1
|
Das S, Valoor R, Ratnayake P, Basu B. Low-Concentration Gelatin Methacryloyl Hydrogel with Tunable 3D Extrusion Printability and Cytocompatibility: Exploring Quantitative Process Science and Biophysical Properties. ACS APPLIED BIO MATERIALS 2024; 7:2809-2835. [PMID: 38602318 DOI: 10.1021/acsabm.3c01194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Three-dimensional (3D) bioprinting of hydrogels with a wide spectrum of compositions has been widely investigated. Despite such efforts, a comprehensive understanding of the correlation among the process science, buildability, and biophysical properties of the hydrogels for a targeted clinical application has not been developed in the scientific community. In particular, the quantitative analysis across the entire developmental path for 3D extrusion bioprinting of such scaffolds is not widely reported. In the present work, we addressed this gap by using widely investigated biomaterials, such as gelatin methacryloyl (GelMA), as a model system. Using extensive experiments and quantitative analysis, we analyzed how the individual components of methacrylated carboxymethyl cellulose (mCMC), needle-shaped nanohydroxyapatite (nHAp), and poly(ethylene glycol)diacrylate (PEGDA) with GelMA as baseline matrix of the multifunctional bioink can influence the biophysical properties, printability, and cellular functionality. The complex interplay among the biomaterial ink formulations, viscoelastic properties, and printability toward the large structure buildability (structurally stable cube scaffolds with 15 mm edge) has been explored. Intriguingly, the incorporation of PEGDA into the GelMA/mCMC matrix offered improved compressive modulus (∼40-fold), reduced swelling ratio (∼2-fold), and degradation rates (∼30-fold) compared to pristine GelMA. The correlation among microstructural pore architecture, biophysical properties, and cytocompatibility is also established for the biomaterial inks. These photopolymerizable bio(material)inks served as the platform for the growth and development of bone and cartilage matrix when human mesenchymal stem cells (hMSCs) are either seeded on two-dimensional (2D) substrates or encapsulated on 3D scaffolds. Taken together, this present study unequivocally establishes a significant step forward in the development of a broad spectrum of shape-fidelity compliant bioink for the 3D bioprinting of multifunctional scaffolds and emphasizes the need for invoking more quantitative analysis in establishing process-microstructure-property correlation.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Remya Valoor
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Praneeth Ratnayake
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
2
|
Liu J, He S, Ma B, Li X, Wang Y, Xiong J. TMT-based quantitative proteomic analysis revealed that FBLN2 and NPR3 are involved in the early osteogenic differentiation of mesenchymal stem cells (MSCs). Aging (Albany NY) 2023; 15:7637-7654. [PMID: 37543430 PMCID: PMC10457061 DOI: 10.18632/aging.204931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/18/2023] [Indexed: 08/07/2023]
Abstract
The delicate equilibrium between osteoblast and adipocyte differentiation of MSCs is highly regulated. We screened for early-stage osteogenesis- or adipogenesis-based MSCs protein expression profiles using TMT-based quantitative proteomic analysis to identify novel participating molecules. Protein annotation, hierarchical clustering, functional stratification, and protein-protein association assessments were performed. Moreover, two upregulated proteins, namely, FBLN2 and NPR3, were validated to participate in the osteogenic differentiation process of MSCs. After that, we independently downregulated FBLN2 and NPR3 over seven days of osteogenic differentiation, and we performed quantitative proteomics analysis to determine how different proteins were regulated in knockdown vs. control cells. Based on gene ontology (GO) and network analyses, FBLN2 deficiency induced functional alterations associated with biological regulation and stimulus-response, whereas NPR3 deficiency induced functional alterations related to cellular and metabolic processes, and so on. These findings suggested that proteomics remains a useful method for an in-depth study of the MSCs differentiation process. This will assist in comprehensively evaluating its role in osteoporosis and provide additional approaches for identifying as-yet-unidentified effector molecules.
Collapse
Affiliation(s)
- Jianyun Liu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang 332005, China
| | - Shan He
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang 332005, China
| | - Baicheng Ma
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang 332005, China
| | - Xingnuan Li
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang 332005, China
| | - Yaqin Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jianjun Xiong
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang 332005, China
| |
Collapse
|
3
|
Migdadi L, Sharar N, Jafar H, Telfah A, Hergenröder R, Wöhler C. Machine Learning in Automated Monitoring of Metabolic Changes Accompanying the Differentiation of Adipose-Tissue-Derived Human Mesenchymal Stem Cells Employing 1H- 1H TOCSY NMR. Metabolites 2023; 13:352. [PMID: 36984792 PMCID: PMC10055867 DOI: 10.3390/metabo13030352] [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: 01/20/2023] [Revised: 02/12/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
The ability to monitor the dynamics of stem cell differentiation is a major goal for understanding biochemical evolution pathways. Automating the process of metabolic profiling using 2D NMR helps us to understand the various differentiation behaviors of stem cells, and therefore sheds light on the cellular pathways of development, and enhances our understanding of best practices for in vitro differentiation to guide cellular therapies. In this work, the dynamic evolution of adipose-tissue-derived human Mesenchymal stem cells (AT-derived hMSCs) after fourteen days of cultivation, adipocyte and osteocyte differentiation, was inspected based on 1H-1H TOCSY using machine learning. Multi-class classification in addition to the novelty detection of metabolites was established based on a control hMSC sample after four days' cultivation and we successively detected the changes of metabolites in differentiated MSCs following a set of 1H-1H TOCSY experiments. The classifiers Kernel Null Foley-Sammon Transform and Kernel Density Estimation achieved a total classification error between 0% and 3.6% and false positive and false negative rates of 0%. This approach was successfully able to automatically reveal metabolic changes that accompanied MSC cellular evolution starting from their undifferentiated status to their prolonged cultivation and differentiation into adipocytes and osteocytes using machine learning supporting the research in the field of metabolic pathways of stem cell differentiation.
Collapse
Affiliation(s)
- Lubaba Migdadi
- Image Analysis Group, TU Dortmund, 44227 Dortmund, Germany
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
| | - Nour Sharar
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
- Cell Therapy Center, University of Jordan, Amman 11942, Jordan
| | - Hanan Jafar
- Cell Therapy Center, University of Jordan, Amman 11942, Jordan
- Department of Anatomy and Histology, College of Medicine, University of Jordan, Amman 11942, Jordan
| | - Ahmad Telfah
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
- Nanotechnology Center, The University of Jordan, Amman 11942, Jordan
| | - Roland Hergenröder
- Leibniz-Institut für Analytische Wissenschaften—ISAS-e.V., 44139 Dortmund, Germany
| | | |
Collapse
|
4
|
Shen L, Yu Y, Zhou Y, Pruett-Miller SM, Zhang GF, Karner CM. SLC38A2 provides proline to fulfil unique synthetic demands arising during osteoblast differentiation and bone formation. eLife 2022; 11:76963. [PMID: 35261338 PMCID: PMC9007586 DOI: 10.7554/elife.76963] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular differentiation is associated with the acquisition of a unique protein signature which is essential to attain the ultimate cellular function and activity of the differentiated cell. This is predicted to result in unique biosynthetic demands that arise during differentiation. Using a bioinformatic approach, we discovered osteoblast differentiation is associated with increased demand for the amino acid proline. When compared to other differentiated cells, osteoblast-associated proteins including RUNX2, OSX, OCN and COL1A1 are significantly enriched in proline. Using a genetic and metabolomic approach, we demonstrate that the neutral amino acid transporter SLC38A2 acts cell autonomously to provide proline to facilitate the efficient synthesis of proline-rich osteoblast proteins. Genetic ablation of SLC38A2 in osteoblasts limits both osteoblast differentiation and bone formation in mice. Mechanistically, proline is primarily incorporated into nascent protein with little metabolism observed. Collectively, these data highlight a requirement for proline in fulfilling the unique biosynthetic requirements that arise during osteoblast differentiation and bone formation.
Collapse
Affiliation(s)
- Leyao Shen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yilin Yu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yunji Zhou
- Department of Biostatistics and Bioinformatics, Duke University, Durham, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, United States
| | - Guo-Fang Zhang
- Sarah W Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, United States
| | - Courtney M Karner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
5
|
Wang F, Chen X, Li J, Wang D, Huang H, Li X, Bi Z, Peng Y, Zhang X, Li G, Wang J, Wang C, Fu Q, Liu L. Dose- and Time-Dependent Effects of Human Mesenchymal Stromal Cell Infusion on Cardiac Allograft Rejection in Mice. Stem Cells Dev 2021; 30:203-213. [PMID: 33371825 DOI: 10.1089/scd.2019.0300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Heart transplantation is the final life-saving therapeutic strategy for many end-stage heart diseases. Long-term immunosuppressive regimens are needed to prevent allograft rejection. Mesenchymal stromal cells (MSCs) have been shown as immunomodulatory therapy for organ transplantation. However, the effect of dose and timing of MSC treatment on heart transplantation has not yet been examined. In this study, we infused three doses (1 × 106, 2 × 106, or 5 × 106 cells) of human MSCs (hMSCs) to the recipient BALB/c mice before (7 days or 24 h) or after (24 h) receiving C57BL/6 cardiac transplants. We found that infusion of high dose hMSCs (5 × 106) at 24 h post-transplantation significantly prolonged the survival time of cardiac grafts. To delineate the underlying mechanism, grafts, spleens, and draining lymph nodes were harvested for analysis. Dose-dependent effect of hMSC treatment was shown in: (1) alleviation of International Society of Heart and Lung Transplantation (ISHLT) score in grafts; (2) reduction of the population of CD4+ and CD8+ T cells; (3) increase of regulatory T (Treg) cells; (4) and decrease of serum levels of inflammatory cytokines and donor-specific antibodies. Taken together, we showed timing critical and dose-dependent immunomodulatory effects of hMSC treatment against acute allograft rejection in a mouse model of heart transplantation.
