of the human umbilical cord and identified by their plastic adherence, surface marker pattern, and differentiation capacity. Then, the MSCs were infected with the HCMV Towne strain, and infection status was assessed via determination of viral entry, replication initiation, viral protein expression, and infectious virion release using western blotting, immunofluorescence assays, and plaque forming assays. The results indicate that the isolated MSCs were fully permissive for HCMV infection and provide a preliminary basis for understanding the pathogenesis of HCMV infection in non-nervous system diseases, including multi-organ malformation during fetal development.
Collapse
Affiliation(s)
- Guan-Hua Qiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fei Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shuang Cheng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Min-Hua Luo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
18
|
Rohart F, Mason EA, Matigian N, Mosbergen R, Korn O, Chen T, Butcher S, Patel J, Atkinson K, Khosrotehrani K, Fisk NM, Lê Cao KA, Wells CA. A molecular classification of human mesenchymal stromal cells. PeerJ 2016; 4:e1845. [PMID: 27042394 PMCID: PMC4811172 DOI: 10.7717/peerj.1845] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are widely used for the study of mesenchymal tissue repair, and increasingly adopted for cell therapy, despite the lack of consensus on the identity of these cells. In part this is due to the lack of specificity of MSC markers. Distinguishing MSC from other stromal cells such as fibroblasts is particularly difficult using standard analysis of surface proteins, and there is an urgent need for improved classification approaches. Transcriptome profiling is commonly used to describe and compare different cell types; however, efforts to identify specific markers of rare cellular subsets may be confounded by the small sample sizes of most studies. Consequently, it is difficult to derive reproducible, and therefore useful markers. We addressed the question of MSC classification with a large integrative analysis of many public MSC datasets. We derived a sparse classifier (The Rohart MSC test) that accurately distinguished MSC from non-MSC samples with >97% accuracy on an internal training set of 635 samples from 41 studies derived on 10 different microarray platforms. The classifier was validated on an external test set of 1,291 samples from 65 studies derived on 15 different platforms, with >95% accuracy. The genes that contribute to the MSC classifier formed a protein-interaction network that included known MSC markers. Further evidence of the relevance of this new MSC panel came from the high number of Mendelian disorders associated with mutations in more than 65% of the network. These result in mesenchymal defects, particularly impacting on skeletal growth and function. The Rohart MSC test is a simple in silico test that accurately discriminates MSC from fibroblasts, other adult stem/progenitor cell types or differentiated stromal cells. It has been implemented in the www.stemformatics.org resource, to assist researchers wishing to benchmark their own MSC datasets or data from the public domain. The code is available from the CRAN repository and all data used to generate the MSC test is available to download via the Gene Expression Omnibus or the Stemformatics resource.
Collapse
Affiliation(s)
- Florian Rohart
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth A. Mason
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Matigian
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Rowland Mosbergen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Othmar Korn
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Tyrone Chen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Suzanne Butcher
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jatin Patel
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Kerry Atkinson
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women’s Hospital, Brisbane, Queensland, Australia
| | - Nicholas M. Fisk
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women’s Hospital, Brisbane, Queensland, Australia
| | - Kim-Anh Lê Cao
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Christine A. Wells
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Zhong L, Huang X, Karperien M, Post JN. The Regulatory Role of Signaling Crosstalk in Hypertrophy of MSCs and Human Articular Chondrocytes. Int J Mol Sci 2015; 16:19225-47. [PMID: 26287176 PMCID: PMC4581295 DOI: 10.3390/ijms160819225] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/07/2015] [Indexed: 12/26/2022] Open
Abstract
Hypertrophic differentiation of chondrocytes is a main barrier in application of mesenchymal stem cells (MSCs) for cartilage repair. In addition, hypertrophy occurs occasionally in osteoarthritis (OA). Here we provide a comprehensive review on recent literature describing signal pathways in the hypertrophy of MSCs-derived in vitro differentiated chondrocytes and chondrocytes, with an emphasis on the crosstalk between these pathways. Insight into the exact regulation of hypertrophy by the signaling network is necessary for the efficient application of MSCs for articular cartilage repair and for developing novel strategies for curing OA. We focus on articles describing the role of the main signaling pathways in regulating chondrocyte hypertrophy-like changes. Most studies report hypertrophic differentiation in chondrogenesis of MSCs, in both human OA and experimental OA. Chondrocyte hypertrophy is not under the strict control of a single pathway but appears to be regulated by an intricately regulated network of multiple signaling pathways, such as WNT, Bone morphogenetic protein (BMP)/Transforming growth factor-β (TGFβ), Parathyroid hormone-related peptide (PTHrP), Indian hedgehog (IHH), Fibroblast growth factor (FGF), Insulin like growth factor (IGF) and Hypoxia-inducible factor (HIF). This comprehensive review describes how this intricate signaling network influences tissue-engineering applications of MSCs in articular cartilage (AC) repair, and improves understanding of the disease stages and cellular responses within an OA articular joint.
