1
|
Marolt Presen D, Goeschl V, Hanetseder D, Ogrin L, Stetco AL, Tansek A, Pozenel L, Bruszel B, Mitulovic G, Oesterreicher J, Zipperle J, Schaedl B, Holnthoner W, Grillari J, Redl H. Prolonged cultivation enhances the stimulatory activity of hiPSC mesenchymal progenitor-derived conditioned medium. Stem Cell Res Ther 2024; 15:434. [PMID: 39551765 PMCID: PMC11572509 DOI: 10.1186/s13287-024-03960-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/25/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cells represent a scalable source of youthful tissue progenitors and secretomes for regenerative therapies. The aim of our study was to investigate the potential of conditioned medium (CM) from hiPSC-mesenchymal progenitors (hiPSC-MPs) to stimulate osteogenic differentiation of human bone marrow-derived mesenchymal stromal cells (MSCs). We also investigated whether prolonged cultivation or osteogenic pre-differentiation of hiPSC-MPs could enhance the stimulatory activity of CM. METHODS MSCs were isolated from 13 donors (age 20-90 years). CM derived from hiPSC-MPs was added to the MSC cultures and the effects on proliferation and osteogenic differentiation were examined after 14 days and 6 weeks. The stimulatory activity of hiPSC-MP-CM was compared with the activity of MSC-derived CM and with the activity of CM prepared from hiPSC-MPs pre-cultured in growth or osteogenic medium for 14 days. Comparative proteomic analysis of CM was performed to gain insight into the molecular components responsible for the stimulatory activity. RESULTS Primary bone marrow-derived MSC exhibited variability, with a tendency towards lower proliferation and tri-lineage differentiation in older donors. hiPSC-MP-CM increased the proliferation and alkaline phosphatase activity of MSC from several adult/aged donors after 14 days of continuous supplementation under osteogenic conditions. However, CM supplementation failed to improve the mineralization of MSC pellets after 6 weeks under osteogenic conditions. hiPSC-MP-CM showed greater enhancement of proliferation and ALP activity than CM derived from bone marrow-derived MSCs. Moreover, 14-day cultivation but not osteogenic pre-differentiation of hiPSC-MPs strongly enhanced CM stimulatory activity. Quantitative proteomic analysis of d14-CM revealed a distinct profile of components that formed a highly interconnected associations network with two clusters, one functionally associated with binding and organization of actin/cytoskeletal components and the other with structural constituents of the extracellular matrix, collagen, and growth factor binding. Several hub proteins were identified that were reported to have functions in cell-extracellular matrix interaction, osteogenic differentiation and development. CONCLUSIONS Our data show that hiPSC-MP-CM enhances early osteogenic differentiation of human bone marrow-derived MSCs and that prolonged cultivation of hiPSC-MPs enhances CM-stimulatory activity. Proteomic analysis of the upregulated protein components provides the basis for further optimization of hiPSC-MP-CM for bone regenerative therapies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria.
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria.
| | - Vanessa Goeschl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Dominik Hanetseder
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Laura Ogrin
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Alexandra-Larissa Stetco
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Anja Tansek
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Laura Pozenel
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Bella Bruszel
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, 3400, Austria
| | - Goran Mitulovic
- Clinical Department of Laboratory Medicine Proteomics Core Facility, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
- Bruker Austria, Lemböckgasse 47b, Vienna, 1230, Austria
| | - Johannes Oesterreicher
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Johannes Zipperle
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
- University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna, 1090, Austria
| | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Gregor-Mendel-Straße 33, Vienna, 1180, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstrasse 13, Vienna, A-1200, Austria
- Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, Vienna, 1200, Austria
| |
Collapse
|
2
|
Tee CA, Roxby DN, Othman R, Denslin V, Bhat KS, Yang Z, Han J, Tucker-Kellogg L, Boyer LA. Metabolic modulation to improve MSC expansion and therapeutic potential for articular cartilage repair. Stem Cell Res Ther 2024; 15:308. [PMID: 39285485 PMCID: PMC11406821 DOI: 10.1186/s13287-024-03923-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Articular cartilage degeneration can result from injury, age, or arthritis, causing significant joint pain and disability without surgical intervention. Currently, the only FDA cell-based therapy for articular cartilage injury is Autologous Chondrocyte Implantation (ACI); however, this procedure is costly, time-intensive, and requires multiple treatments. Mesenchymal stromal cells (MSCs) are an attractive alternative autologous therapy due to their availability and ability to robustly differentiate into chondrocytes for transplantation with good safety profiles. However, treatment outcomes are variable due to donor-to-donor variability as well as intrapopulation heterogeneity and unstandardized MSC manufacturing protocols. Process improvements that reduce cell heterogeneity while increasing donor cell numbers with improved chondrogenic potential during expansion culture are needed to realize the full potential of MSC therapy. METHODS In this study, we investigated the potential of MSC metabolic modulation during expansion to enhance their chondrogenic commitment by varying the nutrient composition, including glucose, pyruvate, glutamine, and ascorbic acid in culture media. We tested the effect of metabolic modulation in short-term (one passage) and long-term (up to seven passages). We measured metabolic state, cell size, population doubling time, and senescence and employed novel tools including micro-magnetic resonance relaxometry (µMRR) relaxation time (T2) to characterize the effects of AA on improved MSC expansion and chondrogenic potential. RESULTS Our data show that the addition of 1 mM L-ascorbic acid-2-phosphate (AA) to cultures for one passage during MSC expansion prior to initiation of differentiation improves chondrogenic differentiation. We further demonstrate that AA treatment reduced the proportion of senescent cells and cell heterogeneity also allowing for long-term expansion that led to a > 300-fold increase in yield of MSCs with enhanced chondrogenic potential compared to untreated cells. AA-treated MSCs with improved chondrogenic potential showed a robust shift in metabolic profile to OXPHOS and higher µMRR T2 values, identifying critical quality attributes that could be implemented in MSC manufacturing for articular cartilage repair. CONCLUSIONS Our results suggest an improved MSC manufacturing process that can enhance chondrogenic potential by targeting MSC metabolism and integrating process analytic tools during expansion.
Collapse
Affiliation(s)
- Ching Ann Tee
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Daniel Ninio Roxby
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Rashidah Othman
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Vinitha Denslin
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore, 117510, Republic of Singapore
| | - Kiesar Sideeq Bhat
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- Department of Bioresources, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Zheng Yang
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore, 117510, Republic of Singapore
- Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block 11, Singapore, 119288, Republic of Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 50 Vassar St, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Lisa Tucker-Kellogg
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore.
- Cancer and Stem Cell Biology and Centre for Computational Biology, Duke-NUS Medical School, 8 College Rd, Singapore, 169857, Republic of Singapore.