Collapse
Affiliation(s)
- Feng Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Organ Transplant Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Jun Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, SunYat-sen University, Guangzhou, China
| | - Huiting Huang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xirui Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zirong Bi
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanwen Peng
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Gang Li
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Jiali Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Changxi Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory on Organ Donation and Transplant Immunology, Guangzhou, China
| | - Qian Fu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Longshan Liu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory on Organ Donation and Transplant Immunology, Guangzhou, China
| |
Collapse
|
6
|
Naot D, Bentley J, Macpherson C, Pitto RP, Bava U, Choi AJ, Matthews BG, Callon KE, Gao R, Horne A, Gamble GD, Reid IR, Cornish J. Molecular characterisation of osteoblasts from bone obtained from people of Polynesian and European ancestry undergoing joint replacement surgery. Sci Rep 2021; 11:2428. [PMID: 33510208 PMCID: PMC7844412 DOI: 10.1038/s41598-021-81731-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022] Open
Abstract
Population studies in Aotearoa New Zealand found higher bone mineral density and lower rate of hip fracture in people of Polynesian ancestry compared to Europeans. We hypothesised that differences in osteoblast proliferation and differentiation contribute to the differences in bone properties between the two groups. Osteoblasts were cultured from bone samples obtained from 30 people of Polynesian ancestry and 25 Europeans who had joint replacement surgeries for osteoarthritis. The fraction of cells in S-phase was determined by flow cytometry, and gene expression was analysed by microarray and real-time PCR. We found no differences in the fraction of osteoblasts in S-phase between the groups. Global gene expression analysis identified 79 differentially expressed genes (fold change > 2, FDR P < 0.1). Analysis of selected genes by real-time PCR found higher expression of COL1A1 and KRT34 in Polynesians, whereas BGLAP, DKK1, NOV, CDH13, EFHD1 and EFNB2 were higher in Europeans (P ≤ 0.01). Osteoblasts from European donors had higher levels of late differentiation markers and genes encoding proteins that inhibit the Wnt signalling pathway. This variability may contribute to the differences in bone properties between people of Polynesian and European ancestry that had been determined in previous studies.
Collapse
Affiliation(s)
- Dorit Naot
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| | - Jarome Bentley
- Middlemore Hospital, Counties Manukau District Health Board, Auckland, 1062, New Zealand
| | | | - Rocco P Pitto
- Middlemore Hospital, Counties Manukau District Health Board, Auckland, 1062, New Zealand
- Department of Orthopaedic Surgery, South Auckland Clinical Campus, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Usha Bava
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Ally J Choi
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Karen E Callon
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Ryan Gao
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Anne Horne
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Gregory D Gamble
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Ian R Reid
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Jillian Cornish
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| |
Collapse
|
7
|
Systems biology analysis of osteogenic differentiation behavior by canine mesenchymal stem cells derived from bone marrow and dental pulp. Sci Rep 2020; 10:20703. [PMID: 33244029 PMCID: PMC7692528 DOI: 10.1038/s41598-020-77656-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Utilization of canine mesenchymal stem cells (cMSCs) for regenerating incorrigible bone diseases has been introduced. However, cMSCs harvested from different sources showed distinct osteogenicity. To clarify this, comparative proteomics-based systems biology analysis was used to analyze osteogenic differentiation behavior by cMSCs harvested from bone marrow and dental pulp. The results illustrated that canine dental pulp stem cells (cDPSCs) contained superior osteogenicity comparing with canine bone marrow-derived MSCs (cBM-MSCs) regarding alkaline phosphatase activity, matrix mineralization, and osteogenic marker expression. Global analyses by proteomics platform showed distinct protein clustering and expression pattern upon an in vitro osteogenic induction between them. Database annotation using Reactome and DAVID revealed contrast and unique expression profile of osteogenesis-related proteins, particularly on signaling pathways, cellular components and processes, and cellular metabolisms. Functional assay and hierarchical clustering for tracking protein dynamic change confirmed that cBM-MSCs required the presences of Wnt, transforming growth factor (TGF)-beta, and bone-morphogenetic protein (BMP) signaling, while cDPSCs mainly relied on BMP signaling presentation during osteogenic differentiation in vitro. Therefore, these findings illustrated the comprehensive data regarding an in vitro osteogenic differentiation behavior by cBM-MSCs and cDPSCs which is crucial for further mechanism study and the establishment of cMSC-based bone tissue engineering (BTE) for veterinary practice.
Collapse
|
8
|
Zeng J, Chen S, Li C, Ye Z, Lin B, Liang Y, Wang B, Ma Y, Chai X, Zhang X, Zhou K, Zhang Q, Zhang H. Mesenchymal stem/stromal cells-derived IL-6 promotes nasopharyngeal carcinoma growth and resistance to cisplatin via upregulating CD73 expression. J Cancer 2020; 11:2068-2079. [PMID: 32127934 PMCID: PMC7052921 DOI: 10.7150/jca.37932] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
Previous studies have implicated the important role of mesenchymal stem/stromal cells (MSCs) within tumor microenvironment (TME) in the pathogenesis and progression of nasopharyngeal carcinoma (NPC), but the potential mechanisms are still unclear. Herein, we showed that an elevated IL-6 level was positively correlated with elevated expression of CD73 in TME of NPC. NPC specimens with an IL-6highCD73high phenotype showed higher expression levels of gp80, gp130, p-STAT3, MMP-9 and α-SMA, and clinically, a poorer prognosis than those with an IL-6lowCD73low phenotype. We found that stimulation with conditioned media derived from IL-6 gene knocked out MSC (MSCIL6KO-CM) down-regulated the expression of CD73, IL-6, gp80, p-STAT3, and proliferative cell nuclear antigen (PCNA) in CNE-2 NPC cells. Meanwhile, NPC cells co-cultured with MSCIL6KO-CM were more sensitive to cisplatin than those co-cultured with MSC-CM. Additionally, MSC-derived IL-6 transcriptionally upregulated CD73 expression via activating STAT3 signaling pathway in NPC cells. In summary, our findings suggest that MSCs promote NPC progression and chemoresistance by upregulation of CD73 expression via activating STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia 19104, USA
| | - Shasha Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
| | - Caihong Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Ziyu Ye
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Yanfang Liang
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, The Fifth People's Hospital of Dongguan, Dongguan 523905, China
| | - Bin Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
| | - Yan Ma
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
| | - Xingxing Chai
- Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Xin Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, 524023 China
| | - Keyuan Zhou
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia 19104, USA
| | - Haitao Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan 523808, China
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
9
|
Chen X, Cai C, Xu D, Liu Q, Zheng S, Liu L, Li G, Zhang X, Li X, Ma Y, Huang L, Chen J, Shi J, Du X, Xia W, Xiang AP, Peng Y. Human Mesenchymal Stem Cell-Treated Regulatory CD23 +CD43 + B Cells Alleviate Intestinal Inflammation. Theranostics 2019; 9:4633-4647. [PMID: 31367246 PMCID: PMC6643430 DOI: 10.7150/thno.32260] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/21/2019] [Indexed: 01/06/2023] Open
Abstract
Rationale: Mesenchymal stem cells (MSCs) have been demonstrated to ameliorate inflammatory bowel disease by their actions on multiple immune cells, especially on regulatory B cells (Breg cells). However, the phenotypes and functions of human MSCs (hMSCs)-treated Breg cell subsets are not yet clear. Methods: Purified B cells were cocultured with MSCs and the phenotypes and immunomodulatory functions of the B cells were analyzed by FACS and proliferation assays in vitro. Also, a trinitrobenzenesulfonic acid-induced mouse colitis model was employed to detect the function of MSC-treated Breg cells in vivo. Results: We demonstrated that coculturing with hMSCs significantly enhanced the immunomodulatory activity of B cells by up-regulating IL-10 expression. We then identified that a novel regulatory B cell population characterized by CD23 and CD43 phenotypic markers could be induced by hMSCs. The CD23+CD43+ Breg cells substantially inhibited the inflammatory cytokine secretion and proliferation of T cells through an IL-10-dependent pathway. More significantly, intraperitoneal injection of hMSCs ameliorated the clinical and histopathological severity in the mouse experimental colitis model, accompanied by an increase in the number of CD23+CD43+ Breg cells. The adoptive transfer of CD23+CD43+ B cells effectively alleviated murine colitis, as compared with the CD23-CD43- B cells. Treatment with CD23+CD43+ B cells, and not hMSCs, substantially improved the symptoms of colitis in B cell-depleted mice. Conclusion: the novel CD23+CD43+ Breg cell subset appears to be involved in the immunomodulatory function of hMSCs and sheds new light on elucidating the therapeutic mechanism of hMSCs for the treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Xiaoyong Chen
- The Biotherapy Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chuang Cai
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dijing Xu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiuli Liu
- The Biotherapy Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Shuwei Zheng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Longshan Liu
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Gang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoping Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuanchen Ma
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jieying Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiahao Shi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin Du
- Hematological Department of Guangdong Province People's Hospital, Guangzhou 510080, China
| | - Wenjie Xia
- Institute of Blood Transfusion, Guangzhou Blood Centre, Guangzhou, 510095, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 511436, China
| | - Yanwen Peng
- The Biotherapy Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
10
|
Safavi AS, Rouhi G, Haghighipour N, Bagheri F, Eslaminejad MB, Sayahpour FA. Efficacy of mechanical vibration in regulating mesenchymal stem cells gene expression. In Vitro Cell Dev Biol Anim 2019; 55:387-394. [PMID: 30993556 DOI: 10.1007/s11626-019-00340-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
This study aimed at investigating the expression of osteoblast and chondrocyte-related genes in mesenchymal stem cells (MSCs), derived from rabbit adipose tissue, under mechanical vibration. The cells were placed securely on a vibrator's platform and subjected to 300 Hz of sinusoidal vibration, with a maximum amplitude of 10 μm, for 45 min per day, and for 14 consequent days, in the absence of biochemical reagents. The negative control group was placed in the conventional culture medium with no mechanical loading. The expression of osteoblast and chondrocyte-related genes was investigated using real-time polymerase chain reaction (real-time PCR). In addition, F-actin fiber structure and alignment with the help of actin filament fluorescence staining were evaluated, and the level of metabolic activity of MSCs was determined by the methyl thiazolyl tetrazolium assay. The real-time PCR study showed a significant increase of bone gene expression in differentiated cells, compared with MSCs (P < 0.05). On the other hand, the level of chondrocyte gene expression was not remarkable. Applying mechanical vibration enhanced F-actin fiber structure and made them aligned in a specific direction. It was also found that during the differentiation process, the metabolic activity of the cells increased (P < 0.05). The results of this work are in agreement with the well-accepted fact that the MSCs, in the absence of growth factors, are sensitive to low-amplitude, high-frequency vibration. Outcomes of this work can be applied in cell therapy and tissue engineering, when regulation of stem cells is required.
