1
|
Soumya BS, Gamit N, Patil M, Shreenidhi VP, Dharmarajan A, Warrier S. Modeling amyotrophic lateral sclerosis with amniotic membrane-derived mesenchymal stem cells: A novel approach for disease modeling. Exp Cell Res 2025; 446:114449. [PMID: 39961464 DOI: 10.1016/j.yexcr.2025.114449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/16/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Advancement of therapeutics for neurodegenerative diseases like amyotrophic lateral sclerosis (ALS) has been predominantly hampered by the dearth of relevant disease models. Despite numerous animal models, significant challenges remain in correlating these with human disease complexities. In this study, the ALS model was created using amniotic membrane-derived mesenchymal stem cells (AM-MSCs) which were differentiated into motor neurons (MN) with specific MN induction media and transiently transfected with mutated human SOD1 G93A plasmid to induce ALS-like condition. Characterization included gene expression analysis, immunocytochemistry, flow cytometry, and Western blot. Functional assays assessed the extent of degeneration and model efficiency. AM-MSCs demonstrated multipotency and were positive for MSC markers. Upon differentiation, the expression of MN markers like MNX1, Olig2, and ChAT were found to be elevated. SOD1 G93A overexpression, downregulated MN markers, upregulated NURR1 gene, reduced acetylcholine (ACh), reduced glutathione, and elevated oxidative stress markers. This robust in-vitro ALS model derived from AM-MSCs offers an alternative to animal models to provide an efficient and cost-effective platform to conduct rapid drug screening.
Collapse
Affiliation(s)
- B S Soumya
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - V P Shreenidhi
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India
| | - Arun Dharmarajan
- School of Human Sciences, Faculty of Life and Physical Sciences, The University of Western Australia, Perth, WA 6009, Australia; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, 560065, India; Division of Regenerative Medicine and Cancer Stem Cells, Department of Biotechnology, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, 600116, India.
| |
Collapse
|
2
|
Mattei DN, Harman RM, Van de Walle GR, Smith R, Grivel JC, Abdelalim EM, Vinardell T. Effect of pregnancy on isolation efficiency and in vitro proliferation of equine peripheral-blood derived mesenchymal stromal cells. Theriogenology 2024; 224:107-118. [PMID: 38761667 DOI: 10.1016/j.theriogenology.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Mesenchymal stromal cells (MSCs) have regenerative and immunomodulatory potential and may be used to treat injured tissues. Pregnancy has been associated with increased MSCs in the peripheral circulation in multiple species, but to date, there are no reports on this matter in horses. This study aimed to evaluate the effect of pregnancy on isolation efficiency and proliferation capacity of equine MSCs derived from the peripheral blood (PB) of mares. Venous blood samples were collected at the 11th month of gestation and 1 month after delivery from clinically healthy Arabian mares that presented normal pregnancies. Blood samples were processed for in vitro cellular culture and hormonal and metabolic profiles. MSCs were isolated and characterized by trilineage differentiation potential, immunophenotyping, analyzed by gene sequencing and proliferation assays. The isolation of peripheral blood mononuclear cells (PBMCs) of pregnant mares were associated with higher isolation efficiency and proliferative capacity of MSCs derived from peripheral blood (PB-MSCs) recovered pre-partum than those isolated post-partum. Although fetal gender, parity, 5α-reduced pregnanes, insulin, and cortisol were shown to affect cellular proliferation, individual factors and the small population studied must be considered. This study suggests that PB-MSCs from pregnant mares could be a valuable alternative source of MSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Debora N Mattei
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Equine Veterinary Medical Center, Member of Qatar Foundation, P.O. Box 5825, Doha, Qatar
| | - Rebecca M Harman
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Rd, Ithaca, NY 14850, USA
| | - Gerlinde R Van de Walle
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Rd, Ithaca, NY 14850, USA
| | - Roger Smith
- Department of Clinical Science and Services, The Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, United Kingdom
| | - Jean Charles Grivel
- Deep Phenotyping Core, Sidra Medicine, PO Box 26999, Al Garrafa St, Ar-Rayyan, Doha, Qatar
| | - Essam M Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Division, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar; Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar
| | - Tatiana Vinardell
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Equine Veterinary Medical Center, Member of Qatar Foundation, P.O. Box 5825, Doha, Qatar.