| | - Laurie A Boyer
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
3
|
Jeyaraman M, Jeyaraman N, Ramasubramanian S, Ranjan R, Jha SK, Gupta A. Bone Marrow Aspirate Concentrate for Treatment of Primary Knee Osteoarthritis: A Prospective, Single-Center, Non-randomized Study with 2-Year Follow-Up. Indian J Orthop 2024; 58:894-904. [PMID: 38948370 PMCID: PMC11208343 DOI: 10.1007/s43465-024-01168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/23/2024] [Indexed: 07/02/2024]
Abstract
Introduction Knee osteoarthritis (OA) is a widespread, disabling condition with no intervention to fully restore cartilage or halt progression. Bone marrow aspirate concentrate (BMAC), an autologous product from bone marrow aspiration, has shown promise as a regenerative therapy due to its cell composition and chondrogenic effects. Our study aims to assess the functional outcomes, including pain, function, satisfaction, and complications post-BMAC injection in knee OA patients. Materials and Methods In this prospective, single-center study, 63 patients with grade II-III knee OA (Kellgren-Lawrence (K-L) scale) unresponsive to conservative management underwent BMAC injection. The procedure involved bone marrow aspiration from the anterior iliac crest, processing to obtain a concentrate, followed by intra-articular injection. Patients were followed for 24 months, assessing outcomes using the Visual Analog Scale (VAS), International Knee Documentation Committee (IKDC) score, and MOCART 2.0 score. Results The cohort, with a slight female predominance and predominantly aged 41-50 years, majorly comprised K-L grade III OA patients. BMAC treatment resulted in significant improvements in VAS pain scores, IKDC functional scores, and MOCART 2.0 scores over the 24-month follow-up. Conclusion BMAC injection provides significant improvement in both pain and functional outcomes at mid-term follow-up in patients with mild-to-moderate OA of the knee. Further high-quality, adequately powered, multi-center, prospective, double-blinded, randomized controlled trials with longer follow-up are necessary to justify the routine clinical use of BMAC for treatment of patients suffering with knee OA. Graphical Abstract
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077 India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India
- Clinical Research Associate, Virginia Tech India, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600095 India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077 India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai, Tamil Nadu 600002 India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306 India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India
- Department of Zoology, Kalindi College, University of Delhi, New Delhi, 110008 India
| | - Ashim Gupta
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045 USA
- Regenerative Orthopaedics, Noida, Uttar Pradesh 201301 India
- Future Biologics, Lawrenceville, GA 30043 USA
- BioIntegrate, Lawrenceville, GA 30043 USA
| |
Collapse
|
4
|
Wang Y, Li HY, Guan SY, Yu SH, Zhou YC, Zheng LW, Zhang J. Different Sources of Bone Marrow Mesenchymal Stem Cells: A Comparison of Subchondral, Mandibular, and Tibia Bone-derived Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:1029-1041. [PMID: 37937557 DOI: 10.2174/011574888x260686231023091127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/04/2023] [Accepted: 09/01/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Stem cell properties vary considerably based on the source and tissue site of mesenchymal stem cells (MSCs). The mandibular condyle is a unique kind of craniofacial bone with a special structure and a relatively high remodeling rate. MSCs here may also be unique to address specific physical needs. OBJECTIVE The aim of this study was to compare the proliferation and multidirectional differentiation potential among MSCs derived from the tibia (TMSCs), mandibular ramus marrow (MMSCs), and condylar subchondral bone (SMSCs) of rats in vitro. METHODS Cell proliferation and migration were assessed by CCK-8, laser confocal, and cell scratch assays. Histochemical staining and real-time PCR were used to evaluate the multidirectional differentiation potential and DNA methylation and histone deacetylation levels. RESULTS The proliferation rate and self-renewal capacity of SMSCs were significantly higher than those of MMSCs and TMSCs. Moreover, SMSCs possessed significantly higher mineralization and osteogenic differentiation potential. Dnmt2, Dnmt3b, Hdac6, Hdac7, Hdac9, and Hdac10 may be instrumental in the osteogenesis of SMSCs. In addition, SMSCs are distinct from MMSCs and TMSCs with lower adipogenic differentiation and chondrogenic differentiation potential. The multidirectional differentiation capacities of TMSCs were exactly the opposite of those of SMSCs, and the results of MMSCs were intermediate. CONCLUSION This research offers a new paradigm in which SMSCs could be a useful source of stem cells for further application in stem cell-based medical therapies due to their strong cell renewal and osteogenic capacity.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Yu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shu-Yuan Guan
- Department of Stomatology, Medical College, Dalian University, Dalian, 116622, Liaoning, China
| | - Si-Han Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Chuan Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li-Wei Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Zhang
- Yunnan Key Laboratory of Stomatology, Kunming Medical University School and Hospital of Stomatology, Kunming, China
| |
Collapse
|
5
|
Li J, Wu Z, Zhao L, Liu Y, Su Y, Gong X, Liu F, Zhang L. The heterogeneity of mesenchymal stem cells: an important issue to be addressed in cell therapy. Stem Cell Res Ther 2023; 14:381. [PMID: 38124129 PMCID: PMC10734083 DOI: 10.1186/s13287-023-03587-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
With the continuous improvement of human technology, the medical field has gradually moved from molecular therapy to cellular therapy. As a safe and effective therapeutic tool, cell therapy has successfully created a research boom in the modern medical field. Mesenchymal stem cells (MSCs) are derived from early mesoderm and have high self-renewal and multidirectional differentiation ability, and have become one of the important cores of cell therapy research by virtue of their immunomodulatory and tissue repair capabilities. In recent years, the application of MSCs in various diseases has received widespread attention, but there are still various problems in the treatment of MSCs, among which the heterogeneity of MSCs may be one of the causes of the problem. In this paper, we review the correlation of MSCs heterogeneity to provide a basis for further reduction of MSCs heterogeneity and standardization of MSCs and hope to provide a reference for cell therapy.
Collapse
Affiliation(s)
- Jingxuan Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Li Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030600, China
| | - Yang Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yazhen Su
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xueyan Gong
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Fancheng Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
6
|
Handke M, Rakow A, Singer D, Miebach L, Schulze F, Bekeschus S, Schoon J, Wassilew GI. Bone marrow from periacetabular osteotomies as a novel source for human mesenchymal stromal cells. Stem Cell Res Ther 2023; 14:315. [PMID: 37924114 PMCID: PMC10625289 DOI: 10.1186/s13287-023-03552-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/27/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are used in regenerative medicine and related research involving immunomodulatory, anti-inflammatory, anti-fibrotic and regenerative functions. Isolation of BM-MSCs from samples obtained during total hip arthroplasty (THA) is routinely possible. Advanced age and comorbidities of the majority of patients undergoing THA limit their applicability. Our study aimed to evaluate the potential of bone marrow obtained during periacetabular osteotomy (PAO) as a novel source of BM-MSCs from young donors by analyzing cell yield and cell characteristics. METHODS Bone samples were obtained from the anterior Os ilium or superior Os pubis during PAO and from the femoral cavity during primary THA. Isolation of bone marrow-derived mononuclear cells (BM-MNCs) was performed by density gradient centrifugation. The samples from PAO and THA patients were compared in terms of BM-MSC yield, colony formation and the proportion of BM-MSCs within the BM-MNC population using flow cytometry analysis. The cells were characterized based on the expression of BM-MSC-specific surface markers. The functionality of the cells was compared by quantifying post-thaw viability, metabolic activity, proliferation capacity, senescence-associated beta galactosidase (SA-β-gal) expression, trilineage differentiation potential and major secretome proteins. RESULTS Isolation of BM-MNCs was possible in a reliable and reproducible manner when using bone from PAO containing more than 0.24 g bone marrow. PAO patients were younger than patients of the THA group. Bone obtained during PAO contained less bone marrow and led to a lower BM-MSC number after the first cell culture passage compared to BM-MSCs obtained during THA. BM-MSCs from PAO samples are characterized by a higher proliferation capacity. This results in a higher yield in cell culture passage two, when normalized to the sample weight. BM-MSCs from PAO patients showed increased secretion of TGF-β1, TIMP2, and VEGF upon osteogenic differentiation. BM-MSCs from PAO and THA patients revealed similar results regarding the onset of SA-β-gal expression and trilineage differentiation capacity. CONCLUSIONS We suggest that bone obtained during PAO is a promising novel source for BM-MSCs from young donors. Limited absolute cell yield due to low sample weight must be considered in early cell culture passages and might be critical for the range of clinical applications possible for BM-MSCs from this source. The higher proliferation capacity and increased growth factor secretion of BM-MSCs from young donors may be beneficial for future regenerative cell therapies, in vitro models, and tissue engineering.