Collapse
Affiliation(s)
- Leilei Zhong
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Xiaobin Huang
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
- School of Life Sciences, Chongqing University, Chongqing 400030, China.
| | - Marcel Karperien
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Janine N Post
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| |
Collapse
|
20
|
Cheng X, Chen JL, Ma ZL, Zhang ZL, Lv S, Mai DM, Liu JJ, Chuai M, Lee KKH, Wan C, Yang X. Biphasic influence of dexamethasone exposure on embryonic vertebrate skeleton development. Toxicol Appl Pharmacol 2014; 281:19-29. [DOI: 10.1016/j.taap.2014.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 08/31/2014] [Accepted: 09/26/2014] [Indexed: 12/26/2022]
|
21
|
Felimban R, Ye K, Traianedes K, Di Bella C, Crook J, Wallace GG, Quigley A, Choong PF, Myers DE. Differentiation of Stem Cells from Human Infrapatellar Fat Pad: Characterization of Cells Undergoing Chondrogenesis. Tissue Eng Part A 2014; 20:2213-23. [DOI: 10.1089/ten.tea.2013.0657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Raed Felimban
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Department of Orthopaedics, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Department of Medical Laboratories, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ken Ye
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Department of Orthopaedics, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Kathy Traianedes
- Departments of Medicine and Clinical Neurosciences, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Claudia Di Bella
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Department of Orthopaedics, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Jeremy Crook
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Intelligent Polymer Research Institute, University of Wollongong, Innovation Campus, Wollongong, New South Wales, Australia
| | - Gordon G. Wallace
- Intelligent Polymer Research Institute, University of Wollongong, Innovation Campus, Wollongong, New South Wales, Australia
| | - Anita Quigley
- Departments of Medicine and Clinical Neurosciences, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Intelligent Polymer Research Institute, University of Wollongong, Innovation Campus, Wollongong, New South Wales, Australia
| | - Peter F.M. Choong
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Department of Orthopaedics, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Damian E. Myers
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
- Department of Orthopaedics, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| |
Collapse
|
22
|
Characterization and evaluation of mesenchymal stem cells derived from human embryonic stem cells and bone marrow. Cell Tissue Res 2014; 358:149-64. [PMID: 24927918 DOI: 10.1007/s00441-014-1926-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 05/15/2014] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells (ESCs) and mesenchymal stem cells (MSCs) have been studied for years as primary cell sources for regenerative biology and medicine. MSCs have been derived from cell and tissue sources, such as bone marrow (BM), and more recently from ESCs. This study investigated MSCs derived from BM, H1- and H9-ESC lines in terms of morphology, surface marker and growth factor receptor expression, proliferative capability, modulation of immune cell growth and multipotency, in order to evaluate ESC-MSCs as a cell source for potential regenerative applications. The results showed that ESC-MSCs exhibited spindle-shaped morphology similar to BM-MSCs but of various sizes, and flow cytometric immunophenotyping revealed expression of characteristic MSC surface markers on all tested cell lines except H9-derived MSCs. Differences in growth factor receptor expression were also shown between cell lines. In addition, ESC-MSCs showed greater capabilities for cell proliferation, and suppression of leukocyte growth compared to BM-MSCs. Using standard protocols, induction of ESC-MSC differentiation along the adipogenic, osteogenic, or chondrogenic lineages was less effective compared to that of BM-MSCs. By adding bone morphogenetic protein 7 (BMP7) into transforming growth factor beta 1 (TGFβ1)-supplemented induction medium, chondrogenesis of ESC-MSCs was significantly enhanced. Our findings suggest that ESC-MSCs and BM-MSCs show differences in their surface marker profiles and the capacities of proliferation, immunomodulation, and most importantly multi-lineage differentiation. Using modified chondrogenic medium with BMP7 and TGFβ1, H1-MSCs can be effectively induced as BM-MSCs for chondrogenesis.