Collapse
Affiliation(s)
- Atiyeh Sadat Safavi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, P. O. Box 1591634311, Tehran, Iran
| | - Gholamreza Rouhi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, P. O. Box 1591634311, Tehran, Iran.
| | | | - Fatemeh Bagheri
- Biotechnology Group, Department of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Frough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
11
|
Li J, Guo X, Li M, Xiao Y, Bao C. [Research progress in the mechanism of protein factors in regulating bone remodeling]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:115-123. [PMID: 30644271 DOI: 10.7507/1002-1892.201808059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective To review the role and mechanism of protein factors in bone remodeling, and provides theoretical basis for further elucidating the pathogenesis and clinical treatment of bone-related diseases. Methods The relevant research results at home and abroad in recent years were extensively consulted, analyzed, and summarized. Results Bone remodeling is an important physiological process to maintain bone homeostasis. Protein, as an important stimulator in bone remodeling, regulates the balance between bone resorption and bone formation. Conclusion At present, the research on the mechanism of protein in bone remodeling is insufficient. Therefore, it is necessary to further study the specific time, process, and interaction network of protein in bone remodeling, and to confirm its mechanism in bone remodeling, so as to reveal and treat the pathogenesis of bone-related diseases.
Collapse
Affiliation(s)
- Ju Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China;National Clinical Research Center of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Xiaodong Guo
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China;National Clinical Research Center of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Mingzheng Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China;National Clinical Research Center of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China;National Clinical Research Center of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041,
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, 610041,
| |
Collapse
|
12
|
Li C, Sunderic K, Nicoll SB, Wang S. Downregulation of Heat Shock Protein 70 Impairs Osteogenic and Chondrogenic Differentiation in Human Mesenchymal Stem Cells. Sci Rep 2018; 8:553. [PMID: 29323151 PMCID: PMC5765044 DOI: 10.1038/s41598-017-18541-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) show promise for bone and cartilage regeneration. Our previous studies demonstrated that hMSCs with periodic mild heating had enhanced osteogenic and chondrogenic differentiation with significantly upregulated heat shock protein 70 (HSP70). However, the role of HSP70 in adult tissue regeneration is not well studied. Here, we revealed an essential regulatory mechanism of HSP70 in osteogenesis and chondrogenesis using adult hMSCs stably transfected with specific shRNAs to knockdown HSP70. Periodic heating at 39 °C was applied to hMSCs for up to 26 days. HSP70 knockdown resulted in significant reductions of alkaline phosphatase activity, calcium deposition, and gene expression of Runx2 and Osterix during osteogenesis. In addition, knockdown of HSP70 led to significant decreases of collagens II and X during chondrogenesis. Thus, downregulation of HSP70 impaired hMSC osteogenic and chondrogenic differentiation as well as the enhancement of these processes by thermal treatment. Taken together, these findings suggest a putative mechanism of thermal-enhanced bone and cartilage formation and underscore the importance of HSP70 in adult bone and cartilage differentiation.
Collapse
Affiliation(s)
- Chenghai Li
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA
| | - Kristifor Sunderic
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA
| | - Steven B Nicoll
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA
| | - Sihong Wang
- Department of Biomedical Engineering, City University of New York-City College, 160 Convent Avenue, New York, NY, 10031, USA.
| |
Collapse
|
13
|
Pan GH, Chen Z, Xu L, Zhu JH, Xiang P, Ma JJ, Peng YW, Li GH, Chen XY, Fang JL, Guo YH, Zhang L, Liu LS. Low-dose tacrolimus combined with donor-derived mesenchymal stem cells after renal transplantation: a prospective, non-randomized study. Oncotarget 2017; 7:12089-101. [PMID: 26933811 PMCID: PMC4914271 DOI: 10.18632/oncotarget.7725] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/29/2016] [Indexed: 12/22/2022] Open
Abstract
Calcineurin inhibitors, including tacrolimus, are largely responsible for advances in allotransplantation. However, the nephrotoxicity associated with these immunosuppressants impairs patients' long-term survival after renal allograft. Therefore, novel regimens that minimize or even eliminate calcineurin inhibitors could improve transplantation outcomes. In this pilot study, we investigated the use of low-dose tacrolimus in combination with mesenchymal stem cells (MSCs), which are immunosuppressive and prolong allograft survival in experimental organ transplant models. Donor-derived, bone marrow MSCs combined with a sparing dose of tacrolimus (0.04-0.05 mg/kg/day) were administered to 16 de novo living-related kidney transplant recipients; 16 other patients received a standard dose of tacrolimus (0.07-0.08 mg/kg/day). The safety of MSC infusion, acute rejection, graft function, graft survival, and patient survival were evaluated over ≥24 months following kidney transplantation. All patients survived and had stable renal function at the 24 month follow-up. The combination of low-dose tacrolimus and MSCs was as effective as standard dose tacrolimus in maintaining graft survival at least 2 years after transplantation. In addition, both groups had similar urea, urine protein, urinary RBC, urinary WBC, 24-h urine protein, and creatinine clearance rates from 7 days to 24 months after transplantation. Furthermore, no differences in the proportion of lymphocytes, CD19, CD3, CD34, CD38, and natural killer cells were detected between the control and experimental groups. None of the MSC recipients experienced immediate or long-term toxicity from the treatment. This preliminary data suggests that the addition of MSCs permits the use of lower dosages of nephrotoxic calcineurin inhibitors following renal transplantation.
Collapse
Affiliation(s)
- Guang-Hui Pan
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zheng Chen
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lu Xu
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jing-Hui Zhu
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, SunYat-sen University, Guangzhou, Guangdong, China
| | - Jun-Jie Ma
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan-Wen Peng
- Center for Stem Cell Biology and Tissue Engineering, SunYat-sen University, Guangzhou, Guangdong, China
| | - Guang-Hui Li
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Yong Chen
- Center for Stem Cell Biology and Tissue Engineering, SunYat-sen University, Guangzhou, Guangdong, China
| | - Jia-Li Fang
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yu-He Guo
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lei Zhang
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Long-Shan Liu
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
|
15
|
Thylur RP, Gowda R, Mishra S, Jun CD. Swiprosin-1: Its Expression and Diverse Biological Functions. J Cell Biochem 2017; 119:150-156. [PMID: 28590012 DOI: 10.1002/jcb.26199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 06/06/2017] [Indexed: 02/02/2023]
Abstract
Swiprosin-1/EFhd2 is a Ca2+ binding adapter protein involved in the various cellular functions. Swiprosin-1 is significantly upregulated in a number of pathological conditions of inflammation, neurodegeneration, and cancer. Swiprosin-1 associated with actin and its expression level amplifies the production of proinflammatory mediators and modulates the activation of transcription factor during immune cells activation. This review aims at providing an overview of the expression and function of swiprosin-1/EFhd2 in various pathophysiological conditions. We also discussed the key role of swiprosin-1 in immune cell activation, cell migration, apoptosis, humoral immunity, cancer invasion and metastasis, neuronal transport, and major signaling cascades. J. Cell. Biochem. 119: 150-156, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ramesh P Thylur
- School of Life Science, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Raghavendra Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Sumita Mishra
- Department of Pediatrics, Division of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chang-Duk Jun
- School of Life Science, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| |
Collapse
|
16
|
Investigation of the Cell Surface Proteome of Human Periodontal Ligament Stem Cells. Stem Cells Int 2016; 2016:1947157. [PMID: 27579043 PMCID: PMC4989088 DOI: 10.1155/2016/1947157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022] Open
Abstract
The present study examined the cell surface proteome of human periodontal ligament stem cells (PDLSC) compared to human fibroblasts. Cell surface proteins were prelabelled with CyDye before processing to extract the membrane lysates, which were separated using 2D electrophoresis. Selected differentially expressed protein “spots” were identified using Mass spectrometry. Four proteins were selected for validation: CD73, CD90, Annexin A2, and sphingosine kinase 1 previously associated with mesenchymal stem cells. Flow cytometric analysis found that CD73 and CD90 were highly expressed by human PDLSC and gingival fibroblasts but not by keratinocytes, indicating that these antigens could be used as potential markers for distinguishing between mesenchymal cells and epithelial cell populations. Annexin A2 was also found to be expressed at low copy number on the cell surface of human PDLSC and gingival fibroblasts, while human keratinocytes lacked any cell surface expression of Annexin A2. In contrast, sphingosine kinase 1 expression was detected in all the cell types examined using immunocytochemical analysis. These proteomic studies form the foundation to further define the cell surface protein expression profile of PDLSC in order to better characterise this cell population and help develop novel strategies for the purification of this stem cell population.
Collapse
|
17
|
Movahedi Najafabadi BAH, Abnosi MH. Boron Induces Early Matrix Mineralization via Calcium Deposition and Elevation of Alkaline Phosphatase Activity in Differentiated Rat Bone Marrow Mesenchymal Stem Cells. CELL JOURNAL 2016; 18:62-73. [PMID: 27054120 PMCID: PMC4819387 DOI: 10.22074/cellj.2016.3988] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/14/2015] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Boron (B) is essential for plant development and might be an essential micronutrient for animals and humans. This study was conducted to characterize the impact of boric acid (BA) on the cellular and molecular nature of differentiated rat bone marrow mesenchymal stem cells (BMSCs). MATERIALS AND METHODS In this experimental study, BMSCs were extracted and expanded to the 3rdpassage, then cultured in Dulbecco's Modified Eagle's Medium (DMEM) complemented with osteogenic media as well as 6 ng/ml and 6 µg/ml of BA. After 5, 10, 15 and 21 days the viability and the level of mineralization was determined using MTT assay and alizarin red respectively. In addition, the morphology, nuclear diameter and cytoplasmic area of the cells were studied with the help of fluorescent dye. The concentration of calcium, activity of alanine transaminase (ALT), aspartate transaminase (AST), lactate dehydrogenase (LDH) and alkaline phosphatase (ALP) as well as sodium and potassium levels were also evaluated using commercial kits and a flame photometer respectively. RESULTS Although 6 µg/ml of BA was found to be toxic, a concentration of 6 ng/ml increased the osteogenic ability of the cell significantly throughout the treatment. In addition it was observed that B treatment caused the early induction of matrix mineralization compared to controls. CONCLUSION Although more investigation is required, we suggest the prescription of a very low concentration of B in the form of BA or foods containing BA, in groups at high risk of osteoporosis or in the case of bone fracture.