Collapse
Affiliation(s)
- Maximilian Handke
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Anastasia Rakow
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Debora Singer
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix- Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Lea Miebach
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix- Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Frank Schulze
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix- Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Janosch Schoon
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475, Greifswald, Germany.
| | - Georgi I Wassilew
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, 17475, Greifswald, Germany.
| |
Collapse
|
7
|
Tsitlakidis S, Hohenbild F, Saur M, Moghaddam A, Kunisch E, Renkawitz T, Gonzalo de Juan I, Westhauser F. Reduced Sodium Portions Favor Osteogenic Properties and Cytocompatibility of 45S5-Based Bioactive Glass Particles. Biomimetics (Basel) 2023; 8:472. [PMID: 37887603 PMCID: PMC10604502 DOI: 10.3390/biomimetics8060472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
Besides its favorable biological properties, the release of sodium (Na) from the well-known 45S5-bioactive glass (BG) composition (in mol%: 46.1, SiO2, 24.5 CaO, 24.5 Na2O, 6.0 P2O5) can hamper its cytocompatibility. In this study, particles of Na-reduced variants of 45S5-BG were produced in exchange for CaO and P2O5 via the sol-gel-route resulting in Na contents of 75%, 50%, 25% or 0% of the original composition. The release of ions from the BGs as well as their impact on the cell environment (pH values), viability and osteogenic differentiation (activity of alkaline phosphatase (ALP)), the expression of osteopontin and osteocalcin in human bone-marrow-derived mesenchymal stromal cells in correlation to the Na-content and ion release of the BGs was assessed. The release of Na-ions increased with increasing Na-content in the BGs. With decreasing Na content, the viability of cells incubated with the BGs increased. The Na-reduced BGs showed elevated ALP activity and a pro-osteogenic stimulation with accelerated osteopontin induction and a pronounced upregulation of osteocalcin. In conclusion, the reduction in Na-content enhances the cytocompatibility and improves the osteogenic properties of 45S5-BG, making the Na-reduced variants of 45S5-BG promising candidates for further experimental consideration.
Collapse
Affiliation(s)
- Stefanos Tsitlakidis
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (S.T.); (M.S.); (E.K.)
| | - Frederike Hohenbild
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (S.T.); (M.S.); (E.K.)
| | - Merve Saur
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (S.T.); (M.S.); (E.K.)
| | - Arash Moghaddam
- PrivatÄrztliches Zentrum Aschaffenburg, Frohsinnstraße 12, 63739 Aschaffenburg, Germany;
| | - Elke Kunisch
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (S.T.); (M.S.); (E.K.)
| | - Tobias Renkawitz
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (S.T.); (M.S.); (E.K.)
| | - Isabel Gonzalo de Juan
- Institut für Materialwissenschaft, Technische Universität Darmstadt, Otto-Berndt-Straße 3, 64287 Darmstadt, Germany
| | - Fabian Westhauser
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (S.T.); (M.S.); (E.K.)
| |
Collapse
|
8
|
Ouzin M, Kogler G. Mesenchymal Stromal Cells: Heterogeneity and Therapeutical Applications. Cells 2023; 12:2039. [PMID: 37626848 PMCID: PMC10453316 DOI: 10.3390/cells12162039] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stromal cells nowadays emerge as a major player in the field of regenerative medicine and translational research. They constitute, with their derived products, the most frequently used cell type in different therapies. However, their heterogeneity, including different subpopulations, the anatomic source of isolation, and high donor-to-donor variability, constitutes a major controversial issue that affects their use in clinical applications. Furthermore, the intrinsic and extrinsic molecular mechanisms underlying their self-renewal and fate specification are still not completely elucidated. This review dissects the different heterogeneity aspects of the tissue source associated with a distinct developmental origin that need to be considered when generating homogenous products before their usage for clinical applications.
Collapse
Affiliation(s)
- Meryem Ouzin
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Hospital Düsseldorf, 40225 Düsseldorf, Germany;
| | | |
Collapse
|
9
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
10
|
Cell surface markers for mesenchymal stem cells related to the skeletal system: A scoping review. Heliyon 2023; 9:e13464. [PMID: 36865479 PMCID: PMC9970931 DOI: 10.1016/j.heliyon.2023.e13464] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) have been described as bone marrow stromal cells, which can form cartilage, bone or hematopoietic supportive stroma. In 2006, the International Society for Cell Therapy (ISCT) established a set of minimal characteristics to define MSCs. According to their criteria, these cells must express CD73, CD90 and CD105 surface markers; however, it is now known they do not represent true stemness epitopes. The objective of the present work was to determine the surface markers for human MSCs associated with skeletal tissue reported in the literature (1994-2021). To this end, we performed a scoping review for hMSCs in axial and appendicular skeleton. Our findings determined the most widely used markers were CD105 (82.9%), CD90 (75.0%) and CD73 (52.0%) for studies performed in vitro as proposed by the ISCT, followed by CD44 (42.1%), CD166 (30.9%), CD29 (27.6%), STRO-1 (17.7%), CD146 (15.1%) and CD271 (7.9%) in bone marrow and cartilage. On the other hand, only 4% of the articles evaluated in situ cell surface markers. Even though most studies use the ISCT criteria, most publications in adult tissues don't evaluate the characteristics that establish a stem cell (self-renewal and differentiation), which will be necessary to distinguish between a stem cell and progenitor populations. Collectively, MSCs require further understanding of their characteristics if they are intended for clinical use.
Collapse
|
11
|
Bone Marrow-Derived Mesenchymal Stem Cell Implants for the Treatment of Focal Chondral Defects of the Knee in Animal Models: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24043227. [PMID: 36834639 PMCID: PMC9958893 DOI: 10.3390/ijms24043227] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis remains an unfortunate long-term consequence of focal cartilage defects of the knee. Associated with functional loss and pain, it has necessitated the exploration of new therapies to regenerate cartilage before significant deterioration and subsequent joint replacement take place. Recent studies have investigated a multitude of mesenchymal stem cell (MSC) sources and polymer scaffold compositions. It is uncertain how different combinations affect the extent of integration of native and implant cartilage and the quality of new cartilage formed. Implants seeded with bone marrow-derived MSCs (BMSCs) have demonstrated promising results in restoring these defects, largely through in vitro and animal studies. A PRISMA systematic review and meta-analysis was conducted using five databases (PubMed, MEDLINE, EMBASE, Web of Science, and CINAHL) to identify studies using BMSC-seeded implants in animal models of focal cartilage defects of the knee. Quantitative results from the histological assessment of integration quality were extracted. Repair cartilage morphology and staining characteristics were also recorded. Meta-analysis demonstrated that high-quality integration was achieved, exceeding that of cell-free comparators and control groups. This was associated with repair tissue morphology and staining properties which resembled those of native cartilage. Subgroup analysis showed better integration outcomes for studies using poly-glycolic acid-based scaffolds. In conclusion, BMSC-seeded implants represent promising strategies for the advancement of focal cartilage defect repair. While a greater number of studies treating human patients is necessary to realize the full clinical potential of BMSC therapy, high-quality integration scores suggest that these implants could generate repair cartilage of substantial longevity.
Collapse
|
12
|
Shanto PC, Park S, Park M, Lee BT. Physico-biological evaluation of 3D printed dECM/TOCN/alginate hydrogel based scaffolds for cartilage tissue regeneration. BIOMATERIALS ADVANCES 2023; 145:213239. [PMID: 36542879 DOI: 10.1016/j.bioadv.2022.213239] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/23/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Cartilage damage is the leading cause of osteoarthritis (OA), especially in an aging society. Mimicking the native cartilage microenvironment for chondrogenic differentiation along with constructing a stable and controlled architectural scaffold is considerably challenging. In this study, three-dimensional (3D) printed scaffolds using tempo-oxidized cellulose nanofiber (TOCN), decellularized extracellular matrix (dECM), and sodium alginate (SA) were fabricated for cartilage tissue regeneration. We prepared three groups (dECM80, dECM50, dECM20) of 3D printable hydrogels with different ratios of TOCN and dECM where SA concentration remained the same. Two-step crosslinking was performed with CaCl2 solution to achieve the highly stable 3D printed scaffolds. Finally, the fundamental physical characterizations showed that increasing the ratio of TOCN with dECM significantly improved the viscoelastic behaviour, stability, mechanical properties, and printability of the scaffolds. Based on the results, the 3D printed dECM50 scaffolds with controlled and identical pore sizes increased the whole-layer integrity and nutrient supply in each layer of the scaffold. Furthermore, evaluation of in vitro and in vivo biocompatibility of the scaffolds with rBMSCs indicated that dECM50 scaffolds provided a suitable microenvironment for cell proliferation and promoted chondrogenesis by remarkably expressing the cartilage-specific markers. This study demonstrates that 3D printed dECM50 scaffolds provide a favourable and promising microenvironment for cartilage tissue regeneration.