Collapse
|
23
|
Guérit D, Brondello JM, Chuchana P, Philipot D, Toupet K, Bony C, Jorgensen C, Noël D. FOXO3A regulation by miRNA-29a Controls chondrogenic differentiation of mesenchymal stem cells and cartilage formation. Stem Cells Dev 2014; 23:1195-205. [PMID: 24467486 DOI: 10.1089/scd.2013.0463] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Skeletal development and cartilage formation require stringent regulation of gene expression for mesenchymal stem cells (MSCs) to progress through stages of differentiation. Since microRNAs (miRNAs) regulate biological processes, the objective of the present study was to identify novel miRNAs involved in the modulation of chondrogenesis. We performed miRNA profiling and identify miR-29a as being one of the most down-regulated miRNAs during the chondrogenesis. Using chromatin immunoprecipitation, we showed that SOX9 down-regulates its transcription. Moreover, the over-expression of miR-29a strongly inhibited the expression of chondrocyte-specific markers during in vitro chondrogenic differentiation of MSCs. We identified FOXO3A as a direct target of miR-29a and showed a down- and up-regulation of FOXO3a protein levels after transfection of, respectively, premiR- and antagomiR-29a oligonucleotides. Finally, we showed that using the siRNA or premiR approach, chondrogenic differentiation was inhibited to a similar extent. Together, we demonstrate that the down-regulation of miR-29a, concomitantly with FOXO3A up-regulation, is essential for the differentiation of MSCs into chondrocytes and in vivo cartilage/bone formation. The delivery of miRNAs that modulate MSC chondrogenesis may be applicable for cartilage regeneration and deserves further investigation.
Collapse
Affiliation(s)
- David Guérit
- 1 Inserm, U 844, Hôpital Saint-Eloi , Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Djouad F, Tejedor G, Toupet K, Maumus M, Bony C, Blangy A, Chuchana P, Jorgensen C, Noël D. Promyelocytic leukemia zinc-finger induction signs mesenchymal stem cell commitment: identification of a key marker for stemness maintenance? Stem Cell Res Ther 2014; 5:27. [PMID: 24564963 PMCID: PMC4055047 DOI: 10.1186/scrt416] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 02/17/2014] [Indexed: 12/30/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) are an attractive cell source for cartilage and bone tissue engineering given their ability to differentiate into chondrocytes and osteoblasts. However, the common origin of these two specialized cell types raised the question about the identification of regulatory pathways determining the differentiation fate of MSCs into chondrocyte or osteoblast. Methods Chondrogenesis, osteoblastogenesis, and adipogenesis of human and mouse MSC were induced by using specific inductive culture conditions. Expression of promyelocytic leukemia zinc-finger (PLZF) or differentiation markers in MSCs was determined by RT-qPCR. PLZF-expressing MSC were implanted in a mouse osteochondral defect model and the neotissue was analyzed by routine histology and microcomputed tomography. Results We found out that PLZF is not expressed in MSCs and its expression at early stages of MSC differentiation is the mark of their commitment toward the three main lineages. PLZF acts as an upstream regulator of both Sox9 and Runx2, and its overexpression in MSC enhances chondrogenesis and osteogenesis while it inhibits adipogenesis. In vivo, implantation of PLZF-expressing MSC in mice with full-thickness osteochondral defects resulted in the formation of a reparative tissue resembling cartilage and bone. Conclusions Our findings demonstrate that absence of PLZF is required for stemness maintenance and its expression is an early event at the onset of MSC commitment during the differentiation processes of the three main lineages.