Collapse
|
18
|
Lo YP, Liu YS, Rimando MG, Ho JHC, Lin KH, Lee OK. Three-dimensional spherical spatial boundary conditions differentially regulate osteogenic differentiation of mesenchymal stromal cells. Sci Rep 2016; 6:21253. [PMID: 26884253 PMCID: PMC4756701 DOI: 10.1038/srep21253] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/20/2016] [Indexed: 01/09/2023] Open
Abstract
The spatial boundary condition (SBC) arising from the surrounding microenvironment imposes specific geometry and spatial constraints that affect organogenesis and tissue homeostasis. Mesenchymal stromal cells (MSCs) sensitively respond to alterations of mechanical cues generated from the SBC. However, mechanical cues provided by a three-dimensional (3D) environment are deprived in a reductionist 2D culture system. This study investigates how SBC affects osteogenic differentiation of MSCs using 3D scaffolds with monodispersed pores and homogenous spherical geometries. MSCs cultured under SBCs with diameters of 100 and 150 μm possessed the greatest capability of osteogenic differentiation. This phenomenon was strongly correlated with MSC morphology, organization of actin cytoskeleton, and distribution of focal adhesion involving α2 and α5 integrins. Further silencing either α2 or α5 integrin significantly reduced the above mentioned mechanosensitivity, indicating that the α2 and α5 integrins as mechano-sensitive molecules mediate MSCs' ability to provide enhanced osteogenic differentiation in response to different spherical SBCs. Taken together, the findings provide new insights regarding how MSCs respond to mechanical cues from the surrounding microenvironment in a spherical SBC, and such biophysical stimuli should be taken into consideration in tissue engineering and regenerative medicine in conjunction with biochemical cues.
Collapse
Affiliation(s)
- Yin-Ping Lo
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 11221, Taiwan
| | - Yi-Shiuan Liu
- Stem Cell Research Center, National Yang-Ming University, Taipei 11221, Taiwan
| | - Marilyn G Rimando
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 11221, Taiwan
| | - Jennifer Hui-Chun Ho
- Center for Stem Cell Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan.,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei 11031, Taiwan.,Department of Ophthalmology, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Keng-Hui Lin
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Oscar K Lee
- Taipei City Hospital, Taipei 10341, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| |
Collapse
|
19
|
Inserra I, Martelli C, Cipollina M, Cicione C, Iavarone F, Taranto GD, Barba M, Castagnola M, Desiderio C, Lattanzi W. Lipoaspirate fluid proteome: A preliminary investigation by LC-MS top-down/bottom-up integrated platform of a high potential biofluid in regenerative medicine. Electrophoresis 2016; 37:1015-26. [PMID: 26719138 DOI: 10.1002/elps.201500504] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/14/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022]
Abstract
The lipoaspirate fluid (LAF) is emerging as a potentially valuable source in regenerative medicine. In particular, our group recently demonstrated that it is able to exert osteoinductive properties in vitro. This original observation stimulated the investigation of the proteomic component of LAF, by means of LC-ESI-LTQ-Orbitrap-MS top-down/bottom-up integrated approach, which represents the object of the present study. Top-down analyses required the optimization of sample pretreatment procedures to enable the correct investigation of the intact proteome. Bottom-up analyses have been directly applied to untreated samples after monodimensional SDS-PAGE separation. The analysis of the acid-soluble fraction of LAF by top-down approach allowed demonstrating the presence of albumin and hemoglobin fragments (i.e. VV- and LVV-hemorphin-7), thymosins β4 and β10 peptides, ubiquitin and acyl-CoA binding protein; adipogenesis regulatory factor, perilipin-1 fragments, and S100A6, along with their PTMs. Part of the bottom-up proteomic profile was reproducibly found in both tested samples. The bottom-up approach allowed demonstrating the presence of proteins, listed among the components of adipose tissue and/or comprised within the ASCs intracellular content and secreted proteome. Our data provide a first glance on the LAF molecular profile, which is consistent with its tissue environment. LAF appeared to contain bioactive proteins, peptides and paracrine factors, suggesting its potential translational exploitation.
Collapse
Affiliation(s)
- Ilaria Inserra
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Martelli
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mara Cipollina
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy.,Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Claudia Cicione
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Iavarone
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Di Taranto
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marta Barba
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimo Castagnola
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy.,Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Claudia Desiderio
- Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Wanda Lattanzi
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, Rome, Italy.,Banca del Tessuto Muscolo-Scheletrico della Regione Lazio, Università Cattolica del, Sacro Cuore, Roma, Italy
| |
Collapse
|
20
|
Peng Y, Chen X, Liu Q, Xu D, Zheng H, Liu L, Liu Q, Liu M, Fan Z, Sun J, Li X, Zou R, Xiang AP. Alteration of naïve and memory B-cell subset in chronic graft-versus-host disease patients after treatment with mesenchymal stromal cells. Stem Cells Transl Med 2014; 3:1023-31. [PMID: 25015640 DOI: 10.5966/sctm.2014-0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although mesenchymal stromal cells (MSCs) possess immunomodulatory properties and exhibit promising efficacy against chronic graft-versus-host disease (cGVHD), little is known about the immune changes by which MSCs ameliorate cGVHD in vivo. Recent studies have suggested that B lymphocytes might play an important role in the pathogenesis of cGVHD. In this study, we investigated changes in the numbers, phenotypes, and subpopulations of B lymphocytes in cGVHD patients who showed a complete response (CR), partial response (PR), or no response (NR) after MSC treatment. We found that the frequencies and numbers of CD27+ memory and pre-germinal center B lymphocytes were significantly increased in the CR and PR cGVHD patients after MSC treatment but decreased in the NR patients. A further analysis of CR/PR cGVHD patients showed that MSC treatment led to a decrease in the plasma levels of B cell-activating factor (BAFF) and increased expression of the BAFF receptor (BAFF-R) on peripheral B lymphocytes but no changes in plasma BAFF levels or BAFF-R expression on B lymphocytes in NR patients. Overall, our findings imply that MSCs might exert therapeutic effects in cGVHD patients, accompanied by alteration of naïve and memory B-cell subsets, modulating plasma BAFF levels and BAFF-R expression on B lymphocytes.
Collapse
Affiliation(s)
- Yanwen Peng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qifa Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dijing Xu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Haiqing Zheng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Longshan Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qiuli Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Muyun Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhiping Fan
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Sun
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaobo Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ruifeng Zou
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
21
|
Mateos J, Pernas PF, Labora JF, Blanco F, Arufe MDC. Proteomic Applications in the Study of Human Mesenchymal Stem Cells. Proteomes 2014; 2:53-71. [PMID: 28250369 PMCID: PMC5302726 DOI: 10.3390/proteomes2010053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/15/2014] [Accepted: 01/26/2014] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are undifferentiated cells with an unlimited capacity for self-renewal and able to differentiate towards specific lineages under appropriate conditions. MSCs are, a priori, a good target for cell therapy and clinical trials as an alternative to embryonic stem cells, avoiding ethical problems and the chance for malignant transformation in the host. However, regarding MSCs, several biological implications must be solved before their application in cell therapy, such as safe ex vivo expansion and manipulation to obtain an extensive cell quantity amplification number for use in the host without risk accumulation of genetic and epigenetic abnormalities. Cell surface markers for direct characterization of MSCs remain unknown, and the precise molecular mechanisms whereby growth factors stimulate their differentiation are still missing. In the last decade, quantitative proteomics has emerged as a promising set of techniques to address these questions, the answers to which will determine whether MSCs retain their potential for use in cell therapy. Proteomics provides tools to globally analyze cellular activity at the protein level. This proteomic profiling allows the elucidation of connections between broad cellular pathways and molecules that were previously impossible to determine using only traditional biochemical analysis. However; thus far, the results obtained must be orthogonally validated with other approaches. This review will focus on how these techniques have been applied in the evaluation of MSCs for their future applications in safe therapies.
Collapse
Affiliation(s)
- Jesús Mateos
- Rheumatology Division, ProteoRed/ISCIII, INIBIC-Hospital Universitario A Coruña, A Coruña 15006, Spain.
| | - Pablo Fernández Pernas
- CIBER-BBN, INIBIC-Hospital Universitario A Coruña, A Coruña 15006, Spain.
- Department of Medicine, University of A Coruña, A Coruña 15006, Spain.
| | - Juan Fafián Labora
- CIBER-BBN, INIBIC-Hospital Universitario A Coruña, A Coruña 15006, Spain.
- Department of Medicine, University of A Coruña, A Coruña 15006, Spain.
| | - Francisco Blanco
- Rheumatology Division, ProteoRed/ISCIII, INIBIC-Hospital Universitario A Coruña, A Coruña 15006, Spain.
- CIBER-BBN, INIBIC-Hospital Universitario A Coruña, A Coruña 15006, Spain.
- Department of Medicine, University of Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - María Del Carmen Arufe
- CIBER-BBN, INIBIC-Hospital Universitario A Coruña, A Coruña 15006, Spain.
- Department of Medicine, University of A Coruña, A Coruña 15006, Spain.
| |
Collapse
|
22
|
Zhu Z, Liu Z, Liu J, Bi M, Yang T, Wang J. Proteomic profiling of human placenta-derived mesenchymal stem cells upon transforming LIM mineralization protein-1 stimulation. Cytotechnology 2014; 67:285-97. [PMID: 24468833 DOI: 10.1007/s10616-013-9684-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 12/23/2013] [Indexed: 02/07/2023] Open
Abstract
Human placenta-derived mesenchymal stem cells (hPDMSCs) can differentiate into different types of cells and thus have tremendous potential for cell therapy and tissue engineering. LIM mineralization protein-1 (LMP-1) plays an important role in osteoblast differentiation, maturation and bone formation. To determine a global effect of LMP-1 on hPDMSCs, we designed a study using a proteomic approach combined with adenovirus-mediated gene transfer of LMP-1 to identify LMP-1-induced changes in hPDMSCs on proteome level. We have generated proteome maps of undifferentiated hPDMSCs and LMP-1 induced hPDMSCs. Two dimensional gel electrophoresis revealed 22 spots with at least 2.0-fold changes in expression and 15 differently expressed proteins were successfully identified by MALDI-TOF-MS. The proteins regulated by LMP-1 included cytoskeletal proteins, cadmium-binding proteins, and metabolic proteins, etc. The expression of some identified proteins was confirmed by further Western blot analyses. Our results will play an important role in better elucidating the underlying molecular mechanism in LMP-1 included hPDMSCs differentiation into osteoblasts.