| |
Collapse
|
3
|
Pereira L, Echarte L, Romero M, Grazioli G, Pérez-Campos H, Francia A, Vicentino W, Mombrú AW, Faccio R, Álvarez I, Touriño C, Pardo H. Synthesis and characterization of a bovine collagen: GAG scaffold with Uruguayan raw material for tissue engineering. Cell Tissue Bank 2024; 25:123-142. [PMID: 34536180 DOI: 10.1007/s10561-021-09960-6] [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: 07/11/2020] [Accepted: 09/06/2021] [Indexed: 11/28/2022]
Abstract
Tissue engineering (TE) and regenerative medicine offer strategies to improve damaged tissues by using scaffolds and cells. The use of collagen-based biomaterials in the field of TE has been intensively growing over the past decades. Mesenchymal stromal cells (MSCs) and dental pulp stem cells (DPSCs) are promising cell candidates for development of clinical composites. In this study, we proposed the development of a bovine collagen type I: chondroitin-6-sulphate (CG) scaffold, obtained from Uruguayan raw material (certified as free bovine spongiform encephalopathy), with CG crosslinking enhancement using different gamma radiation doses. Structural, biomechanical and chemical characteristics of the scaffolds were assessed by Scanning Electron Microscopy, axial tensile tests, FT-IR and Raman Spectroscopy, respectively. Once we selected the most appropriate scaffold for future use as a TE product, we studied the behavior of MSCs and DPSCs cultured on the scaffold by cytotoxicity, proliferation and differentiation assays. Among the diverse porous scaffolds obtained, the one with the most adequate properties was the one exposed to 15 kGy of gamma radiation. This radiation dose contributed to the crosslinking of molecules, to the formation of new bonds and/or to the reorganization of the collagen fibers. The selected scaffold was non-cytotoxic for the tested cells and a suitable substrate for cell proliferation. Furthermore, the scaffold allowed MSCs differentiation to osteogenic, chondrogenic, and adipogenic lineages. Thus, this work shows a promising approach to the synthesis of a collagen-scaffold suitable for TE.
Collapse
Affiliation(s)
- L Pereira
- Centro NanoMat, Facultad de Química, Instituto Polo Tecnológico de Pando, UdelaR, Camino Aparicio Saravia s/n, 9100, Pando, Canelones, Uruguay
| | - L Echarte
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - M Romero
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - G Grazioli
- Cátedra de Materiales Dentales, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - H Pérez-Campos
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A Francia
- Fisiología general y bucodental, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - W Vicentino
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A W Mombrú
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - R Faccio
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - I Álvarez
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - C Touriño
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay.
| | - H Pardo
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay.
| |
Collapse
|
4
|
Taguchi T, Lopez M, Takawira C. Viable tendon neotissue from adult adipose-derived multipotent stromal cells. Front Bioeng Biotechnol 2024; 11:1290693. [PMID: 38260742 PMCID: PMC10800559 DOI: 10.3389/fbioe.2023.1290693] [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: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Background: Tendon healing is frequently prolonged, unpredictable, and results in poor tissue quality. Neotissue formed by adult multipotent stromal cells has the potential to guide healthy tendon tissue formation. Objectives: The objective of this study was to characterize tendon neotissue generated by equine adult adipose-derived multipotent stromal cells (ASCs) on collagen type I (COLI) templates under 10% strain in a novel bioreactor. The tested hypothesis was that ASCs assume a tendon progenitor cell-like morphology, express tendon-related genes, and produce more organized extracellular matrix (ECM) in tenogenic versus stromal medium with perfusion and centrifugal fluid motion. Methods: Equine ASCs on COLI sponge cylinders were cultured in stromal or tenogenic medium within bioreactors during combined perfusion and centrifugal fluid motion for 7, 14, or 21 days under 10% strain. Viable cell distribution and number, tendon-related gene expression, and micro- and ultra-structure were evaluated with calcein-AM/EthD-1 staining, resazurin reduction, RT-PCR, and light, transmission, and scanning electron microscopy. Fibromodulin was localized with immunohistochemistry. Cell number and gene expression were compared between culture media and among culture periods (p < 0.05). Results: Viable cells were distributed throughout constructs for up to 21 days of culture, and cell numbers were higher in tenogenic medium. Individual cells had a round or rhomboid shape with scant ECM in stromal medium in contrast to clusters of parallel, elongated cells surrounded by highly organized ECM in tenogenic medium after 21 days of culture. Transcription factor, extracellular matrix, and mature tendon gene expression profiles confirmed ASC differentiation to a tendon progenitor-like cell in tenogenic medium. Construct micro- and ultra-structure were consistent with tendon neotissue and fibromodulin was present in the ECM after culture in tenogenic medium. Conclusion: Long-term culture in custom bioreactors with combined perfusion and centrifugal tenogenic medium circulation supports differentiation of equine adult ASCs into tendon progenitor-like cells capable of neotissue formation.