Collapse
|
25
|
Mathieu M, Iampietro M, Chuchana P, Guérit D, Djouad F, Noël D, Jorgensen C. Involvement of angiopoietin-like 4 in matrix remodeling during chondrogenic differentiation of mesenchymal stem cells. J Biol Chem 2014; 289:8402-12. [PMID: 24505142 DOI: 10.1074/jbc.m113.539825] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered for cartilage engineering given their ability to differentiate into chondrocytes. Chondrogenic differentiation of MSCs is currently triggered by micromass culture in the presence of a member of the TGF-β superfamily. However, the main constituents of the cartilaginous matrix, aggrecan and type II collagen, are degraded at the end of the differentiation process through induction of matrix metallopeptidase (MMP)13. We hypothesized that MSCs undergoing chondrogenic differentiation produce an intermediate cytokine that triggers this matrix remodeling. Analysis of transcriptomic data identified angiopoietin-like 4 (ANGPTL4) as one of the most strongly up-regulated gene encoding a secreted factor during TGF-β-induced chondrogenesis. To gain insight into the role of ANGPTL4 during chondrogenesis, we used recombinant ANGPTL4 as well as a RNA interference approach. Addition of exogenous ANGPTL4 during the course of TGF-β-induced differentiation reduced the mRNA levels of aggrecan and type II collagen, although it increased those of MMP1 and MMP13. Accordingly, deposition of aggrecan and total collagens was diminished, whereas release of MMP1 and MMP13 was increased. Conversely, transfection of MSCs with an siRNA targeting ANGPTL4 prior to induction of chondrogenesis increased expression of type II collagen and aggrecan, whereas it repressed that of MMP1, MMP3, and MMP13. A neutralizing antibody against integrin αVβ5, a known receptor for ANGPTL4, mimicked some of the effects observed after siRNA-mediated ANGPTL4 silencing. Our data provide evidence that ANGPTL4 promotes cartilage matrix remodeling by inhibiting expression of its two key components and by up-regulating the level of certain MMPs.
Collapse
Affiliation(s)
- Marc Mathieu
- From Inserm, U844, Hôpital Saint-Eloi, Montpellier F-34091, France
| | | | | | | | | | | | | |
Collapse
|
26
|
Boyd AL, Salci KR, Shapovalova Z, McIntyre BAS, Bhatia M. Nonhematopoietic cells represent a more rational target of in vivo hedgehog signaling affecting normal or acute myeloid leukemia progenitors. Exp Hematol 2013; 41:858-869.e4. [PMID: 23747997 DOI: 10.1016/j.exphem.2013.05.287] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 11/30/2022]
Abstract
Recent work has shown that leukemic stem cell self-renewal in chronic myeloid leukemia is dependent on cell-intrinsic hedgehog (Hh) signaling, and early clinical trials suggest that targeting this pathway is also therapeutic in patients with acute myeloid leukemia (AML). In this study, we aimed to better understand Hh signaling in normal hematopoiesis and AML by molecularly and functionally analyzing more than 200 primary human AML patient samples compared with nonleukemic controls. Gene expression analysis indicated that Hh pathway transcripts were similarly regulated in AML and nonleukemic controls, regardless of whether samples were purified based on primitive phenotypes. Consistent with these results, pharmacologic inhibition of Smoothened (SMO) did not preferentially reduce in vitro colony formation of AML versus normal progenitors. Using a unique analytic approach, messenger RNA expression of membrane receptor SMO was found to be unexpectedly rare within all hematopoietic samples analyzed, which is indicative of heterogeneity at the level of Hh signaling machinery. In contrast, abundant SMO expression could be readily detected in the nonhematopoietic fraction of human and murine bone marrow (BM) cells. Our predictions of increased SMO(+) cell frequencies within nonhematopoietic BM fractions were further supported by single-cell protein analyses. Although we did not find support for cell-autonomous sensitivity of AML cells to Hh pathway inhibition, we alternatively suggest that nonhematopoietic BM cells represent an indirect target through which primitive normal and leukemic cells can be modulated. These findings suggest current approaches to applying Hh inhibition should be carefully reevaluated to account for BM niche cell regulation that might be selectively Hh responsive.