Collapse
Affiliation(s)
- Zhen Zhu
- Stomatology Hospital, Jilin University, Changchun, 130021, People's Republic of China
| | | | | | | | | | | |
Collapse
|
23
|
Granéli C, Thorfve A, Ruetschi U, Brisby H, Thomsen P, Lindahl A, Karlsson C. Novel markers of osteogenic and adipogenic differentiation of human bone marrow stromal cells identified using a quantitative proteomics approach. Stem Cell Res 2014; 12:153-65. [DOI: 10.1016/j.scr.2013.09.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 09/17/2013] [Accepted: 09/19/2013] [Indexed: 12/22/2022] Open
|
24
|
Anjos L, Gomes AS, Redruello B, Reinhardt R, Canário AV, Power DM. PTHrP-induced modifications of the sea bream (Sparus auratus) vertebral bone proteome. Gen Comp Endocrinol 2013; 191:102-12. [PMID: 23747812 DOI: 10.1016/j.ygcen.2013.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 04/03/2013] [Accepted: 05/28/2013] [Indexed: 02/04/2023]
Abstract
Endocrine factors play an essential role in the formation and turnover of the skeleton in vertebrates. In the present study sea bream vertebral bone transcripts for PTH1R and PTH3R were identified and the action of intermittent administration of parathyroid hormone related protein (PTHrP) on the proteome of vertebral bone was analysed. Treatment of immature sea bream (Sparus auratus, n=6) for 5days with homologous recombinant PTHrP(1-125; 150ng/g body weight) modified bone metabolism and caused a significant (p<0.05) reduction in both tartrate resistant acid phosphatase (TRACP) and alkaline phosphatase (ALP) in relation to control fish. However, the ratio of TRACP: ALP in PTHrP treated fish (1.3 to 2.2 cf. control) suggested it had an anabolic response. A sea bream vertebral bone proteome of 157 protein spots was generated and putative identity assigned to 118 (75.2%) proteins of which 72% had homology to proteins/transcripts from teleosts many of which have not previously been reported in teleost bone. Classification of bone proteins using gene ontology revealed those with protein or metal/ion (e.g., calcium, magnesium, zinc) binding (∼53%) activities were most abundant. The expression of eight proteins was significantly (p<0.05) modified in the vertebra of PTHrP treated compared to control fish; three were up-regulated, betainehomocystein S-methyltransferase, glial fibrillary acidic protein, parvalbumin beta and five were down-regulated, annexin A5, apolipoprotein A1, myosin light chain 2, fast skeletal myosin light chain 3, troponin C. In conclusion, intermittent administration of PTHrP to sea bream is associated with an anabolic response in vertebral bone metabolism and modifies calcium binding proteins in the proteome.
Collapse
Affiliation(s)
- Liliana Anjos
- Comparative and Molecular Endocrinology Group, CCMAR, CIMAR Laboratório Associado, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.
| | | | | | | | | | | |
Collapse
|
25
|
Donor-Derived Mesenchymal Stem Cells Combined With Low-Dose Tacrolimus Prevent Acute Rejection After Renal Transplantation. Transplantation 2013; 95:161-8. [DOI: 10.1097/tp.0b013e3182754c53] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
26
|
Brohlin M, Kingham PJ, Novikova LN, Novikov LN, Wiberg M. Aging effect on neurotrophic activity of human mesenchymal stem cells. PLoS One 2012; 7:e45052. [PMID: 23028757 PMCID: PMC3444498 DOI: 10.1371/journal.pone.0045052] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 08/15/2012] [Indexed: 12/18/2022] Open
Abstract
Clinical efficacy of stem cells for nerve repair is likely to be influenced by issues including donor age and in vitro expansion time. We isolated human mesenchymal stem cells (MSC) from bone marrow of young (16–18 years) and old (67–75 years) donors and analyzed their capacity to differentiate and promote neurite outgrowth from dorsal root ganglia (DRG) neurons. Treatment of MSC with growth factors (forskolin, basic fibroblast growth factor, platelet derived growth factor-AA and glial growth factor-2) induced protein expression of the glial cell marker S100 in cultures from young but not old donors. MSC expressed various neurotrophic factor mRNA transcripts. Growth factor treatment enhanced the levels of BDNF and VEGF transcripts with corresponding increases in protein release in both donor cell groups. MSC in co-culture with DRG neurons significantly enhanced total neurite length which, in the case of young but not old donors, was further potentiated by treatment of the MSC with the growth factors. Stem cells from young donors maintained their proliferation rate over a time course of 9 weeks whereas those from the old donors showed increased population doubling times. MSC from young donors, differentiated with growth factors after long-term culture, maintained their ability to enhance neurite outgrowth of DRG. Therefore, MSC isolated from young donors are likely to be a favourable cell source for nerve repair.
Collapse
Affiliation(s)
- Maria Brohlin
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, Umeå, Sweden
| | - Paul J. Kingham
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, Umeå, Sweden
- * E-mail:
| | - Liudmila N. Novikova
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, Umeå, Sweden
| | - Lev N. Novikov
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, Umeå, Sweden
| | - Mikael Wiberg
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, Umeå, Sweden
- Department of Surgical and Perioperative Sciences, Section of Hand and Plastic Surgery, Umeå University, Umeå, Sweden
| |
Collapse
|
27
|
Gurkan UA, Golden R, Kishore V, Riley CP, Adamec J, Akkus O. Immune and inflammatory pathways are involved in inherent bone marrow ossification. Clin Orthop Relat Res 2012; 470:2528-40. [PMID: 22798134 PMCID: PMC3830098 DOI: 10.1007/s11999-012-2459-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Bone marrow plays a key role in bone formation and healing. Although a subset of marrow explants ossifies in vitro without excipient osteoinductive factors, some explants do not undergo ossification. The disparity of outcome suggests a significant heterogeneity in marrow tissue in terms of its capacity to undergo osteogenesis. QUESTIONS/PURPOSES We sought to identify: (1) proteins and signaling pathways associated with osteogenesis by contrasting the proteomes of ossified and poorly ossified marrow explants; and (2) temporal changes in proteome and signaling pathways of marrow ossification in the early and late phases of bone formation. METHODS Explants of marrow were cultured. Media conditioned by ossified (n = 4) and poorly ossified (n = 4) subsets were collected and proteins unique to each group were identified by proteomic analysis. Proteomic data were processed to assess proteins specific to the early phase (Days 1-14) and late phase (Days 15-28) of the culture period. Pathways involved in bone marrow ossification were identified through bioinformatics. RESULTS Twenty-eight proteins were unique to ossified samples and eight were unique to poorly ossified ones. Twelve proteins were expressed during the early phase and 15 proteins were specific to the late phase. Several identified pathways corroborated those reported for bone formation in the literature. Immune and inflammatory pathways were specific to ossified samples. CONCLUSIONS The marrow explant model indicates the inflammatory and immune pathways to be an integral part of the osteogenesis process.
Collapse
Affiliation(s)
- Umut Atakan Gurkan
- />Harvard-MIT Division of Health Sciences and Technology, Brigham and Women’s Hospital, Harvard Medical School, 65 Landsdowne Street, PRB 252, Cambridge, MA 02139 USA
| | - Ryan Golden
- />Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN USA
| | - Vipuil Kishore
- />Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| | - Catherine P. Riley
- />Department of Research and Development Pathology Associates, Medical Laboratories, Spokane, WA 99204 USA
| | - Jiri Adamec
- />Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE USA
| | - Ozan Akkus
- />Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- />Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- />Department of Orthopaedics, University Hospitals of Cleveland, Cleveland, OH USA
| |
Collapse
|
28
|
IFN-γ-primed human bone marrow mesenchymal stem cells induce tumor cell apoptosis in vitro via tumor necrosis factor-related apoptosis-inducing ligand. Int J Biochem Cell Biol 2012; 44:1305-14. [DOI: 10.1016/j.biocel.2012.04.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 04/14/2012] [Accepted: 04/18/2012] [Indexed: 12/26/2022]
|
29
|
Faça VM. Human mesenchymal stromal cell proteomics: contribution for identification of new markers and targets for medicine intervention. Expert Rev Proteomics 2012; 9:217-30. [PMID: 22462791 DOI: 10.1586/epr.12.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) have become of great interest for cell-based therapy owing to their roles in tissue repair and immune suppression. MSCs have the ability to differentiate into specialized tissues, including bone, cartilage and muscle, among several others. Furthermore, it has been found that MSCs can also serve as cellular factories that secrete mediators to stimulate in situ regeneration of injured tissues. Proteomics has contributed significantly to the identification of new proteins to improve cellular characterization of MSCs, to identify new targets for therapeutic intervention and to elucidate important pathways utilized by MSCs to differentiate into distinct tissues. As proteomics technology advances, several studies can be revisited and analyzed in depth, employing state-of-the-art approaches, helping to uncover the cellular mechanisms utilized by MSCs to exert their regenerative functionalities. In this article, we will review the progress made so far and discuss further opportunities for proteomics to contribute to the clinical applications of MSCs.