Collapse
|
5
|
Rajasingh S, Vembuli H, Perales S, Rajasingh J. Safe and Noninvasive Method for Generating Induced Mesenchymal Stem Cells from Urinary Epithelial Cells. Methods Mol Biol 2024; 2835:1-15. [PMID: 39105901 DOI: 10.1007/978-1-0716-3995-5_1] [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: 08/07/2024]
Abstract
Mesenchymal stem cells (MSCs) exhibit remarkable versatility and hold immense potential for tissue regeneration. They are actively investigated in clinical trials for various diseases and injuries, showcasing their therapeutic promise. However, traditional sources of MSCs have limitations in terms of scalability and storage. To address these challenges, this study aims to provide a method of creating an alternative source of induced pluripotent stem cells (iPSCs)-derived MSCs (iMSCs) from urinary epithelial cells (UECs) through a noninvasive procedure. This distinct subset of UECs found in urine samples offers an invaluable resource for generating autologous UE-iPSCs. iPSCs have distinct advantages over embryonic stem cells, as they can be generated from somatic cells, eliminating the need for human embryos and associated ethical concerns. Advancements in iPSC technology enable the differentiation of iMSCs, allowing researchers to create disease models, gain insights into disease mechanisms, and develop targeted therapies. This straightforward and noninvasive method aims to enhance the production of high-quality, autologous iMSCs with significant replicative and differentiation potential, making them suitable for regenerative therapy.
Collapse
Affiliation(s)
- Sheeja Rajasingh
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hemanathan Vembuli
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Selene Perales
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
6
|
Liao S, Li J, Gao S, Han Y, Han X, Wu Y, Bi J, Xu M, Bi W. Sulfatinib, a novel multi-targeted tyrosine kinase inhibitor of FGFR1, CSF1R, and VEGFR1-3, suppresses osteosarcoma proliferation and invasion via dual role in tumor cells and tumor microenvironment. Front Oncol 2023; 13:1158857. [PMID: 37361567 PMCID: PMC10286821 DOI: 10.3389/fonc.2023.1158857] [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: 02/04/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction Tumor progression is driven by intrinsic malignant behaviors caused by gene mutation or epigenetic modulation, as well as crosstalk with the components in the tumor microenvironment (TME). Considering the current understanding of the tumor microenvironment, targeting the immunomodulatory stromal cells such as cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) could provide a potential therapeutic strategy. Here, we investigated the effect of sulfatinib, a multi-targeted tyrosine kinase inhibitor (TKI) of FGFR1, CSF1R, and VEGFR1-3, on the treatment of osteosarcoma (OS). Methods In vitro, the antitumor effect was tested by clony formation assay and apoptosis assay.The inhibition of tumor migration and invasion was detected by Transwell assay, and the de-polarization of macrophage was detected by flow cytometry.In vivo, subcutaneous and orthotopic tumor models were established to verify antitumor effect, and the underlying mechanism was verified by immunohistochemistry(IHC), immunofluorescence(IF) and flow cytometry. Results Sulfatinib suppressed OS cell migration and invasion by inhibiting epithelial-mesenchymal transition (EMT) by blocking the secretion of basic fibroblast growth factor (bFGF) in an autocrine manner. In addition, it regulated immune TME via inhibition of the migration of skeletal stem cells (SSCs) to the TME and the differentiation from SSCs to CAFs. Moreover, sulfatinib can suppress OS by modulation of the TME by inhibiting M2 polarization of macrophages. Systemic treatment of sulfatinib can reduce immunosuppression cells M2-TAMs, Tregs, and myeloid-derived suppressor cells (MDSCs) and increase cytotoxic T-cell infiltration in tumors, the lungs, and the spleens. Discussion Our preclinical experiments have shown that sulfatinib can inhibit the proliferation, migration, and invasion of OS by playing a dual role on tumor cells and the tumor microenvironment simultaneously and systematically reverse immunosuppression to immune activation status, which could be translated into clinical trials.