Collapse
Affiliation(s)
- Allison L Boyd
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | | | | | | | | |
Collapse
|
27
|
Li F, Niyibizi C. Cells derived from murine induced pluripotent stem cells (iPSC) by treatment with members of TGF-beta family give rise to osteoblasts differentiation and form bone in vivo. BMC Cell Biol 2012; 13:35. [PMID: 23241430 PMCID: PMC3541062 DOI: 10.1186/1471-2121-13-35] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 12/06/2012] [Indexed: 01/17/2023] Open
Abstract
Background Induced pluripotent stem cells (iPSC) are generated by reprogramming somatic cells into embryonic like state (ESC) using defined factors. There is great interest in these cells because of their potential for application in regenerative medicine. Results iPSC reprogrammed from murine tail tip fibroblasts were exposed to retinoic acid alone (RA) or in combination with TGF-β1 and 3, basic fibroblast growth factor (bFGF) or bone morphogenetic protein 2 (BMP-2). The resulting cells expressed selected putative mesenchymal stem cells (MSCs) markers; differentiated toward osteoblasts and adipocytic cell lineages in vitro at varying degrees. TGF-beta1 and 3 derived-cells possessed higher potential to give rise to osteoblasts than bFGF or BMP-2 derived-cells while BMP-2 derived cells exhibited a higher potential to differentiate toward adipocytic lineage. TGF-β1 in combination with RA derived-cells seeded onto HA/TCP ceramics and implanted in mice deposited typical bone. Immunofluorescence staining for bone specific proteins in cell seeded scaffolds tissue sections confirmed differentiation of the cells into osteoblasts in vivo. Conclusions The results demonstrate that TGF-beta family of proteins could potentially be used to generate murine iPSC derived-cells with potential for osteoblasts differentiation and bone formation in vivo and thus for application in musculoskeletal tissue repair and regeneration.
Collapse
Affiliation(s)
- Feng Li
- Department of Orthopaedics and Rehabilitation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | |
Collapse
|
28
|
Ullah M, Hamouda H, Stich S, Sittinger M, Ringe J. A reliable protocol for the isolation of viable, chondrogenically differentiated human mesenchymal stem cells from high-density pellet cultures. Biores Open Access 2012; 1:297-305. [PMID: 23514965 PMCID: PMC3559221 DOI: 10.1089/biores.2012.0279] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Administration of chondrogenically differentiated mesenchymal stem cells (MSC) is discussed as a promising approach for the regenerative treatment of injured or diseased cartilage. The high-density pellet culture is the standard culture for chondrogenic differentiation, but cells in pellets secrete extracellular matrix (ECM) that they become entrapped in. Protocols for cell isolation from pellets often result in cell damage and dedifferentiation towards less differentiated MSC. Therefore, our aim was to develop a reliable protocol for the isolation of viable, chondrogenically differentiated MSC from high-density pellet cultures. Human bone marrow MSC were chondrogenically stimulated with transforming growth factor-β3, and the cartilaginous structure of the pellets was verified by alcian blue staining of cartilage proteoglycans, antibody staining of cartilage collagen type II, and quantitative real-time reverse-transcription polymerase chain reaction of the marker genes COL2A1 and SOX9. Trypsin and collagenases II and P were tested alone or in combination, and for different concentrations and times, to find a protocol for optimized pellet digestion. Whereas trypsin was not able to release viable cells, 90-min digestion with 300 U of collagenase II, 20 U of collagenase P, and 2 mM CaCl2 worked quite well and resulted in about 2.5×10(5) cells/pellet. The protocol was further optimized for the separation of released cells and ECM from each other. Cells were alcian blue and collagen type II positive and expressed COL2A1 and SOX9, verifying a chondrogenic character. However, they had different morphological shapes. The ECM was also uniformly alcian blue and collagen type II positive but showed different organizational and structural forms. To conclude, our protocol allows the reliable isolation of a defined number of viable, chondrogenically differentiated MSC from high-density pellet cultures. Such cells, as well as the ECM components, are of interest as research tools and for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mujib Ullah
- Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Houda Hamouda
- AG Glycodesign and Glycoanalytics, Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry; Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Stich
- Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sittinger
- Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen Ringe
- Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
29
|
Bhang SH, Jeon JY, La WG, Seong JY, Hwang JW, Ryu SE, Kim BS. Enhanced chondrogenic marker expression of human mesenchymal stem cells by interaction with both TGF-β3 and hyaluronic acid. Biotechnol Appl Biochem 2011; 58:271-6. [PMID: 21838802 DOI: 10.1002/bab.39] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study was designed to evaluate the additive effects of transforming growth factor-beta3 (TGF-β3) and hyaluronic acid (HA) on chondrogenic differentiation of human mesenchymal stem cells (hMSCs). The hMSCs were cultured on collagen type I-, HA-, or fibronectin-coated cell culture dishes with or without TGF-β3 added to the culture medium. Four weeks after cell culture, chondrogenic differentiation of hMSCs was determined by evaluating the expression of cartilage-specific markers using real-time polymerase chain reaction, immunocytochemistry, and Western blot analysis. hMSCs cultured on HA-coated dishes with TGF-β3 supplementation revealed a prominent increase in collagen type II, aggrecan, and Sox9. When hMSCs were cultured without TGF-β3 supplementation, only hMSCs cultured on HA-coated dishes showed prominent expression of the cartilage-specific markers. This study shows that chondrogenic differentiation of hMSCs can be enhanced additively by interactions with both a specific cell-adhesion matrix and a soluble growth factor.