Collapse
Affiliation(s)
- Vitor Marcel Faça
- Department of Biochemistry & Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Brazil.
| |
Collapse
|
30
|
Peter S, Evans C, Ow SY, Scutt AM, Wright PC, Biggs CA. Proteomic analysis of the impact of static culturing on the expansion of rat bone marrow mesenchymal stem cells. Biotechnol Lett 2012; 34:1589-96. [DOI: 10.1007/s10529-012-0935-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 04/17/2012] [Indexed: 10/28/2022]
|
31
|
Tiwari A, Tursky ML, Mushahary D, Wasnik S, Collier FM, Suma K, Kirkland MA, Pande G. Ex vivo expansion of haematopoietic stem/progenitor cells from human umbilical cord blood on acellular scaffolds prepared from MS-5 stromal cell line. J Tissue Eng Regen Med 2012; 7:871-83. [PMID: 22511368 DOI: 10.1002/term.1479] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 11/18/2011] [Accepted: 01/16/2012] [Indexed: 12/13/2022]
Abstract
Lineage-specific expansion of haematopoietic stem/progenitor cells (HSPCs) from human umbilical cord blood (UCB) is desirable because of their several applications in translational medicine, e.g. treatment of cancer, bone marrow failure and immunodeficiencies. The current methods for HSPC expansion use either cellular feeder layers and/or soluble growth factors and selected matrix components coated on different surfaces. The use of cell-free extracellular matrices from bone marrow cells for this purpose has not previously been reported. We have prepared insoluble, cell-free matrices from a murine bone marrow stromal cell line (MS-5) grown under four different conditions, i.e. in presence or absence of osteogenic medium, each incubated under 5% and 20% O₂ tensions. These acellular matrices were used as biological scaffolds for the lineage-specific expansion of magnetically sorted CD34⁺ cells and the results were evaluated by flow cytometry and colony-forming assays. We could get up to 80-fold expansion of some HSPCs on one of the matrices and our results indicated that oxygen tension played a significant role in determining the expansion capacity of the matrices. A comparative proteomic analysis of the matrices indicated differential expression of proteins, such as aldehyde dehydrogenase and gelsolin, which have previously been identified as playing a role in HSPC maintenance and expansion. Our approach may be of value in identifying factors relevant to tissue engineering-based ex vivo HSPC expansion, and it may also provide insights into the constitution of the niche in which these cells reside in the bone marrow.
Collapse
Affiliation(s)
- Abhilasha Tiwari
- CSIR Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India; Deakin University, Waurn Ponds, Geelong, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kim HJ, Ji BR, Kim JS, Lee HN, Ha DH, Kim CW. Proteomic analysis of proteins associated with cellular senescence by calorie restriction in mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2012; 48:186-95. [DOI: 10.1007/s11626-012-9485-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/06/2012] [Indexed: 01/01/2023]
|
33
|
Lo T, Tsai CF, Shih YRV, Wang YT, Lu SC, Sung TY, Hsu WL, Chen YJ, Lee OK. Phosphoproteomic Analysis of Human Mesenchymal Stromal Cells during Osteogenic Differentiation. J Proteome Res 2011; 11:586-98. [DOI: 10.1021/pr200868p] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ting Lo
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Chia-Feng Tsai
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yu-Ru V. Shih
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Wang
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Sheng-Chieh Lu
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Ting-Yi Sung
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Wen-Lian Hsu
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yu-Ju Chen
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Oscar K. Lee
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Bozic D, Grgurevic L, Erjavec I, Brkljacic J, Orlic I, Razdorov G, Grgurevic I, Vukicevic S, Plancak D. The proteome and gene expression profile of cementoblastic cells treated by bone morphogenetic protein-7 in vitro. J Clin Periodontol 2011; 39:80-90. [PMID: 22093042 DOI: 10.1111/j.1600-051x.2011.01794.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2011] [Indexed: 11/28/2022]
Abstract
AIM Regenerative periodontal therapy is often unpredictable and limited. Cementum regeneration is necessary for the proper repair of a periodontal ligament. The precise mechanism how bone morphogenetic protein-7 (BMP7) induces differentiation and mineralization of cementoblasts remains undetermined. The purpose of this study was to evaluate the effect of BMP7 on early proteome and gene expression profile of cementoblastic OCCM.30 cells in vitro. MATERIALS AND METHODS Immortalized murine cementoblasts (OCCM.30) were exposed to BMP7 and evaluated for: (1) proliferation; (2) mineralization; (3) early proteome profile using liquid chromatography-mass spectrometry (LC-MS); and (4) gene expression by quantitative RT-PCR. RESULTS Bone morphogenetic protein-7 increased the cell proliferation at 24 h and 48 h, while higher doses suppressed the cell proliferation at 48 h. BMP7 induced the mineralization of cementoblasts following 8 days of therapy. Using LC-MS we identified 1117 proteins from the cell lysate. Many belonged to extracellular matrix formation such as PCPE1, collagens, annexins and integrin receptors. RT-PCR analyses revealed a BMP7 dose-dependent upregulation of BMP1, TGFβ1, osterix, osteoprotegerin, procollagen I and II, PCPE1, and noggin, while BMP6 and chordin expression were decreased. The high BMP7 dose down regulated most of the genes 24 h following therapy. CONCLUSION Bone morphogenetic protein-7 promotes differentiation and mineralization of cementoblasts via inducing PCPE1 and BMP1 responsible for processing of type I collagen.
Collapse
Affiliation(s)
- Darko Bozic
- Department of Periodontology, University of Zagreb, School of Dental Medicine, Croatia
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hong Y, Xu GX. Proteome changes during bone mesenchymal stem cell differentiation into photoreceptor-like cells in vitro. Int J Ophthalmol 2011; 4:466-73. [PMID: 22553704 DOI: 10.3980/j.issn.2222-3959.2011.05.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 09/19/2011] [Indexed: 01/14/2023] Open
Abstract
Human bone marrow stem cell (BMSC) may be directed to differentiate into multiple cell types, including adipocyte, chondrocyte, osteocyte and photoreceptor, among others. At present, little is known about the features of the BMSC and the protein control mechanism underlying their differentiation into photoreceptor-like cells. In the present study, BMSCs are induced to differentiate into photoreceptor-like cells in an in vitro model simulating the in vivo microenvironment. Up to 32 proteins are identified and differentially expressed through two-dimensional difference gel electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry to establish a differential protein database for photoreceptor-like cells from BMSC-induced differentiation. Western blot analysis further confirms the expression of some of the identified proteins. The present study proposes the total protein expression and possible molecular mechanism during the differentiation of BMSCs into photoreceptor cells.
Collapse
Affiliation(s)
- Yu Hong
- Fujian Institute of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | | |
Collapse
|
36
|
Human platelet lysate supports ex vivo expansion and enhances osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. Cell Biol Int 2011; 35:639-43. [PMID: 21235529 DOI: 10.1042/cbi20100361] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MSCs (mesenchymal stem cells) with their versatile growth and differentiation potential are ideal candidates for use in regenerative medicine and are currently making their way into clinical trials, which requires the development of xeno-free protocols for their culture. In this study, MSCs were cultured in 10% FCS or 7.5% HPL (human platelet lysate)-supplemented media. We found that both groups of MSCs showed a comparable morphology, phenotype and proliferation. The percentage of cells in the S- and G2-/M-phases, however, was slightly up-regulated (P<0.01) in HPL group. HPL contains PDGF (platelet derived growth factor)-AB and IGF (insulin-like growth factor)-1. In addition, compared with FCS group, MSCs in HPL group showed an increase in osteogenic differentiation and a decrease in adipogenic differentiation. In conclusion, MSCs in HPL-supplemented media maintained similar growing potential and phenotype, while osteogenic potential was enhanced. HPL offers a promising alternative to FCS for MSC expansion for clinical application, especially in bone injury diseases.
Collapse
|
37
|
Pietilä M, Palomäki S, Lehtonen S, Ritamo I, Valmu L, Nystedt J, Laitinen S, Leskelä HV, Sormunen R, Pesälä J, Nordström K, Vepsäläinen A, Lehenkari P. Mitochondrial function and energy metabolism in umbilical cord blood- and bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2011; 21:575-88. [PMID: 21615273 DOI: 10.1089/scd.2011.0023] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive choice for a variety of cellular therapies. hMSCs can be isolated from many different tissues and possess unique mitochondrial properties that can be used to determine their differentiation potential. Mitochondrial properties may possibly be used as a quality measure of hMSC-based products. Accordingly, the present work focuses on the mitochondrial function of hMSCs from umbilical cord blood (UCBMSC) cells and bone marrow cells from donors younger than 18 years of age (BMMSC <18) and those more than 50 years of age (BMMSC >50). Changes of ultrastructure and energy metabolism during osteogenic differentiation in all hMSC types were studied in detail. Results show that despite similar surface antigen characteristics, the UCBMSCs had smaller cell surface area and possessed more abundant rough endoplasmic reticulum than BMMSC >50. BMMSC <18 were morphologically more UCBMSC-like. UCBMSC showed dramatically higher mitochondrial-to-cytoplasm area ratio and elevated superoxide and manganese superoxide dismutase (MnSOD) levels as compared with BMMSC >50 and BMMSC <18. All hMSCs types showed changes indicative of mitochondrial activation after 2 weeks of osteogenic differentiation, and the increase in mitochondrial-to-cytoplasm area ratio appears to be one of the first steps in the differentiation process. However, BMMSC >50 showed a lower level of mitochondrial maturation and differentiation capacity. UCBMSCs and BMMSCs also showed a different pattern of exocytosed proteins and glycoproteoglycansins. These results indicate that hMSCs with similar cell surface antigen expression have different mitochondrial and functional properties, suggesting different maturation levels and other significant biological variations of the hMSCs. Therefore, it appears that mitochondrial analysis presents useful characterization criteria for hMSCs intended for clinical use.