Collapse
Affiliation(s)
- Song Liao
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianxiong Li
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Song Gao
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuchen Han
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xinli Han
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yanan Wu
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jingyou Bi
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Meng Xu
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wenzhi Bi
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Nair RS, Sobhan PK, Shenoy SJ, Prabhu MA, Rema AM, Ramachandran S, C Geetha S, V Pratheesh K, Mony MP, Raj R, Anilkumar TV. A porcine cholecystic extracellular matrix conductive scaffold for cardiac tissue repair. J Biomed Mater Res B Appl Biomater 2022; 110:2039-2049. [PMID: 35305082 DOI: 10.1002/jbm.b.35058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/12/2021] [Accepted: 08/22/2021] [Indexed: 11/08/2022]
Abstract
Cardiac tissue engineering using cells, scaffolds or signaling molecules is a promising approach for replacement or repair of damaged myocardium. This study addressed the contemporary need for a conductive biomimetic nanocomposite scaffold for cardiac tissue engineering by examining the use of a gold nanoparticle-incorporated porcine cholecystic extracellular matrix for the same. The scaffold had an electrical conductivity (0.74 ± 0.03 S/m) within the range of native myocardium. It was a suitable substrate for the growth and differentiation of cardiomyoblast (H9c2) as well as rat mesenchymal stem cells to cardiomyocyte-like cells. Moreover, as an epicardial patch, the scaffold promoted neovascularisation and cell proliferation in infarcted myocardium of rats. It was concluded that the gold nanoparticle coated cholecystic extracellular matrix is a prospective biomaterial for cardiac tissue engineering.
Collapse
Affiliation(s)
- Reshma S Nair
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Praveen K Sobhan
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Sachin J Shenoy
- Division of In Vivo Models and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Mukund A Prabhu
- Department of Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Aswathy M Rema
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Surya Ramachandran
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Surendran C Geetha
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Kanakarajan V Pratheesh
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Manjula P Mony
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Reshmi Raj
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Thapasimuthu V Anilkumar
- Division of Experimental Pathology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India.,School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| |
Collapse
|
8
|
Taguchi T, Zhang N, Angibeau D, Spivey KP, Lopez MJ. Evaluation of canine adipose-derived multipotent stromal cell differentiation to ligamentoblasts on tensioned collagen type I templates in a custom bioreactor culture system. Am J Vet Res 2021; 82:924-934. [PMID: 34669492 DOI: 10.2460/ajvr.82.11.924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate differentiation of canine adipose-derived multipotent stromal cells (ASCs) into ligamentoblasts on tensioned collagen type I (Col1) templates in a perfusion culture system. SAMPLES Infrapatellar fat pad ASCs from healthy stifle joints of 6 female mixed-breed dogs. PROCEDURES Third-passage ASCs (6 × 106 cells/template) were loaded onto suture-augmented Col1 templates under 15% static strain in perfusion bioreactors. Forty-eight ASC-Col1 constructs were incubated with ligamentogenic (ligamentogenic constructs; n = 24) or stromal medium (stromal constructs; 24) for up to 21 days. Specimens were collected from each construct after 2 hours (day 0) and 7, 14, and 21 days of culture. Cell number, viability, distribution, and morphology; construct collagen content; culture medium procollagen-I-N-terminal peptide concentration; and gene expression were compared between ligamentogenic and stromal constructs. RESULTS ASCs adhered to collagen fibers. Cell numbers increased from days 0 to 7 and days 14 to 21 for both construct types. Relative to stromal constructs, cell morphology and extracellular matrix were more mature and collagen content on day 21 and procollagen-I-N-terminal peptide concentration on days 7 and 21 were greater for ligamentogenic constructs. Ligamentogenic constructs had increased expression of the genes biglycan on day 7, decorin throughout the culture period, and Col1, tenomodulin, fibronectin, and tenascin-c on day 21; expression of Col1, tenomodulin, and tenascin-c increased between days 7 and 21. CONCLUSIONS AND CLINICAL RELEVANCE Ligamentogenic medium was superior to stromal medium for differentiation of ASCs to ligamentoblasts on suture-augmented Col1 scaffolds. Customized ligament neotissue may augment treatment options for dogs with cranial cruciate ligament rupture.