Collapse
Affiliation(s)
- Suk Ho Bhang
- School of Chemical and Biological Engineering, Seoul National University, Gwanak-gu, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Multipotent mesenchymal stromal cells or mesenchymal stem cells (MSCs) are mainly isolated from bone marrow or fat tissue. Owing to their potential for multilineage differentiation towards bone, cartilage and fat tissue, they were initially evaluated in innovative strategies for tissue engineering. More recently, they have gained interest for their immunomodulatory properties and have been tested in various clinical trials that aim to modulate the host immune response in graft-versus-host disease or autoimmune diseases. MSC-mediated immunomodulation occurs through the secretion of soluble mediators. The clinical applications of MSCs for rheumatic diseases focus on their potential to promote tissue repair/regeneration and prevent inflammation. This article will focus on the mechanisms by which MSCs might exhibit a therapeutic potential in rheumatology. Special attention is given to their potential for innovative future strategies.
Collapse
Affiliation(s)
| | - Danièle Noël
- INSERM U 844, Hôpital Saint-Eloi, 34295 Montpellier, France
| |
Collapse
|
31
|
Human bone marrow mesenchymal stem cells: a systematic reappraisal via the genostem experience. Stem Cell Rev Rep 2011; 7:32-42. [PMID: 20198518 DOI: 10.1007/s12015-010-9125-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Genostem (acronym for "Adult mesenchymal stem cells engineering for connective tissue disorders. From the bench to the bed side") has been an European consortium of 30 teams working together on human bone marrow Mesenchymal Stem Cell (MSC) biological properties and repair capacity. Part of Genostem activity has been dedicated to the study of basic issues on undifferentiated MSCs properties and on signalling pathways leading to the differentiation into 3 of the connective tissue lineages, osteoblastic, chondrocytic and tenocytic. We have evidenced that native bone marrow MSCs and stromal cells, forming the niche of hematopoietic stem cells, were the same cellular entity located abluminally from marrow sinus endothelial cells. We have also shown that culture-amplified, clonogenic and highly-proliferative MSCs were bona fide stem cells, sharing with other stem cell types the major attributes of self-renewal and of multipotential priming to the lineages to which they can differentiate (osteoblasts, chondrocytes, adipocytes and vascular smooth muscle cells/pericytes). Extensive transcription profiling and in vitro and in vivo assays were applied to identify genes involved in differentiation. Thus we have described novel factors implicated in osteogenesis (FHL2, ITGA5, Fgf18), chondrogenesis (FOXO1A) and tenogenesis (Smad8). Another part of Genostem activity has been devoted to studies of the repair capacity of MSCs in animal models, a prerequisite for future clinical trials. We have developed novel scaffolds (chitosan, pharmacologically active microcarriers) useful for the repair of both bone and cartilage. Finally and most importantly, we have shown that locally implanted MSCs effectively repair bone, cartilage and tendon.
Collapse
|
32
|
Díaz-Prado S, Muiños-López E, Hermida-Gómez T, Cicione C, Rendal-Vázquez ME, Fuentes-Boquete I, de Toro FJ, Blanco FJ. Human amniotic membrane as an alternative source of stem cells for regenerative medicine. Differentiation 2011; 81:162-71. [PMID: 21339039 DOI: 10.1016/j.diff.2011.01.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/28/2010] [Accepted: 01/11/2011] [Indexed: 12/18/2022]
Abstract
The human amniotic membrane (HAM) is a highly abundant and readily available tissue. This amniotic tissue has considerable advantageous characteristics to be considered as an attractive material in the field of regenerative medicine. It has low immunogenicity, anti-inflammatory properties and their cells can be isolated without the sacrifice of human embryos. Since it is discarded post-partum it may be useful for regenerative medicine and cell therapy. Amniotic membranes have already been used extensively as biologic dressings in ophthalmic, abdominal and plastic surgery. HAM contains two cell types, from different embryological origins, which display some characteristic properties of stem cells. Human amnion epithelial cells (hAECs) are derived from the embryonic ectoderm, while human amnion mesenchymal stromal cells (hAMSCs) are derived from the embryonic mesoderm. Both populations have similar immunophenotype and multipotential for in vitro differentiation into the major mesodermal lineages, however they differ in cell yield. Therefore, HAM has been proposed as a good candidate to be used in cell therapy or regenerative medicine to treat damaged or diseased tissues.