Collapse
Affiliation(s)
- Prayas Chakma Shanto
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Seongsu Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea; Institute of Tissue Regeneration, Soonchunhyang University, Cheonan 31151, Republic of Korea.
| |
Collapse
|
13
|
Haeusner S, Jauković A, Kupczyk E, Herrmann M. Review: cellularity in bone marrow autografts for bone and fracture healing. Am J Physiol Cell Physiol 2023; 324:C517-C531. [PMID: 36622067 DOI: 10.1152/ajpcell.00482.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of autografts, as primary cell and tissue source, is the current gold standard approach to treat critical size bone defects and nonunion defects. The unique mixture of the autografts, containing bony compartments and bone marrow (BM), delivers promising results. Although BM mesenchymal stromal cells (BM-MSCs) still represent a major target for various healing approaches in current preclinical research and respective clinical trials, their occurrence in the human BM is typically low. In vitro expansion of this cell type is regulatory challenging as well as time and cost intensive. Compared with marginal percentages of resident BM-MSCs in BM, BM mononuclear cells (BM-MNCs) contained in BM aspirates, concentrates, and bone autografts represent a readily available abundant cell source, applicable within hours during surgical procedures without the need for time-consuming and regulatory challenging cell expansion. This benefit is one reason why autografting has become a clinical standard procedure. However, the exact anatomy and cellularity of BM-MNCs in humans, which is strongly correlated to their unique mode of action and wide application range remains to be elucidated. The aim of this review was to present an overview of the current knowledge on these specific cell types found in human BM, emphasize the contribution of BM-MNCs in bone healing, highlight donor site dependence, and discuss limitations in the current isolation and subsequent characterization procedures. Hereby, the most recent and relevant examples of human BM-MNC cell characterization, flow cytometric analyses, and findings are summarized, with a strong focus on bone therapy.
Collapse
Affiliation(s)
- S Haeusner
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital of Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - A Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - E Kupczyk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - M Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital of Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
14
|
Pacheco-Vergara MJ, Ricci JL, Mijares D, Bromage TG, Rabieh S, Coelho PG, Witek L. 3D printed mesoporous bioactive glass, bioglass 45S5, and β-TCP scaffolds for regenerative medicine: A comparative in vitro study. Biomed Mater Eng 2023; 34:439-458. [PMID: 36744331 DOI: 10.3233/bme-222524] [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: 02/04/2023]
Abstract
BACKGROUND While autografts to date remain the "gold standard" for bone void fillers, synthetic bone grafts have garnered attention due to their favorable advantages such as ability to be tailored in terms of their physical and chemical properties. Bioactive glass (BG), an inorganic material, has the capacity to form a strong bond with bone by forming a bone-like apatite surface, enhancing osteogenesis. Coupled with additive manufacturing (3D printing) it is possible to maximize bone regenerative properties of the BG. OBJECTIVE The objective of this study was to synthesize and characterize 3D printed mesoporous bioactive glass (MBG), BG 45S5, and compare to β-Tricalcium phosphate (β-TCP) based scaffolds; test cell viability and osteogenic differentiation on human osteoprogenitor cells in vitro. METHODS MBG, BG 45S5, and β-TCP were fabricated into colloidal gel suspensions, tested with a rheometer, and manufactured into scaffolds using a 3D direct-write micro-printer. The materials were characterized in terms of microstructure and composition with Thermogravimetric Analyzer/Differential Scanning Calorimeter (TGA/DSC), Fourier Transform Infrared Spectroscopy (FTIR), X-ray Diffraction (XRD), Micro-Computed Tomography (μ-CT), Scanning Electron Microscopy (SEM), Energy Dispersive X-ray Spectroscopy (EDS), and Mattauch-Herzog-Inductively Coupled Plasma-Mass Spectrometry (MH-ICP-MS). RESULTS Scaffolds were tested for cell proliferation and osteogenic differentiation using human osteoprogenitor cells. Osteogenic media was used for differentiation, and immunocytochemistry for osteogenic markers Runx-2, Collagen-I, and Osteocalcin. The cell viability results after 7 days of culture yielded significantly higher (p < 0.05) results in β-TCP scaffolds compared to BG 45S5 and MBG groups. CONCLUSION All materials expressed osteogenic markers after 21 days of culture in expansion and osteogenic media.
Collapse
Affiliation(s)
| | - John L Ricci
- Biomaterials Division, New York University College of Dentistry, New York, NY, USA
| | - Dindo Mijares
- Biomaterials Division, New York University College of Dentistry, New York, NY, USA
| | - Timothy G Bromage
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Sasan Rabieh
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Paulo G Coelho
- Division of Plastic Surgery, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lukasz Witek
- Biomaterials Division, New York University College of Dentistry, New York, NY, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, USA
| |
Collapse
|
15
|
Fibrin glue does not promote migration and proliferation of bone marrow derived mesenchymal stem cells in collagenic membranes: an in vitro study. Sci Rep 2022; 12:20660. [PMID: 36450814 PMCID: PMC9712600 DOI: 10.1038/s41598-022-25203-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
During Autologous Matrix-Induced Chondrogenesis (AMIC), the membrane is often glued into the chondral defect. However, whether fibrin glue influences cells proliferation and migration remain unclear. This study evaluated the impact of fibrin glue addition to biologic membranes loaded with bone marrow-derived mesenchymal stem cells (B-MSCs). A porcine derived collagen membrane (Cartimaix, Matricel GmbH, Germany) was used. B-MSCs were harvested from three different unrelated donors. The membranes were embedded in mounting medium with DAPI (ABCAM, Cambridge, UK) and analysed at 1-, 2-, 3-, 4-, 6-, and at 8-week follow-up. The DAPI ties the DNA of the cell nucleus, emitting blue fluorescence. DAPI/nuclei signals were analysed with fluorescence microscopy at 100-fold magnification. The group without fibrin glue demonstrated greater migration of the B-MSCs within the membrane at week 4 (P < 0.001), 6 (P < 0.001), and 8 (P < 0.001). No difference was found at week 1, 2, and 3. The group without fibrin glue demonstrated greater proliferation of B-MSCs within the membrane. These differences were significant at week 1 (P = 0.02), 2 (P = 0.008), 3 (P = 0.0009), 4 (P < 0.0001), 6 (P < 0.0001), 8 (P < 0.0001). Concluding, in the present setting, the use of fibrin in a collagenic biomembrane impairs B-MSCs proliferation and migration in vitro.
Collapse
|
16
|
Trivanovic D, Harder J, Leucht M, Kreuzahler T, Schlierf B, Holzapfel BM, Rudert M, Jakob F, Herrmann M. Immune and stem cell compartments of acetabular and femoral bone marrow in hip osteoarthritis patients. Osteoarthritis Cartilage 2022; 30:1116-1129. [PMID: 35569800 DOI: 10.1016/j.joca.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Hip osteoarthritis (OA) affects all components of the osteochondral unit, leading to bone marrow (BM) lesions, and unknown consequences on BM cell functionality. We analyzed the cellular composition in OA-affected acetabula compared to proximal femur shafts obtained of hip OA patients to reveal yet not explored immune and stem cell compartments. DESIGN Combining flow cytometry, cellular assays and transcription analyses, we performed extensive ex vivo phenotyping of acetabular BM cells from 18 hip OA patients, comparing them with their counterparts from patient-matched femoral shaft BM samples. Findings were related to differences in skeletal sites and age. RESULTS Acetabular BM had a greater frequency of T-lymphocytes, non-hematopoietic cells and colony-forming units fibroblastic potential than femoral BM. The incidence of acetabular CD45+CD3+ T-lymphocytes increased (95% CI: 0.1770 to 0.0.8416), while clonogenic hematopoietic progenitors declined (95% CI: -0.9023 to -0.2399) with age of patients. On the other side, in femoral BM, we observed higher B-lymphocyte, myeloid and erythroid cell frequencies. Acetabular mesenchymal stromal cells (MSCs) showed a senescent profile associated with the expression of survival and inflammation-related genes. Efficient osteogenic and chondrogenic differentiation was detected in acetabular MSCs, while adipogenesis was more pronounced in their femoral counterparts. CONCLUSION Our results suggest that distinctions in BM cellular compartments and MSCs may be due to the influence of the OA-stressed microenvironment, but also acetabular vs femoral shaft-specific peculiarities cannot be excluded. These results bring new knowledge on acetabular BM cell populations and may be addressed as novel pathogenic mechanisms and therapeutic targets in OA.