Collapse
Affiliation(s)
- Mika Pietilä
- Department of Anatomy and Cell Biology, Institute of Biomedicine, University of Oulu, Oulu, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kasap M, Karaoz E, Akpinar G, Aksoy A, Erman G. A unique Golgi apparatus distribution may be a marker for osteogenic differentiation of hDP-MSCs. Cell Biochem Funct 2011; 29:489-95. [DOI: 10.1002/cbf.1776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/20/2011] [Accepted: 06/03/2011] [Indexed: 01/07/2023]
Affiliation(s)
- Murat Kasap
- Department of Medical Biology/KABI Proteomics Laboratory; Kocaeli University Medical School; Kocaeli; Turkey
| | - Erdal Karaoz
- Applied Stem Cell and Gene Therapy Research Center; Kocaeli University; Kocaeli; Turkey
| | - Gurler Akpinar
- Department of Medical Biology/KABI Proteomics Laboratory; Kocaeli University Medical School; Kocaeli; Turkey
| | - Ayca Aksoy
- Applied Stem Cell and Gene Therapy Research Center; Kocaeli University; Kocaeli; Turkey
| | - Gulay Erman
- Applied Stem Cell and Gene Therapy Research Center; Kocaeli University; Kocaeli; Turkey
| |
Collapse
|
39
|
Niehage C, Steenblock C, Pursche T, Bornhäuser M, Corbeil D, Hoflack B. The cell surface proteome of human mesenchymal stromal cells. PLoS One 2011; 6:e20399. [PMID: 21637820 PMCID: PMC3102717 DOI: 10.1371/journal.pone.0020399] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Accepted: 04/23/2011] [Indexed: 12/12/2022] Open
Abstract
Background Multipotent human mesenchymal stromal cells (hMSCs) are considered as promising biological tools for regenerative medicine. Their antibody-based isolation relies on the identification of reliable cell surface markers. Methodology/Principal Findings To obtain a comprehensive view of the cell surface proteome of bone marrow-derived hMSCs, we have developed an analytical pipeline relying on cell surface biotinylation of intact cells using cell impermeable, cleavable sulfo-NHS-SS-biotin to enrich the plasma membrane proteins and mass spectrometry for identification with extremely high confidence. Among the 888 proteins identified, we found ≈200 bona fide plasma membrane proteins including 33 cell adhesion molecules and 26 signaling receptors. In total 41 CD markers including 5 novel ones (CD97, CD112, CD239, CD276, and CD316) were identified. The CD markers are distributed homogenously within plastic-adherent hMSC populations and their expression is modulated during the process of adipogenesis or osteogenesis. Moreover, our in silico analysis revealed a significant difference between the cell surface proteome of hMSCs and that of human embryonic stem cells reported previously. Conclusions/Significance Collectively, our analytical methods not only provide a basis for further studies of mechanisms maintaining the multipotency of hMSCs within their niches and triggering their differentiation after signaling, but also a toolbox for a refined antibody-based identification of hMSC populations from different tissues and their isolation for therapeutic intervention.
Collapse
Affiliation(s)
- Christian Niehage
- Biotechnology Center, Dresden University of Technology, Dresden, Germany
| | | | - Theresia Pursche
- Biotechnology Center, Dresden University of Technology, Dresden, Germany
| | - Martin Bornhäuser
- Department of Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Denis Corbeil
- Biotechnology Center, Dresden University of Technology, Dresden, Germany
| | - Bernard Hoflack
- Biotechnology Center, Dresden University of Technology, Dresden, Germany
- * E-mail:
| |
Collapse
|
40
|
Saad FA, Hofstaetter JG. Proteomic analysis of mineralising osteoblasts identifies novel genes related to bone matrix mineralisation. INTERNATIONAL ORTHOPAEDICS 2011; 35:447-51. [PMID: 20556378 PMCID: PMC3047647 DOI: 10.1007/s00264-010-1076-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 06/03/2010] [Accepted: 06/03/2010] [Indexed: 11/30/2022]
Abstract
Bone matrix mineralisation plays a critical role in the determination of the overall biomechanical competence of bone. However, the molecular mechanisms of bone matrix mineralisation have not been fully elucidated. We used a proteomic approach to identify proteins and genes that may play a role in osteoblast matrix mineralisation. Proteomic differential display revealed a protein band that appeared only in mineralising mouse 7F2 osteoblasts. In-gel protein digestion and mass spectrometry proteomic analysis of this protein band identified 16 proteins. Furthermore, their corresponding transcripts were upregulated. This identification of proteins that may be associated with bone matrix mineralisation presents important new information toward a better understanding of the precise mechanisms of this process.
Collapse
Affiliation(s)
- Fawzy A Saad
- Department of Orthopaedic Surgery, Harvard Medical School, Children's Hospital Boston, Boston, MA, 02115, USA.
| | | |
Collapse
|
41
|
Giusta MS, Andrade H, Santos AV, Castanheira P, Lamana L, Pimenta AMC, Goes AM. Proteomic analysis of human mesenchymal stromal cells derived from adipose tissue undergoing osteoblast differentiation. Cytotherapy 2011; 12:478-90. [PMID: 20230220 DOI: 10.3109/14653240903580270] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Stem cells derived from human adipose tissue (ASC) have the capacity for renewal, are easily obtained and have plasticity properties that allow them to differentiate into several cell types, including osteoblast cells. With the aim of understanding the issue of the osteogenic process and finding reliable biomarkers in cells undergoing the osteogeneic differentiation process, this work took advantage of a proteomic approach to identify proteins involved in osteogenesis. METHODS For this purpose, ASC were analyzed under three conditions: S0, in the absence of stimulation; S1, with 2 weeks of osteogenic medium stimulation; and S2, with 4 weeks of osteogenic medium stimulation. The identification of ASC was carried out by flow cytometry using antibodies specific to known undifferentiated stem cell-surface markers. Cell viability, enzymatic activity, mineral deposition, collagen structure and production and gene analyzes were evaluated for each condition. RESULTS Phenotypic modifications were observed during the in vitro osteogenic differentiation process by two-dimensional (2-D) differential image gel electrophoresis (DIGE). The proteins were identified by mass espectrometry in tandem (MS/MS) analyzes using Matrix-assisted laser desorption/ionization with TOF/TOF is a tandem mass spectrometry method where two time-of-flight mass spectrometers are used consecutively (MALDI-TOF/TOF). A total of 51 differentially expressed proteins was identified when comparing the three observed conditions. Sixteen different spots were identified in the S0 stage compared with S2, while 28 different spots were found in S2 compared with S0. S1 expressed seven different spots compared with S0 and S2. CONCLUSIONS These findings suggest the involvement of several proteins directly related to the osteogenic pathway, which can be used to improve understanding of the osteogenic process.
Collapse
Affiliation(s)
- M S Giusta
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
42
|
Mrozik KM, Zilm PS, Bagley CJ, Hack S, Hoffmann P, Gronthos S, Bartold PM. Proteomic characterization of mesenchymal stem cell-like populations derived from ovine periodontal ligament, dental pulp, and bone marrow: analysis of differentially expressed proteins. Stem Cells Dev 2011; 19:1485-99. [PMID: 20050811 DOI: 10.1089/scd.2009.0446] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Postnatal mesenchymal stem/stromal-like cells (MSCs) including periodontal ligament stem cells (PDLSCs), dental pulp stem cells (DPSCs), and bone marrow stromal cells (BMSCs) are capable of self-renewal and differentiation into multiple mesenchymal cell lineages. Despite their similar expression of MSC-associated and osteoblastic markers, MSCs retain the capacity to generate structures resembling the microenvironments from which they are derived in vivo and represent a promising therapy for the regeneration of complex tissues in the clinical setting. With this in mind, systematic approaches are required to identify the differential protein expression patterns responsible for lineage commitment and mediating the formation of these complex structures. This is the first study to compare the differential proteomic expression profiles of ex vivo-expanded ovine PDLSCs, DPSCs, and BMSCs derived from an individual donor. The two-dimensional electrophoresis was performed and regulated proteins were identified by liquid chromatography--electrospray-ionization tandem mass spectrometry (MS and MS/MS), database searching, and de novo sequencing. In total, 58 proteins were differentially expressed between at least 2 MSC populations in both sheep, 12 of which were up-regulated in one MSC population relative to the other two. In addition, the regulation of selected proteins was also conserved between equivalent human MSC populations. We anticipate that differential protein expression profiling will provide a basis for elucidating the protein expression patterns and molecular cues that are crucial in specifying the characteristic growth and developmental capacity of dental and non-dental tissue-derived MSC populations. These expression patterns can serve as important tools for the regeneration of particular tissues in future stem cell-based tissue engineering studies using animal models.
Collapse
Affiliation(s)
- Krzysztof M Mrozik
- Colgate Australian Clinical Dental Research Centre, Dental School, The University of Adelaide, Adelaide, Australia.
| | | | | | | | | | | | | |
Collapse
|
43
|
Dütting S, Brachs S, Mielenz D. Fraternal twins: Swiprosin-1/EFhd2 and Swiprosin-2/EFhd1, two homologous EF-hand containing calcium binding adaptor proteins with distinct functions. Cell Commun Signal 2011; 9:2. [PMID: 21244694 PMCID: PMC3036668 DOI: 10.1186/1478-811x-9-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 01/18/2011] [Indexed: 11/10/2022] Open
Abstract
Changes in the intracellular calcium concentration govern cytoskeletal rearrangement, mitosis, apoptosis, transcriptional regulation or synaptic transmission, thereby, regulating cellular effector and organ functions. Calcium binding proteins respond to changes in the intracellular calcium concentration with structural changes, triggering enzymatic activation and association with downstream proteins. One type of calcium binding proteins are EF-hand super family proteins. Here, we describe two recently discovered homologous EF-hand containing adaptor proteins, Swiprosin-1/EF-hand domain containing 2 (EFhd2) and Swiprosin-2/EF-hand domain containing 1 (EFhd1), which are related to allograft inflammatory factor-1 (AIF-1). For reasons of simplicity and concision we propose to name Swiprosin-1/EFhd2 and Swiprosin-2/EFhd1 from now on EFhd2 and EFhd1, according to their respective gene symbols. AIF-1 and Swiprosin-1/EFhd2 are already present in Bilateria, for instance in Drosophila melanogaster and Caenhorhabditis elegans. Swiprosin-2/EFhd1 arose later from gene duplication in the tetrapodal lineage. Secondary structure prediction of AIF-1 reveals disordered regions and one functional EF-hand. Swiprosin-1/EFhd2 and Swiprosin-2/EFhd1 exhibit a disordered region at the N-terminus, followed by two EF-hands and a coiled-coil domain. Whereas both proteins are similar in their predicted overall structure they differ in a non-homologous stretch of 60 amino acids just in front of the EF-hands. AIF-1 controls calcium-dependent cytoskeletal rearrangement in innate immune cells by means of its functional EF-hand. We propose that Swiprosin-1/EFhd2 as well is a cytoskeleton associated adaptor protein involved in immune and brain cell function. Pro-inflammatory conditions are likely to modulate expression and function of Swiprosin-1/EFhd2. Swiprosin-2/EFhd1, on the other hand, modulates apoptosis and differentiation of neuronal and muscle precursor cells, probably through an association with mitochondria. We suggest furthermore that Swiprosin-2/EFhd1 is part of a cellular response to oxidative stress, which could explain its pro-survival activity in neuronal, muscle and perhaps some malignant tissues.