Collapse
Affiliation(s)
- Silvia Díaz-Prado
- Department of Medicine, INIBIC-University of A Coruña, Spain; CIBER-BBN-Cellular Therapy Area, Spain
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Díaz-Prado S, Muiños-López E, Hermida-Gómez T, Rendal-Vázquez ME, Fuentes-Boquete I, de Toro FJ, Blanco FJ. Isolation and characterization of mesenchymal stem cells from human amniotic membrane. Tissue Eng Part C Methods 2010; 17:49-59. [PMID: 20673138 DOI: 10.1089/ten.tec.2010.0136] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION The human amniotic membrane is a highly abundant and readily available tissue that may be useful for regenerative medicine and cell therapy. AIM To compare two previously published protocols for the isolation of human amnion mesenchymal stromal cells (hAMSCs), including their phenotypic characterization and in vitro potential for differentiation toward osteogenic, adipogenic, and chondrogenic mesodermal lineages. MATERIALS AND METHODS Human placentas were obtained from selected caesarean-sectioned births. Two different protocols (Alviano et al. (1) and Soncini et al. (2) ) for the isolation of hAMSCs were performed. After monolayer expansion of adherent cells from both protocols, the cells were characterized by flow cytometry and for multipotentiality, as assessed by their capability to differentiate toward adipocyte-, osteoblast-, and chondrocyte-like cells. RESULTS Both protocols yielded hAMSCs that showed plastic adherence, fibroblast-like growth, and well-defined human MSC markers. The cell yield and mesodermal differentiation capability of hAMSCs were higher in cells isolated using the Soncini protocol. CONCLUSIONS Our data demonstrated the successful isolation of hAMSCs from full-term placentas using two published protocols. Differences between the two protocols in cell yield and in vitro differentiation potential are shown.
Collapse
Affiliation(s)
- Silvia Díaz-Prado
- 1 Department of Medicine, INIBIC-University of A Coruña , A Coruña, Spain
| | | | | | | | | | | | | |
Collapse
|
34
|
Baker BM, Nathan AS, Gee AO, Mauck RL. The influence of an aligned nanofibrous topography on human mesenchymal stem cell fibrochondrogenesis. Biomaterials 2010; 31:6190-200. [PMID: 20494438 DOI: 10.1016/j.biomaterials.2010.04.036] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 04/21/2010] [Indexed: 01/13/2023]
Abstract
Fibrocartilaginous tissues serve critical load-bearing functions in numerous joints throughout the body. As these structures are often injured, there exists great demand for engineered tissue for repair or replacement. This study assessed the ability of human marrow-derived mesenchymal stem cells (MSCs) to elaborate a mechanically functional, fibrocartilaginous matrix in a nanofibrous microenvironment. Nanofibrous scaffolds, composed of ultra-fine biodegradable polymer fibers, replicate the structural and mechanical anisotropy of native fibrous tissues and serve as a 3D micro-pattern for directing cell orientation and ordered matrix formation. MSCs were isolated from four osteoarthritic (OA) patients, along with meniscal fibrochondrocytes (FC) which have proven to be a potent cell source for engineering fibrocartilage. Cell-seeded nanofibrous scaffolds were cultured in a chemically-defined medium formulation and mechanical, biochemical, and histological features were evaluated over 9 weeks. Surprisingly, and contrary to previous studies with juvenile bovine cells, matrix assembly by adult human MSCs was dramatically hindered compared to donor-matched FCs cultured similarly. Unlike FCs, MSCs did not proliferate, resulting in sparsely colonized constructs. Increases in matrix content, and therefore changes in tensile properties, were modest in MSC-seeded constructs compared to FC counterparts, even when normalized to the lower cell number in these constructs. To rule out the influence of OA sourcing on MSC functional potential, constructs from healthy young donors were generated; these constructs matured no differently than those formed with OA MSCs. Importantly, there was no difference in matrix production of MSCs and FCs when cultured in pellet form, highlighting the sensitivity of human MSCs to their 3D microenvironment.