Collapse
Affiliation(s)
- D Trivanovic
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - J Harder
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - M Leucht
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - T Kreuzahler
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - B Schlierf
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - B M Holzapfel
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Department of Orthopaedic Surgery, König-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Department of Orthopaedic Surgery, University Clinics, Ludwig-Maximilians University Munich, Munich, 81377, Germany
| | - M Rudert
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Department of Orthopaedic Surgery, König-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - F Jakob
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - M Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany.
| |
Collapse
|
17
|
Tan L, Liu X, Dou H, Hou Y. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment — specific factors involved in the regulation of MSC plasticity. Genes Dis 2022; 9:296-309. [PMID: 35224147 PMCID: PMC8843883 DOI: 10.1016/j.gendis.2020.10.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotent stromal cells, have attracted extensive attention in the field of regenerative medicine and cell therapy due to the capacity of self-renewal, multilineage differentiation, and immune regulation. MSCs have different cellular effects in different diseases, and even have markedly different curative effects with different tissue sources, indicating the plasticity of MSCs. The phenotypes, secreted factors, and proliferative, migratory, differentiating, and immunomodulatory effects of MSCs depend on certain mediators present in their microenvironment. Understanding microenvironmental factors and their internal mechanisms in MSC responses may help in subsequent prediction and improvement of clinical benefits. This review highlighted the recent advances in MSC plasticity in the physiological and pathological microenvironment and multiple microenvironmental factors regulating MSC plasticity. It also highlighted some progress in the underlying molecular mechanisms of MSC remodeling in the microenvironment. It might provide references for the improvement in vitro culture of MSCs, clinical application, and in vivo induction.
Collapse
|
18
|
Kanawa M, Igarashi A, Fujimoto K, Saskianti T, Nakashima A, Higashi Y, Kurihara H, Kato Y, Kawamoto T. The Identification of Marker Genes for Predicting the Osteogenic Differentiation Potential of Mesenchymal Stromal Cells. Curr Issues Mol Biol 2021; 43:2157-2166. [PMID: 34940124 PMCID: PMC8929155 DOI: 10.3390/cimb43030150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have the potential to differentiate into a variety of mature cell types and are a promising source of regenerative medicine. The success of regenerative medicine using MSCs strongly depends on their differentiation potential. In this study, we sought to identify marker genes for predicting the osteogenic differentiation potential by comparing ilium MSC and fibroblast samples. We measured the mRNA levels of 95 candidate genes in nine ilium MSC and four fibroblast samples before osteogenic induction, and compared them with alkaline phosphatase (ALP) activity as a marker of osteogenic differentiation after induction. We identified 17 genes whose mRNA expression levels positively correlated with ALP activity. The chondrogenic and adipogenic differentiation potentials of jaw MSCs are much lower than those of ilium MSCs, although the osteogenic differentiation potential of jaw MSCs is comparable with that of ilium MSCs. To select markers suitable for predicting the osteogenic differentiation potential, we compared the mRNA levels of the 17 genes in ilium MSCs with those in jaw MSCs. The levels of 7 out of the 17 genes were not substantially different between the jaw and ilium MSCs, while the remaining 10 genes were expressed at significantly lower levels in jaw MSCs than in ilium MSCs. The mRNA levels of the seven similarly expressed genes were also compared with those in fibroblasts, which have little or no osteogenic differentiation potential. Among the seven genes, the mRNA levels of IGF1 and SRGN in all MSCs examined were higher than those in any of the fibroblasts. These results suggest that measuring the mRNA levels of IGF1 and SRGN before osteogenic induction will provide useful information for selecting competent MSCs for regenerative medicine, although the effectiveness of the markers is needed to be confirmed using a large number of MSCs, which have various levels of osteogenic differentiation potential.
Collapse
Affiliation(s)
- Masami Kanawa
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima 734-8533, Japan;
| | - Akira Igarashi
- Division of Advanced Technology and Development, BML, Inc., Saitama 350-1101, Japan;
- Department of Dental and Medical Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan; (K.F.); (T.S.); (Y.K.)
| | - Katsumi Fujimoto
- Department of Dental and Medical Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan; (K.F.); (T.S.); (Y.K.)
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan
| | - Tania Saskianti
- Department of Dental and Medical Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan; (K.F.); (T.S.); (Y.K.)
- Department of Pediatric Dentistry, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan;
| | - Yukihito Higashi
- Research Center for Radiation Genome Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8533, Japan;
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan;
| | - Yukio Kato
- Department of Dental and Medical Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan; (K.F.); (T.S.); (Y.K.)
| | - Takeshi Kawamoto
- Department of Dental and Medical Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8533, Japan; (K.F.); (T.S.); (Y.K.)
- Writing Center, Hiroshima University, Higashi-Hiroshima 739-8512, Japan
- Correspondence: ; Tel.: +81-82-424-6207
| |
Collapse
|
19
|
Fosca M, Basoli V, Della Bella E, Russo F, Vadala G, Alini M, Rau JV, Verrier S. Raman spectroscopy in skeletal tissue disorders and tissue engineering: present and prospective. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:949-965. [PMID: 34579558 DOI: 10.1089/ten.teb.2021.0139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Musculoskeletal disorders are the most common reason of chronic pain and disability representing worldwide an enormous socio-economic burden. In this review, new biomedical application fields for Raman spectroscopy (RS) technique related to skeletal tissues are discussed showing that it can provide a comprehensive profile of tissue composition in situ, in a rapid, label-free, and non-destructive manner. RS can be used as a tool to study tissue alterations associated to aging, pathologies, and disease treatments. The main advantage with respect to currently applied methods in clinics is its ability to provide specific information on molecular composition, which goes beyond other diagnostic tools. Being compatible with water, RS can be performed without pre-treatment on unfixed, hydrated tissue samples, without any labelling and chemical fixation used in histochemical methods. This review provides first the description of basic principles of RS as a biotechnology tool and introduces into the field of currently available RS based techniques, developed to enhance Raman signal. The main spectral processing statistical tools, fingerprint identification and available databases are mentioned. The recent literature has been analysed for such applications of RS as tendon and ligaments, cartilage, bone, and tissue engineered constructs for regenerative medicine. Several cases of proof-of-concept preclinical studies have been described. Finally, advantages, limitations, future perspectives, and challenges for translation of RS into clinical practice have been also discussed.
Collapse
Affiliation(s)
- Marco Fosca
- Istituto di Struttura della Materia Consiglio Nazionale delle Ricerche, 204549, Roma, Lazio, Italy;
| | - Valentina Basoli
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| | - Elena Della Bella
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| | - Fabrizio Russo
- Campus Bio-Medico University Hospital, 220431, Roma, Lazio, Italy;
| | - Gianluca Vadala
- Campus Bio-Medico University Hospital, 220431, Roma, Lazio, Italy;
| | - Mauro Alini
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| | - Julietta V Rau
- Istituto di Struttura della Materia Consiglio Nazionale delle Ricerche, 204549, Roma, Lazio, Italy.,I M Sechenov First Moscow State Medical University, 68477, Moskva, Moskva, Russian Federation;
| | - Sophie Verrier
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| |
Collapse
|
20
|
Monaco G, Ladner YD, El Haj AJ, Forsyth NR, Alini M, Stoddart MJ. Mesenchymal Stromal Cell Differentiation for Generating Cartilage and Bone-Like Tissues In Vitro. Cells 2021; 10:cells10082165. [PMID: 34440934 PMCID: PMC8391162 DOI: 10.3390/cells10082165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 01/22/2023] Open
Abstract
In the field of tissue engineering, progress has been made towards the development of new treatments for cartilage and bone defects. However, in vitro culture conditions for human bone marrow mesenchymal stromal cells (hBMSCs) have not yet been fully defined. To improve our understanding of cartilage and bone in vitro differentiation, we investigated the effect of culture conditions on hBMSC differentiation. We hypothesized that the use of two different culture media including specific growth factors, TGFβ1 or BMP2, as well as low (2% O2) or high (20% O2) oxygen tension, would improve the chondrogenic and osteogenic potential, respectively. Chondrogenic and osteogenic differentiation of hBMSCs isolated from multiple donors and expanded under the same conditions were directly compared. Chondrogenic groups showed a notable upregulation of chondrogenic markers compared with osteogenic groups. Greater sGAG production and deposition, and collagen type II and I accumulation occurred for chondrogenic groups. Chondrogenesis at 2% O2 significantly reduced ALP gene expression and reduced type I collagen deposition, producing a more stable and less hypertrophic chondrogenic phenotype. An O2 tension of 2% did not inhibit osteogenic differentiation at the protein level but reduced ALP and OC gene expression. An upregulation of ALP and OC occurred during osteogenesis in BMP2 containing media under 20% O2; BMP2 free osteogenic media downregulated ALP and also led to higher sGAG release. A higher mineralization was observed in the presence of BMP2 during osteogenesis. This study demonstrates how the modulation of O2 tension, combined with tissue-specific growth factors and media composition can be tailored in vitro to promote chondral or endochondral differentiation while using the same donor cell population.