Collapse
Affiliation(s)
- Takashi Taguchi
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Nan Zhang
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Dominique Angibeau
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Kathryn P Spivey
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Mandi J Lopez
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| |
Collapse
|
9
|
Wiśniewska J, Sadowska A, Wójtowicz A, Słyszewska M, Szóstek-Mioduchowska A. Perspective on Stem Cell Therapy in Organ Fibrosis: Animal Models and Human Studies. Life (Basel) 2021; 11:life11101068. [PMID: 34685439 PMCID: PMC8538998 DOI: 10.3390/life11101068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) components that result from the disruption of regulatory processes responsible for ECM synthesis, deposition, and remodeling. Fibrosis develops in response to a trigger or injury and can occur in nearly all organs of the body. Thus, fibrosis leads to severe pathological conditions that disrupt organ architecture and cause loss of function. It has been estimated that severe fibrotic disorders are responsible for up to one-third of deaths worldwide. Although intensive research on the development of new strategies for fibrosis treatment has been carried out, therapeutic approaches remain limited. Since stem cells, especially mesenchymal stem cells (MSCs), show remarkable self-renewal, differentiation, and immunomodulatory capacity, they have been intensively tested in preclinical studies and clinical trials as a potential tool to slow down the progression of fibrosis and improve the quality of life of patients with fibrotic disorders. In this review, we summarize in vitro studies, preclinical studies performed on animal models of human fibrotic diseases, and recent clinical trials on the efficacy of allogeneic and autologous stem cell applications in severe types of fibrosis that develop in lungs, liver, heart, kidney, uterus, and skin. Although the results of the studies seem to be encouraging, there are many aspects of cell-based therapy, including the cell source, dose, administration route and frequency, timing of delivery, and long-term safety, that remain open areas for future investigation. We also discuss the contemporary status, challenges, and future perspectives of stem cell transplantation for therapeutic options in fibrotic diseases as well as we present recent patents for stem cell-based therapies in organ fibrosis.
Collapse
|
10
|
Rajasingh S, Sigamani V, Selvam V, Gurusamy N, Kirankumar S, Vasanthan J, Rajasingh J. Comparative analysis of human induced pluripotent stem cell-derived mesenchymal stem cells and umbilical cord mesenchymal stem cells. J Cell Mol Med 2021; 25:8904-8919. [PMID: 34390186 PMCID: PMC8435459 DOI: 10.1111/jcmm.16851] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/19/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022] Open
Abstract
Generation of induced pluripotent stem cells (iPSCs) and their differentiation into mesenchymal stem/stromal cells (iMSCs) have created exciting source of cells for autologous therapy. In this study, we have compared the therapeutic potential of iMSCs generated from urinary epithelial (UE) cells with the available umbilical cord MSCs (UC‐MSCs). For this, adult UE cells were treated with the mRNA of pluripotent genes (OCT4, NANOG, SOX2, KLF4, MYC and LIN28) and a cocktail of miRNAs under specific culture conditions for generating iPSCs. Our non‐viral and mRNA‐based treatment regimen demonstrated a high reprogramming efficiency to about 30% at passage 0. These UE‐iPSCs were successfully differentiated further into ectoderm, endoderm and mesoderm lineage of cells. Moreover, these UE‐iPSCs were subsequently differentiated into iMSCs and were compared with the UC‐MSCs. These iMSCs were capable of differentiating into osteocytes, chondrocytes and adipocytes. Our qRT‐PCR and Western blot data showed that the CD73, CD90 and CD105 gene transcripts and proteins were highly expressed in iMSCs and UC‐MSCs but not in other cells. The comparative qRT‐PCR data showed that the iMSCs maintained their MSC characteristics without any chromosomal abnormalities even at later passages (P15), during which the UC‐MSCs started losing their MSC characteristics. Importantly, the wound‐healing property demonstrated through migration assay was superior in iMSCs when compared to the UC‐MSCs. In this study, we have demonstrated an excellent non‐invasive and pain‐free method of obtaining iMSCs for regenerative therapy. These homogeneous autologous highly proliferative iMSCs may provide an alternative source of cells to UC‐MSCs for treating various diseases.
Collapse
Affiliation(s)
- Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Vijay Selvam
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Shivaani Kirankumar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Jayavardini Vasanthan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
11
|
Al Naem M, Bourebaba L, Kucharczyk K, Röcken M, Marycz K. Therapeutic mesenchymal stromal stem cells: Isolation, characterization and role in equine regenerative medicine and metabolic disorders. Stem Cell Rev Rep 2021; 16:301-322. [PMID: 31797146 DOI: 10.1007/s12015-019-09932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a popular treatment modality in equine orthopaedics. Regenerative therapies are especially interesting for pathologies like complicated tendinopathies of the distal limb, osteoarthritis, osteochondritis dissecans (OCD) and more recently metabolic disorders. Main sources for MSC harvesting in the horse are bone marrow, adipose tissue and umbilical cord blood. While the acquisition of umbilical cord blood is fairly easy and non-invasive, extraction of bone marrow and adipose tissue requires more invasive techniques. Characterization of the stem cells as a result of any isolation method, is also a crucial step for the confirmation of the cells' stemness properties; thus, three main characteristics must be fulfilled by these cells, namely: adherence, expression of a series of well-defined differentiation clusters as well as pluripotency. EVs, resulting from the paracrine action of MSCs, also play a key role in the therapeutic mechanisms mediated by stem cells; MSC-EVs are thus largely implicated in the regulation of proliferation, maturation, polarization and migration of various target cells. Evidence that EVs alone represent a complex network 0involving different soluble factors and could then reflect biophysical characteristics of parent cells has fuelled the importance of developing highly specific techniques for their isolation and analysis. All these aspects related to the functional and technical understanding of MSCs will be discussed and summarized in this review.