Collapse
Affiliation(s)
- Brendon M Baker
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
35
|
Franco HL, Lee KY, Rubel CA, Creighton CJ, White LD, Broaddus RR, Lewis MT, Lydon JP, Jeong JW, DeMayo FJ. Constitutive activation of smoothened leads to female infertility and altered uterine differentiation in the mouse. Biol Reprod 2010; 82:991-9. [PMID: 20130264 PMCID: PMC2857637 DOI: 10.1095/biolreprod.109.081513] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Previous work has identified Indian hedgehog (Ihh) as a major mediator of progesterone signaling during embryo implantation. Ihh acts through its downstream effector smoothened (Smo) to activate the GLI family of transcription factors. In order to gain a better understanding of Ihh action during embryo implantation, we expressed a Cre-recombinase-dependent constitutively activated SMO in the murine uterus using the Pgr(tm2(cre)Lyd) (PR(cre)) mouse model [Pgr(tm2(cre)Lyd+)Gt(ROSA)26Sor(tm1(Smo/EYFP)Amc)(+) (PR(cre/+)SmoM2(+))]. Female PR(cre/+)SmoM2(+) mice were infertile. They exhibited normal serum progesterone levels and normal ovulation, but their ova failed to be fertilized in vivo and their uterus failed to undergo the artificially induced decidual response. Examination of the PR(cre/+)SmoM2(+) uteri revealed numerous features such as uterine hypertrophy, the presence of a stratified luminal epithelial cell layer, a reduced number of uterine glands, and an endometrial stroma that had lost its normal morphologic characteristics. Microarray analysis of 3-mo-old PR(cre/+)SmoM2(+) uteri demonstrated a chondrocytic signature and confirmed that constitutive activation of PR(cre/+)SmoM2(+) increased extracellular matrix production. Thus, constitutive activation of Smo in the mouse uterus alters postnatal uterine differentiation which interferes with early pregnancy. These results provide new insight into the role of Hedgehog signaling during embryo implantation.
Collapse
Affiliation(s)
- Heather L. Franco
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Kevin Y. Lee
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Cory A. Rubel
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Chad J. Creighton
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Lisa D. White
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Russell R. Broaddus
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Michael T. Lewis
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jae-Wook Jeong
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Francesco J. DeMayo
- Department of Molecular and Cellular Biology, Department of Molecular and Human Genetics, The Dan L. Duncan Cancer Center, and The Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas,Correspondence: Francesco J. DeMayo, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030. FAX: 713 790 1275; e-mail:
| |
Collapse
|
36
|
Daher RJ, Chahine NO, Greenberg AS, Sgaglione NA, Grande DA. New methods to diagnose and treat cartilage degeneration. Nat Rev Rheumatol 2009; 5:599-607. [PMID: 19786989 DOI: 10.1038/nrrheum.2009.204] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lesions in articular cartilage can result in significant musculoskeletal morbidity and display unique biomechanical characteristics that make repair difficult, at best. Several surgical procedures have been devised in an attempt to relieve pain, restore function, and delay or stop the progression of cartilaginous lesions. Advanced MRI and ultrasonography protocols are currently used in the evaluation of tissue repair and to improve diagnostic capability. Other nonoperative modalities, such as injection of intra-articular hyaluronic acid or supplementary oral glucosamine and chondroitin sulfate, have shown potential efficacy as anti-inflammatory and symptom-modifying agents. The emerging field of tissue engineering, involving the use of a biocompatible, structurally and mechanically stable scaffold, has shown promising early results in cartilage tissue repair. Scaffolds incorporating specific cell sources and bioactive molecules have been the focus in this new exciting field. Further work is required to better understand the behavior of chondrocytes and the variables that influence their ability to heal articular lesions. The future of cartilage repair will probably involve a combination of treatments in an attempt to achieve a regenerative tissue that is both biomechanically stable and, ideally, identical to the surrounding native tissues.
Collapse
Affiliation(s)
- Robert J Daher
- Department of Orthopedics, Long Island Jewish Medical Center, New Hyde Park, NY, USA
| | | | | | | | | |
Collapse
|