Collapse
Affiliation(s)
- Graziana Monaco
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
| | - Yann D. Ladner
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
- Institute for Biomechanics, ETH Zurich, Lengghalde 5, CH-8008 Zurich, Switzerland
| | - Alicia J. El Haj
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TT, UK;
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
| | - Mauro Alini
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
| | - Martin J. Stoddart
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
- Correspondence:
| |
Collapse
|
21
|
Aquino JB, Sierra R, Montaldo LA. Diverse cellular origins of adult blood vascular endothelial cells. Dev Biol 2021; 477:117-132. [PMID: 34048734 DOI: 10.1016/j.ydbio.2021.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
During embryonic stages, vascular endothelial cells (ECs) originate from the mesoderm, at specific extraembryonic and embryonic regions, through a process called vasculogenesis. In the adult, EC renewal/replacement mostly depend on local resident ECs or endothelial progenitor cells (EPCs). Nevertheless, contribution from circulating ECs/EPCs was also reported. In addition, cells lacking from EC/EPC markers with in vitro extended plasticity were shown to originate endothelial-like cells (ELCs). Most of these cells consist of mesenchymal stromal progenitors, which would eventually get mobilized from the bone marrow after injury. Based on that, current knowledge on different mouse and human bone marrow stromal cell (BM-SC) subpopulations, able to contribute with mesenchymal stromal/stem cells (MSCs), is herein reviewed. Such analyses underline an unexpected heterogeneity among sinusoidal LepR+ stromal/CAR cells. For instance, in a recent report a subgroup of LepR+ stromal/CAR progenitors, which express GLAST and is traced in Wnt1Cre;R26RTom mice, was found to contribute with ELCs in vivo. These GLAST + Wnt1+ BM-SCs were shown to get mobilized to the peripheral blood and to contribute with liver regeneration. Other sources of ELCs, such as adipose, neural and dental pulp tissues, were also published. Finally, mechanisms likely involved in the enhanced cellular plasticity properties of bone marrow/adipose tissue stromal cells, able to originate ELCs, are assessed. In the future, strategies to analyze the in vivo expression profile of stromal cells, with MSC properties, in combination with screening of active genomic regions at the single cell-level, during early postnatal development and/or after injury, will likely help understanding properties of these ELC sources.
Collapse
Affiliation(s)
- Jorge B Aquino
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina.
| | - Romina Sierra
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| | - Laura A Montaldo
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| |
Collapse
|
22
|
Recent Developed Strategies for Enhancing Chondrogenic Differentiation of MSC: Impact on MSC-Based Therapy for Cartilage Regeneration. Stem Cells Int 2021; 2021:8830834. [PMID: 33824665 PMCID: PMC8007380 DOI: 10.1155/2021/8830834] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/20/2021] [Accepted: 03/04/2021] [Indexed: 12/19/2022] Open
Abstract
Articular cartilage is susceptible to damage, but its self-repair is hindered by its avascular nature. Traditional treatment methods are not able to achieve satisfactory repair effects, and the development of tissue engineering techniques has shed new light on cartilage regeneration. Mesenchymal stem cells (MSCs) are one of the most commonly used seed cells in cartilage tissue engineering. However, MSCs tend to lose their multipotency, and the composition and structure of cartilage-like tissues formed by MSCs are far from those of native cartilage. Thus, there is an urgent need to develop strategies that promote MSC chondrogenic differentiation to give rise to durable and phenotypically correct regenerated cartilage. This review provides an overview of recent advances in enhancement strategies for MSC chondrogenic differentiation, including optimization of bioactive factors, culture conditions, cell type selection, coculture, gene editing, scaffolds, and physical stimulation. This review will aid the further understanding of the MSC chondrogenic differentiation process and enable improvement of MSC-based cartilage tissue engineering.
Collapse
|
23
|
Zha K, Li X, Yang Z, Tian G, Sun Z, Sui X, Dai Y, Liu S, Guo Q. Heterogeneity of mesenchymal stem cells in cartilage regeneration: from characterization to application. NPJ Regen Med 2021; 6:14. [PMID: 33741999 PMCID: PMC7979687 DOI: 10.1038/s41536-021-00122-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
Articular cartilage is susceptible to damage but hard to self-repair due to its avascular nature. Traditional treatment methods are not able to produce satisfactory effects. Mesenchymal stem cells (MSCs) have shown great promise in cartilage repair. However, the therapeutic effect of MSCs is often unstable partly due to their heterogeneity. Understanding the heterogeneity of MSCs and the potential of different types of MSCs for cartilage regeneration will facilitate the selection of superior MSCs for treating cartilage damage. This review provides an overview of the heterogeneity of MSCs at the donor, tissue source and cell immunophenotype levels, including their cytological properties, such as their ability for proliferation, chondrogenic differentiation and immunoregulation, as well as their current applications in cartilage regeneration. This information will improve the precision of MSC-based therapeutic strategies, thus maximizing the efficiency of articular cartilage repair.
Collapse
Affiliation(s)
- Kangkang Zha
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhen Yang
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Guangzhao Tian
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiqiang Sun
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xiang Sui
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
| | - Yongjing Dai
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
| | - Shuyun Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China.
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW One aim in bone tissue engineering is to develop human cell-based, 3D in vitro bone models to study bone physiology and pathology. Due to the heterogeneity of cells among patients, patient's own cells are needed to be obtained, ideally, from one single cell source. This review attempts to identify the appropriate cell sources for development of such models. RECENT FINDINGS Bone marrow and peripheral blood are considered as suitable sources for extraction of osteoblast/osteocyte and osteoclast progenitor cells. Recent studies on these cell sources have shown no significant differences between isolated progenitor cells. However, various parameters such as medium composition affect the cell's proliferation and differentiation potential which could make the peripheral blood-derived stem cells superior to the ones from bone marrow. Peripheral blood can be considered a suitable source for osteoblast/osteocyte and osteoclast progenitor cells, being less invasive for the patient. However, more investigations are needed focusing on extraction and differentiation of both cell types from the same donor sample of peripheral blood.
Collapse
Affiliation(s)
- Sana Ansari
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
25
|
Herrmann M, Diederichs S, Melnik S, Riegger J, Trivanović D, Li S, Jenei-Lanzl Z, Brenner RE, Huber-Lang M, Zaucke F, Schildberg FA, Grässel S. Extracellular Vesicles in Musculoskeletal Pathologies and Regeneration. Front Bioeng Biotechnol 2021; 8:624096. [PMID: 33553127 PMCID: PMC7855463 DOI: 10.3389/fbioe.2020.624096] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence of musculoskeletal diseases is steadily increasing with aging of the population. In the past years, extracellular vesicles (EVs) have gained attention in musculoskeletal research. EVs have been associated with various musculoskeletal pathologies as well as suggested as treatment option. EVs play a pivotal role in communication between cells and their environment. Thereby, the EV cargo is highly dependent on their cellular origin. In this review, we summarize putative mechanisms by which EVs can contribute to musculoskeletal tissue homeostasis, regeneration and disease, in particular matrix remodeling and mineralization, pro-angiogenic effects and immunomodulatory activities. Mesenchymal stromal cells (MSCs) present the most frequently used cell source for EV generation for musculoskeletal applications, and herein we discuss how the MSC phenotype can influence the cargo and thus the regenerative potential of EVs. Induced pluripotent stem cell-derived mesenchymal progenitor cells (iMPs) may overcome current limitations of MSCs, and iMP-derived EVs are discussed as an alternative strategy. In the last part of the article, we focus on therapeutic applications of EVs and discuss both practical considerations for EV production and the current state of EV-based therapies.