Collapse
Affiliation(s)
- Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kucharczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
12
|
Gopalakrishnan Usha P, Jalajakumari S, Sheela UB, Mohan D, Berry C, Tripathi A, Thankappan Nair ST. Engineering cartilage graft using mesenchymal stem cell laden polyacrylamide-galactoxyloglucan hydrogel for transplantation. J Biomater Appl 2021; 36:541-551. [PMID: 34018854 DOI: 10.1177/08853282211019521] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hydrogels are reported to have various biomedical field applications, and many reports also suggest that soft gels promote stem cell differentiation. Chondrogenic differentiation of mesenchymal stem cells (MSC) is significant in articular cartilage repair. This study focuses on polysaccharide-based hydrogels which enhance chondrocyte lineage differentiation of MSC when grown in the hydrogels. This study implies that the prepared hydrogels promote specific lineage without any external chemical induction factors. The techniques, including immunofluorescence and functional assays to assess the differentiation and in vivo implantation, were employed. All observations paved the way towards confirmation that the galactoxyloglucan-based hydrogel is an attractive candidate for supporting stem cell growth and cartilaginous differentiation.
Collapse
Affiliation(s)
- Preethi Gopalakrishnan Usha
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| | - Sreekutty Jalajakumari
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| | - Unnikrishnan Babukuttan Sheela
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| | - Deepa Mohan
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| | - Catherine Berry
- Centre for the Cellular Microenvironment, Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Anuj Tripathi
- Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Sreelekha T Thankappan Nair
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| |
Collapse
|
13
|
Bukowska J, Szóstek-Mioduchowska AZ, Kopcewicz M, Walendzik K, Machcińska S, Gawrońska-Kozak B. Adipose-Derived Stromal/Stem Cells from Large Animal Models: from Basic to Applied Science. Stem Cell Rev Rep 2020; 17:719-738. [PMID: 33025392 PMCID: PMC8166671 DOI: 10.1007/s12015-020-10049-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2020] [Indexed: 12/16/2022]
Abstract
Adipose-derived stem cells (ASCs) isolated from domestic animals fulfill the qualitative criteria of mesenchymal stem cells, including the capacity to differentiate along multiple lineage pathways and to self-renew, as well as immunomodulatory capacities. Recent findings on human diseases derived from studying large animal models, have provided evidence that administration of autologous or allogenic ASCs can improve the process of healing. In a narrow group of large animals used in bioresearch studies, pigs and horses have been shown to be the best suited models for study of the wound healing process, cardiovascular and musculoskeletal disorders. To this end, current literature demonstrates that ASC-based therapies bring considerable benefits to animal health in both spontaneously occurring and experimentally induced clinical cases. The purpose of this review is to provide an overview of the diversity, isolation, and characterization of ASCs from livestock. Particular attention has been paid to the functional characteristics of the cells that facilitate their therapeutic application in large animal models of human disease. In this regard, we describe outcomes of ASCs utilization in translational research with pig and horse models of disease. Furthermore, we evaluate the current status of ASC-based therapy in veterinary practice, particularly in the rapidly developing field of equine regenerative medicine. In conclusion, this review presents arguments that support the relevance of animal ASCs in the field of regenerative medicine and it provides insights into the future perspectives of ASC utilization in animal husbandry.