Collapse
Affiliation(s)
- Sebastian Dütting
- Division of Molecular Immunology, Department of Medicine III, Nikolaus Fiebiger Center, University of Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | | | | |
Collapse
|
44
|
Weng JY, Du X, Geng SX, Peng YW, Wang Z, Lu ZS, Wu SJ, Luo CW, Guo R, Ling W, Deng CX, Liao PJ, Xiang AP. Mesenchymal stem cell as salvage treatment for refractory chronic GVHD. Bone Marrow Transplant 2010; 45:1732-40. [PMID: 20818445 PMCID: PMC3035976 DOI: 10.1038/bmt.2010.195] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Refractory chronic GVHD (cGVHD) is an important complication after allogeneic hematopoietic SCT and is prognostic of poor outcome. MSCs are involved in tissue repair and modulating immune responses in vitro and in vivo. From April 2005 to October 2008, 19 patients with refractory cGVHD were treated with MSCs derived from the BM of volunteers. The median dose of MSCs was 0.6 × 10(6) cells per kg body weight. Fourteen of 19 patients (73.7%) responded well to MSCs, achieving a CR (n=4) or a PR (n=10). The immunosuppressive agent could be tapered to less than 50% of the starting dose in 5 of 14 surviving patients, and five patients could discontinue immunosuppressive agents. The median duration between MSC administration and immunosuppressive therapy discontinuation was 324 days (range, 200-550 days). No patients experienced adverse events during or immediately after MSC infusion. The 2-year survival rate was 77.7% in this study. Clinical improvement was accompanied by the increasing ratio of CD5+CD19+/CD5-CD19+ B cells and CD8+CD28-/CD8+CD28+ T cells. In conclusion, transfusion of MSCs expanded in vitro, irrespective of the donor, might be a safe and effective salvage therapy for patients with steroid-resistant, cGVHD.
Collapse
Affiliation(s)
- J Y Weng
- Department of Haematology, Guangdong General Hospital, Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, Guangdong, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kim S, Min WK, Chun S, Lee W, Chung HJ, Choi SJ, Yang SE, Yang YS, Yoo JI. Protein expression profiles during osteogenic differentiation of mesenchymal stem cells derived from human umbilical cord blood. TOHOKU J EXP MED 2010; 221:141-50. [PMID: 20495303 DOI: 10.1620/tjem.221.141] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mesenchymal stem cells (MSCs) can potentially differentiate along multiple lineages and be expanded in vitro, making them highly attractive candidates for cell therapy and tissue engineering applications. This study sought to investigate the critical proteins involved in osteogenic differentiation of mesenchymal stem cells derived from umbilical cord blood (UCB-MSCs). MSCs, which were isolated from three different preparations of human UCB, were osteoinduced, and total proteins were extracted from the cells. Two-dimensional polyacrylamide gel electrophoresis (2-D PAGE) was performed on the day (d) of induction d0, and on d2, d7, and d21 of differentiation. The optical density (OD) of each spot was measured, and spots with a mean OD of three cell lines of MSCs that increased > 30 or decreased < 0.1 relative to a previous time point were selected. Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry (MALDI-TOF/MS) was used to identify the proteins. Through database searches, the properties and functions of the proteins were investigated and then classified according to the Gene Ontology classification. Among the 308 spots observed in the 2-D gel, 16 proteins with a mean OD ratio > 30, and 20 proteins with a mean OD ratio < 0.1 were identified during the differentiation process. Additionally, the distribution of differentially expressed proteins according to cellular component and molecular function criteria differed depending on whether protein expression increased or decreased during differentiation. The results of this study will comprise an initial proteomic database for UCB-MSCs differentiation.
Collapse
Affiliation(s)
- Sollip Kim
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kilian KA, Bugarija B, Lahn BT, Mrksich M. Geometric cues for directing the differentiation of mesenchymal stem cells. Proc Natl Acad Sci U S A 2010; 107:4872-7. [PMID: 20194780 PMCID: PMC2841932 DOI: 10.1073/pnas.0903269107] [Citation(s) in RCA: 1355] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Significant efforts have been directed to understanding the factors that influence the lineage commitment of stem cells. This paper demonstrates that cell shape, independent of soluble factors, has a strong influence on the differentiation of human mesenchymal stem cells (MSCs) from bone marrow. When exposed to competing soluble differentiation signals, cells cultured in rectangles with increasing aspect ratio and in shapes with pentagonal symmetry but with different subcellular curvature-and with each occupying the same area-display different adipogenesis and osteogenesis profiles. The results reveal that geometric features that increase actomyosin contractility promote osteogenesis and are consistent with in vivo characteristics of the microenvironment of the differentiated cells. Cytoskeletal-disrupting pharmacological agents modulate shape-based trends in lineage commitment verifying the critical role of focal adhesion and myosin-generated contractility during differentiation. Microarray analysis and pathway inhibition studies suggest that contractile cells promote osteogenesis by enhancing c-Jun N-terminal kinase (JNK) and extracellular related kinase (ERK1/2) activation in conjunction with elevated wingless-type (Wnt) signaling. Taken together, this work points to the role that geometric shape cues can play in orchestrating the mechanochemical signals and paracrine/autocrine factors that can direct MSCs to appropriate fates.
Collapse
Affiliation(s)
- Kristopher A. Kilian
- Department of Chemistry
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637
| | - Branimir Bugarija
- Department of Human Genetics, and
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637
| | - Bruce T. Lahn
- Department of Human Genetics, and
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637
| | - Milan Mrksich
- Department of Chemistry
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
47
|
Ly L, Wasinger VC. Mass and charge selective protein fractionation for the differential analysis of T-cell and CD34+ stem cell proteins from cord blood. J Proteomics 2010; 73:571-8. [DOI: 10.1016/j.jprot.2009.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/11/2009] [Accepted: 09/02/2009] [Indexed: 12/11/2022]
|
48
|
D'Alessandro A, Liumbruno G, Grazzini G, Pupella S, Lombardini L, Zolla L. Umbilical cord blood stem cells: Towards a proteomic approach. J Proteomics 2010; 73:468-82. [DOI: 10.1016/j.jprot.2009.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/04/2009] [Accepted: 06/16/2009] [Indexed: 02/07/2023]
|
49
|
Anderson JM, Kushwaha M, Tambralli A, Bellis SL, Camata RP, Jun HW. Osteogenic differentiation of human mesenchymal stem cells directed by extracellular matrix-mimicking ligands in a biomimetic self-assembled peptide amphiphile nanomatrix. Biomacromolecules 2009; 10:2935-44. [PMID: 19746964 PMCID: PMC2760643 DOI: 10.1021/bm9007452] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study investigated the ability of nanoscale, biomimetic peptide amphiphile (PA) scaffolds inscribed with specific cellular adhesive ligands to direct the osteogenic differentiation of human mesenchymal stem cells (hMSCs) without osteogenic supplements. PA sequences were synthesized to mimic the native bone extracellular matrix (ECM), expressing different isolated ligands (i.e., RGDS, DGEA, KRSR). All PAs were presented as self-assembled two-dimensional coatings for the seeded hMSCs. Initial attachment results demonstrated that the different PAs could be individually recognized based on the incorporated adhesive ligands. Long-term studies assessed osteogenic differentiation up to 35 days. The RGDS-containing PA nanomatrix expressed significantly greater alkaline phosphatase activity, indicating the early promotion of osteogenic differentiation. A progressive shift toward osteogenic morphology and positive staining for mineral deposition provided further confirmation of the RGDS-containing PA nanomatrix. Overall, the PA nanomatrix clearly has great promise for directing the osteogenic differentiation of hMSCs without the aid of supplements by mimicking the native ECM, providing an adaptable environment that allows for different adhesive ligands to control cellular behaviors. This research model establishes the beginnings of a new versatile approach to regenerate bone tissues by closely following the principles of natural tissue formation.
Collapse
Affiliation(s)
- Joel M. Anderson
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Meenakshi Kushwaha
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Ajay Tambralli
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Susan L. Bellis
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233
- Department of Physiology & Biophysics, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Renato P. Camata
- Department of Physics, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233
| |
Collapse
|
50
|
Wu L, Wei X, Ling J, Liu L, Liu S, Li M, Xiao Y. Early osteogenic differential protein profile detected by proteomic analysis in human periodontal ligament cells. J Periodontal Res 2009; 44:645-56. [PMID: 19453858 DOI: 10.1111/j.1600-0765.2008.01174.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Human periodontal ligament cells play a pivotal role in maintaining periodontal ligament space, contain progenitors that are able to differentiate into cementoblasts/osteoblasts and have a tremendous potential to regenerate periodontal tissue. However, the exact molecular mechanisms governing the differentiation mechanisms of progenitors in periodontal ligament cells remain largely unknown. This study was carried out to investigate the differentially expressed proteins involved in the osteogenic differentiation of progenitors presented in periodontal ligament cells. MATERIAL AND METHODS Using two-dimensional gel electrophoresis, mass spectrometry and peptide mass fingerprinting, we analyzed the differential protein profiles of periodontal ligament cells undergoing mineralization. RESULTS Compared with undifferentiated periodontal ligament cells, 61 proteins in periodontal ligament cells undergoing differentiation showed at least a 1.5-fold change in intensity, of which 29 differentially expressed proteins were successfully identified by matrix-assisted laser-desorption ionization time-of-flight mass spectrometry. The expression of some of the identified proteins was further confirmed by western blotting and reverse transcription-polymerase chain reaction analysis. The identified proteins were cytoskeleton proteins and cytoskeleton-associated proteins, nuclear proteins and cell membrane-bound molecules. CONCLUSION Our results suggest that the proteins identified in this study may be associated with the unique function of periodontal ligament cells in maintaining periodontal tissue homeostasis, thus providing a comprehensive reference for understanding and investigating in greater detail the molecular mechanisms of periodontal ligament cells involved in periodontal regeneration.
Collapse
Affiliation(s)
- L Wu
- Department of Orthodontics, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong, China
| | | | | | | | | | | | | |
Collapse
|