Collapse
Affiliation(s)
- Marietta Herrmann
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Svitlana Melnik
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Drenka Trivanović
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Shushan Li
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Rolf E. Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Susanne Grässel
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| |
Collapse
|
26
|
Pomicter AD, Clair PM, Yan D, Heaton WL, Eiring AM, Anderson MB, Richards SM, Gililland J, O'Hare T, Deininger MW. Femoral Heads from Total Hip Arthroplasty as a Source of Adult Hematopoietic Cells. Acta Haematol 2021; 144:458-464. [PMID: 33412552 DOI: 10.1159/000511953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022]
Abstract
Normal human bone marrow cells are critical for studies of hematopoiesis and as controls to assess toxicity. As cells from commercial vendors are expensive, many laboratories resort to cancer-free bone marrow specimens obtained during staging or to umbilical cord blood cells, which may be abnormal or reflect a much younger age group compared to the disease samples under study. We piloted the use of femoral heads as an alternative and inexpensive source of normal bone marrow. Femoral heads were obtained from 21 successive patients undergoing elective hip arthroplasty. Mononuclear cells (MNCs) were purified with Ficoll, and CD3+, CD14+, and CD34+ cells were purified with antibody-coated microbeads. The median yield of MNCs was 8.95 × 107 (range, 1.62 × 105-2.52 × 108), and the median yield of CD34+ cells was 1.40 × 106 (range, 3.60 × 105-9.90 × 106). Results of downstream applications including qRT-PCR, colony-forming assays, and ex vivo proliferation analysis were of high quality and comparable to those obtained with standard bone marrow aspirates. We conclude that femoral heads currently discarded as medical waste are a cost-efficient source of bone marrow cells for research use.
Collapse
Affiliation(s)
- Anthony D Pomicter
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Phillip M Clair
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Dongqing Yan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - William L Heaton
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Anna M Eiring
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Anderson
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Stephen M Richards
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Jeremy Gililland
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Thomas O'Hare
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, Utah, USA
| | - Michael W Deininger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA,
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, Utah, USA,
| |
Collapse
|
27
|
Wangler S, Kamali A, Wapp C, Wuertz-Kozak K, Häckel S, Fortes C, Benneker LM, Haglund L, Richards RG, Alini M, Peroglio M, Grad S. Uncovering the secretome of mesenchymal stromal cells exposed to healthy, traumatic, and degenerative intervertebral discs: a proteomic analysis. Stem Cell Res Ther 2021; 12:11. [PMID: 33413584 PMCID: PMC7789679 DOI: 10.1186/s13287-020-02062-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been introduced as promising cell source for regenerative medicine. Besides their multilineage differentiation capacity, MSCs release a wide spectrum of bioactive factors. This secretome holds immunomodulatory and regenerative capacities. In intervertebral disc (IVD) cells, application of MSC secretome has been shown to decrease the apoptosis rate, induce proliferation, and promote production of extracellular matrix (ECM). For clinical translation of secretome-based treatment, characterization of the secretome composition is needed to better understand the induced biological processes and identify potentially effective secretomes. METHODS This study aimed to investigate the proteome released by bone marrow-derived MSCs following exposure to a healthy, traumatic, or degenerative human IVD environment by mass spectroscopy and quantitative immunoassay analyses. Exposure of MSCs to the proinflammatory stimulus interleukin 1β (IL-1β) was used as control. RESULTS Compared to MSC baseline secretome, there were 224 significantly up- or downregulated proteins following healthy, 179 following traumatic, 223 following degenerative IVD, and 160 proteins following IL-1β stimulus. Stimulation of MSCs with IVD conditioned media induced a more complex MSC secretome, involving more biological processes, compared to stimulation with IL-1β. The MSC response to stimulation with IVD conditioned medium was dependent on their pathological status. CONCLUSIONS The MSC secretome seemed to match the primary need of the IVD: homeostasis maintenance in the case of healthy IVDs, versus immunomodulation, adjustment of ECM synthesis and degradation disbalance, and ECM (re) organization in the case of traumatic and degenerative IVDs. These findings highlight the importance of cell preconditioning in the development of tailored secretome therapies. The secretome of human bone marrow-derived mesenchymal stromal cells (MSCs) stimulated with intervertebral disc (IVD) conditioned medium was analyzed by proteomic profiling. Depending on the pathological state of the IVD, the MSC secretome protein composition indicated immunomodulatory or anabolic activity of the secretome. These findings may have implications for tailored secretome therapy for the IVD and other tissues.
Collapse
Affiliation(s)
- Sebastian Wangler
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amir Kamali
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Christina Wapp
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Karin Wuertz-Kozak
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY, USA
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, Germany
| | - Sonja Häckel
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Lorin M Benneker
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lisbet Haglund
- Department of Surgery, Division of Orthopaedics, Faculty of Medicine, McGill University, Montreal, Canada
| | - R Geoff Richards
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Marianna Peroglio
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Sibylle Grad
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland.
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
Xia Y, Chen J, Ding J, Zhang J, Chen H. IGF1- and BM-MSC-incorporating collagen-chitosan scaffolds promote wound healing and hair follicle regeneration. Am J Transl Res 2020; 12:6264-6276. [PMID: 33194028 PMCID: PMC7653568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Full-thickness skin injury affects millions of people worldwide each year. Although bone marrow-derived mesenchymal stem cells (BM-MSCs) have been shown to promote cutaneous wound healing, they cannot functionally promote wound healing with the recovery of appendages such as hair follicles. We previously found that growth factor plus BM-MSCs could effectively promote wound healing and hair follicle regeneration. In the present study, we grafted insulin-like growth factor 1 (IGF1), a multifunctional cell growth factor, and BM-MSCs into a collagen-chitosan scaffold to investigate their effects on functional wound healing. Using scanning electron microscopy, histological staining, and quantitative analysis, we found that IGF1- and BM-MSC-incorporating collagen-chitosan scaffolds promote cutaneous wound healing with effective regeneration of hair follicles in a rat full-thickness skin injury model. In addition, IGF1/BM-MSCs inhibit inflammatory cytokines during wound healing. In vitro, we found that IGF1 promotes the proliferation and migration of BM-MSCs via the IGFR-mediated ERK1/2 signaling pathway. Collectively, in this study, we first demonstrated that IGF1 enhances BM-MSC-mediated wound healing as well as hair follicle regeneration. Our data suggest that the topical application of IGF1 and BM-MSCs incorporated in collagen-chitosan scaffolds can be used as a feasible and effective therapeutic approach to improve functional cutaneous wound healing.
Collapse
Affiliation(s)
- Ying Xia
- Medical Cosmetology Department, Plastic Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang Province, P. R. China
| | - Jianshe Chen
- Department of Dermatology, The First Affiliated Hospital, Xiamen UniversityXiamen 361000, Fujian Province, P. R. China
| | - Juan Ding
- Medical Cosmetology Department, Plastic Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang Province, P. R. China
| | - Jianqing Zhang
- Medical Cosmetology Department, Plastic Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang Province, P. R. China
| | - Hong Chen
- Medical Cosmetology Department, Plastic Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang Province, P. R. China
| |
Collapse
|
29
|
Efficient Non-Viral Gene Modification of Mesenchymal Stromal Cells from Umbilical Cord Wharton's Jelly with Polyethylenimine. Pharmaceutics 2020; 12:pharmaceutics12090896. [PMID: 32971730 PMCID: PMC7559368 DOI: 10.3390/pharmaceutics12090896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stromal cells (MSC) derived from human umbilical cord Wharton’s jelly (WJ) have a wide therapeutic potential in cell therapy and tissue engineering because of their multipotential capacity, which can be reinforced through gene therapy in order to modulate specific responses. However, reported methodologies to transfect WJ-MSC using cationic polymers are scarce. Here, WJ-MSC were transfected using 25 kDa branched- polyethylenimine (PEI) and a DNA plasmid encoding GFP. PEI/plasmid complexes were characterized to establish the best transfection efficiencies with lowest toxicity. Expression of MSC-related cell surface markers was evaluated. Likewise, immunomodulatory activity and multipotential capacity of transfected WJ-MSC were assessed by CD2/CD3/CD28-activated peripheral blood mononuclear cells (PBMC) cocultures and osteogenic and adipogenic differentiation assays, respectively. An association between cell number, PEI and DNA content, and transfection efficiency was observed. The highest transfection efficiency (15.3 ± 8.6%) at the lowest toxicity was achieved using 2 ng/μL DNA and 3.6 ng/μL PEI with 45,000 WJ-MSC in a 24-well plate format (200 μL). Under these conditions, there was no significant difference between the expression of MSC-identity markers, inhibitory effect on CD3+ T lymphocytes proliferation and osteogenic/adipogenic differentiation ability of transfected WJ-MSC, as compared with non-transfected cells. These results suggest that the functional properties of WJ-MSC were not altered after optimized transfection with PEI.