Collapse
Affiliation(s)
- Joanna Bukowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | | | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Sylwia Machcińska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Barbara Gawrońska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| |
Collapse
|
14
|
Kim KH, Park TS, Cho BW, Kim TM. Nanoparticles from Equine Fetal Bone Marrow-Derived Cells Enhance the Survival of Injured Chondrocytes. Animals (Basel) 2020; 10:ani10101723. [PMID: 32977476 PMCID: PMC7598183 DOI: 10.3390/ani10101723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/07/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Reports on the potential of using mesenchymal stem cells (MSCs) for treating equine degenerative joint diseases have been increasing over the past few years, in spite of limitations such as uncontrolled differentiation, immunogenicity, and tumorigenicity. We elucidated an allogenic alternative, where equine bone marrow-derived cells (BMC)-derived nanoparticles (BMC-NPs) can be used to promote the growth of chondrocytes, and simultaneously reduce their cytokine-induced apoptosis. The role of BMC-NPs in activation of signaling pathway molecules was also established. Abstract Recent studies have shown that mesenchymal stem cells (MSCs) can play a restorative role against degenerative joint diseases in horses. The purpose of this study was to investigate whether fetal bone marrow-derived cells (BMC)-derived nanoparticles (BMC-NPs) can stimulate the survival of equine chondrocytes. Equine fetal BMCs were isolated and characterized, and the role of BMC-NPs s in equine chondrocytes undergoing inflammatory cell death was examined. BMCs have several characteristics, such as the potential to differentiate into chondrocytes and osteocytes. Additionally, BMCs expressed immunoregulatory genes in response to treatment with tumor necrosis factor-alpha (TNF-α) and Interleukin 1 beta (IL-1β). We found that BMC-NPs were taken up by equine chondrocytes. Functionally, BMC-NPs promoted the growth of chondrocytes, and reduced apoptosis induced by inflammatory cytokines. Furthermore, we observed that BMC-NPs upregulated the phosphorylation of protein kinase B (Akt) in the presence of IL-1β, and reduced the phosphorylation of TNF-α-induced activation of extracellular signal-regulated kinase 1/2 (ERK1/2) in the chondrocytes. Cumulatively, our study demonstrated that equine fetal BMC-NPs have the potential to stimulate the survival of chondrocytes damaged by inflammatory cytokines. Thus, BMC-NPs may become an alternative cell-free allogenic therapeutic for degenerative joint diseases in horses.
Collapse
Affiliation(s)
- Ki Hoon Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do 25354, Korea; (K.H.K.); (T.S.P.)
| | - Tae Sub Park
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do 25354, Korea; (K.H.K.); (T.S.P.)
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do 25354, Korea
| | - Byung-Wook Cho
- Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University, Samnangin-ro 1268-50, Miryang, Gyeongsangnam-do 50463, Korea;
| | - Tae Min Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do 25354, Korea; (K.H.K.); (T.S.P.)
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do 25354, Korea
- Correspondence: ; Tel.: +82-33-339-5896
| |
Collapse
|
15
|
Quality control and immunomodulatory potential for clinical-grade equine bone marrow-derived mesenchymal stromal cells and conditioned medium. Res Vet Sci 2020; 132:407-415. [PMID: 32768869 DOI: 10.1016/j.rvsc.2020.07.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/10/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
This study aimed to assess the safety and reproducibility of cell therapy for its use in clinical practice. We performed immunophenotypic characterization of equine bone marrow-derived mesenchymal stromal cells (BMMSCs) by flow cytometry using CD90, CD19, CD14, CD105, CD45, and HLA-DR markers (n = 4); GTG banding cytogenetic analysis (n = 3); and microbiological quality control (n = 4). The immunomodulatory potentials of BMMSCs (n = 4) and its conditioned medium (CM, n = 3) were investigated by in vitro lymphocyte inhibition assay using phytohemagglutinin (PHA)-stimulated peripheral blood mononuclear cells (PBMCs). BMMSCs populations isolated from all animals showed high expression of CD90 and CD105, and low expression of CD19, CD4, CD45, and HLA-DR. Of the 60 metaphases analyzed, 5% presented aneuploidy on random chromosomes and no contamination was found based on microbiological analyses. Both treatments significantly inhibited lymphocyte proliferation (> 50%), compared with PHA-stimulated PBMCs (p < 0.0001). These promising results for BMMSCs and CM justify their potential as a therapeutic approach for inflammatory diseases. The techniques used in this study were effective in assessing the quality and determining the minimum criteria for the clinical use of BMMSCs in veterinary medicine.