Collapse
|
30
|
Robert AW, Marcon BH, Dallagiovanna B, Shigunov P. Adipogenesis, Osteogenesis, and Chondrogenesis of Human Mesenchymal Stem/Stromal Cells: A Comparative Transcriptome Approach. Front Cell Dev Biol 2020; 8:561. [PMID: 32733882 PMCID: PMC7362937 DOI: 10.3389/fcell.2020.00561] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022] Open
Abstract
Adipogenesis, osteogenesis and chondrogenesis of human mesenchymal stem/stromal cells (MSC) are complex and highly regulated processes. Over the years, several studies have focused on understanding the mechanisms involved in the MSC commitment to the osteogenic, adipogenic and/or chondrogenic phenotypes. High-throughput methodologies have been used to investigate the gene expression profile during differentiation. Association of data analysis of mRNAs, microRNAs, circular RNAs and long non-coding RNAs, obtained at different time points over these processes, are important to depict the complexity of differentiation. This review will discuss the results that were highlighted in transcriptome analyses of MSC undergoing adipogenic, osteogenic and chondrogenic differentiation. The focus is to shed light on key molecules, main signaling pathways and biological processes related to different time points of adipogenesis, osteogenesis and chondrogenesis.
Collapse
Affiliation(s)
- Anny W Robert
- Instituto Carlos Chagas - Fiocruz Paraná, Curitiba, Brazil
| | - Bruna H Marcon
- Instituto Carlos Chagas - Fiocruz Paraná, Curitiba, Brazil
| | | | | |
Collapse
|
31
|
Luo S, Wu S, Xu J, Zhang X, Zou L, Yao R, Jin L, Li Y. Osteogenic differentiation of BMSCs on MoS2 composite nanofibers with different cell seeding densities. APPLIED NANOSCIENCE 2020. [DOI: 10.1007/s13204-020-01473-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
32
|
Xiao Y, Zhu Q, Liu X, Jiang M, Hao H, Zhu H, Cowan PJ, He X, Liu Q, Zhou S, Liu Z. High-fat diet selectively decreases bone marrow lin - /CD117 + cell population in aging mice through increased ROS production. J Tissue Eng Regen Med 2020; 14:884-892. [PMID: 32337800 DOI: 10.1002/term.3047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
Abstract
Bone marrow (BM) stem cells (BMSCs) are an important source for cell therapy. The outcome of cell therapy could be ultimately associated with the number and function of donor BMSCs. The present study was to evaluate the effect of long-term high-fat diet (HFD) on the population of BMSCs and the role of reactive oxygen species (ROS) in aging mice. Forty-week-old male C57BL/6 mice were fed with HFD for 3 months with regular diet as control. Experiments were repeated when ROS production was reduced in mice treated with N-acetylcysteine (NAC) or using mice overexpressing antioxidant enzyme network (AON) of superoxide dismutase (SOD)1, SOD3, and glutathione peroxidase. BM and blood cells were analyzed with flowcytometry for lineage negative (lin- ) and Sca-1+ , or lin- /CD117+ , or lin- /CD133+ cells. Lin- /CD117+ cell population was significantly decreased with increased intracellular ROS and apoptosis and decreased proliferation in BM, not in blood, in HFD-treated mice without change for Sca-1+ or CD133+ cell populations in BM or blood. NAC treatment or AON overexpression effectively prevented HFD-induced intracellular ROS production and reduction of BM lin- /CD117+ population. These data suggested that long-term HFD selectively decreased BM lin- /CD117+ cell population in aging mice through increased ROS production.
Collapse
Affiliation(s)
- Yichao Xiao
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.,Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Qingyi Zhu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.,Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Xuanyou Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Meng Jiang
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Hua Zhu
- Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH, USA
| | - Peter J Cowan
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Immunology Research Centre, St. Vincent's Hospital, Melbourne, Victoria, Australia
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
33
|
Della Bella E, Menzel U, Basoli V, Tourbier C, Alini M, Stoddart MJ. Differential Regulation of circRNA, miRNA, and piRNA during Early Osteogenic and Chondrogenic Differentiation of Human Mesenchymal Stromal Cells. Cells 2020; 9:cells9020398. [PMID: 32050423 PMCID: PMC7072123 DOI: 10.3390/cells9020398] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
The goal of the present study is to identify the differential expression of circular RNA (circRNA), miRNA, and piwi-interacting RNA (piRNA) after lineage commitment towards osteo- and chondrogenesis of human bone marrow mesenchymal stromal cells (hMSCs). The cells were maintained for 7 days in either osteogenic or chondrogenic medium. RNA sequencing was performed to assess the expression of miRNA and piRNA, while RNA hybridization arrays were used to identify which circRNA were differentially expressed. qPCR validation of a selection of targets for both osteogenic and chondrogenic differentiation was carried out. The differential expression of several circRNA, miRNA, and piRNA was identified and validated. The expression of total and circular isoforms of FKBP5 was upregulated both in osteo- and chondrogenesis and it was influenced by the presence of dexamethasone. ZEB1, FADS2, and SMYD3 were also identified as regulated in differentiation and/or by dexamethasone. In conclusion, we have identified a set of different non-coding RNAs that are differentially regulated in early osteogenic and chondrogenic differentiation, paving the way for further investigation to understand how dexamethasone controls the expression of those genes and what their function is in MSC differentiation.
Collapse
Affiliation(s)
- Elena Della Bella
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (U.M.); (V.B.); (C.T.); (M.A.)
| | - Ursula Menzel
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (U.M.); (V.B.); (C.T.); (M.A.)
| | - Valentina Basoli
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (U.M.); (V.B.); (C.T.); (M.A.)
| | - Céline Tourbier
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (U.M.); (V.B.); (C.T.); (M.A.)
- Department of Cranio-Maxiofacial Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79085 Freiburg, Germany
| | - Mauro Alini
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (U.M.); (V.B.); (C.T.); (M.A.)
| | - Martin J. Stoddart
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (U.M.); (V.B.); (C.T.); (M.A.)
- Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
- Correspondence:
| |
Collapse
|
34
|
Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal Stromal Cell-Based Bone Regeneration Therapies: From Cell Transplantation and Tissue Engineering to Therapeutic Secretomes and Extracellular Vesicles. Front Bioeng Biotechnol 2019; 7:352. [PMID: 31828066 PMCID: PMC6890555 DOI: 10.3389/fbioe.2019.00352] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Effective regeneration of bone defects often presents significant challenges, particularly in patients with decreased tissue regeneration capacity due to extensive trauma, disease, and/or advanced age. A number of studies have focused on enhancing bone regeneration by applying mesenchymal stromal cells (MSCs) or MSC-based bone tissue engineering strategies. However, translation of these approaches from basic research findings to clinical use has been hampered by the limited understanding of MSC therapeutic actions and complexities, as well as costs related to the manufacturing, regulatory approval, and clinical use of living cells and engineered tissues. More recently, a shift from the view of MSCs directly contributing to tissue regeneration toward appreciating MSCs as "cell factories" that secrete a variety of bioactive molecules and extracellular vesicles with trophic and immunomodulatory activities has steered research into new MSC-based, "cell-free" therapeutic modalities. The current review recapitulates recent developments, challenges, and future perspectives of these various MSC-based bone tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Traweger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Spinal Cord Injury & Tissue Regeneration Center Salzburg, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|