Collapse
|
16
|
Jahanbakhsh A, Nourbakhsh MS, Bonakdar S, Shokrgozar MA, Haghighipour N. Evaluation of alginate modification effect on cell-matrix interaction, mechanotransduction and chondrogenesis of encapsulated MSCs. Cell Tissue Res 2020; 381:255-272. [PMID: 32405685 DOI: 10.1007/s00441-020-03216-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 04/04/2020] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) are promising cell candidates for cartilage regeneration. Furthermore, it is important to control the cell-matrix interactions that have a direct influence on cell functions. Providing an appropriate microenvironment for cell differentiation in response to exogenous stimuli is a critical step towards the clinical utilization of MSCs. In this study, hydrogels consisted of different proportions of alginates that were modified using gelatin, collagen type I and arginine-glycine-aspartic acid (RGD) and were evaluated regarding their effects on mesenchymal stem cells. The effect of applying hydrostatic pressure on MSCs encapsulated in collagen-modified alginate with and without chondrogenic medium was evaluated 7, 14 and 21 days after culture, which is a comprehensive evaluation of chondrogenesis in 3D hydrogels with mechanical and chemical stimulants. Alcian blue, safranin O and dimethyl methylene blue (DMMB) staining showed the chondrogenic phenotype of cells seeded in the collagen- and RGD-modified alginate hydrogels with the highest intensity after 21 days of culture. The results of real-time PCR for cartilage-specific extracellular matrix genes indicated the chondrogenic differentiation of MSCs in all hydrogels. Also, the synergic effects of chemical and mechanical stimuli are indicated. The highest expression levels of the studied genes were observed in the cells embedded in collagen-modified alginate by loading after 14 days of exposure to the chondrogenic medium. The effect of using IHP on encapsulated MSCs in modified alginate with collagen type I is equal or even higher than using TGF-beta on encapsulated cells. The results of immunohistochemical assessments also confirmed the real-time PCR data.
Collapse
Affiliation(s)
- Azadeh Jahanbakhsh
- Biomaterial Group, Faculty of New Sciences and Technologies, Semnan University, Semnan, Iran
| | - Mohammad Sadegh Nourbakhsh
- Biomaterial Group, Faculty of New Sciences and Technologies, Semnan University, Semnan, Iran.
- Faculty of Materials and Metallurgical Engineering, Semnan University, Semnan, Iran.
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | | | | |
Collapse
|
17
|
Silva M, Monteiro GA, Fialho AM, Bernardes N, da Silva CL. Conditioned Medium From Azurin-Expressing Human Mesenchymal Stromal Cells Demonstrates Antitumor Activity Against Breast and Lung Cancer Cell Lines. Front Cell Dev Biol 2020; 8:471. [PMID: 32733876 PMCID: PMC7363770 DOI: 10.3389/fcell.2020.00471] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022] Open
Abstract
Recently, cell-based therapies have been explored as a strategy to enhance the specificity of anticancer therapeutic agents. In this perspective, human mesenchymal stromal cells (MSC) hold a promising future as cell delivery systems for anticancer proteins due to their unique biological features. In this study, we engineered human MSC to secrete a human codon-optimized version of azurin (hazu), a bacterial protein that has demonstrated anticancer activity toward different cancer models both in vitro and in vivo. To this end, microporation was used to deliver plasmid DNA encoding azurin into MSC derived from bone marrow (BM) and umbilical cord matrix (UCM), leading to expression and secretion of hazu to the conditioned medium (CM). Engineered hazu-MSC were shown to preserve tumor tropism toward breast (MCF-7) and lung (A549) cancer cell lines, comparable to non-modified MSC. Azurin was detected in the CM of transfected MSC and, upon treatment with hazu-MSC-CM, we observed a decrease in cancer cell proliferation, migration, and invasion, and an increase in cell death for both cancer cell lines. Moreover, expression of azurin caused no changes in MSC expression profile of cytokines relevant in the context of cancer progression, thus suggesting that the antitumoral effects induced by hazu-MSC secretome might be due to the presence of azurin independently. In conclusion, data shown herein indicate that MSC-produced azurin in a CM configuration elicits an anticancer effect.
Collapse
Affiliation(s)
- Marília Silva
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Gabriel Amaro Monteiro
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Arsenio M Fialho
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno Bernardes
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudia Lobato da Silva
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
18
|
Fabrication and characterization of herbal drug enriched Guar galactomannan based nanofibrous mats seeded with GMSC's for wound healing applications. Int J Biol Macromol 2020; 148:737-749. [DOI: 10.1016/j.ijbiomac.2020.01.188] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/05/2019] [Accepted: 01/20/2020] [Indexed: 12/30/2022]
|
19
|
Kang JM, Rajangam T, Rhie JW, Kim SH. Characterization of cell signaling, morphology, and differentiation potential of human mesenchymal stem cells based on cell adhesion mechanism. J Cell Physiol 2020; 235:6915-6928. [PMID: 32017071 DOI: 10.1002/jcp.29587] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022]
Abstract
It is essential to characterize the cellular properties of mesenchymal stem cell populations to maintain quality specifications and control in regenerative medicine. Biofunctional materials have been designed as artificial matrices for the stimulation of cell adhesion and specific cellular functions. We have developed recombinant maltose-binding protein (MBP)-fused proteins as artificial adhesion matrices to control human mesenchymal stem cell (hMSC) fate by using an integrin-independent heparin sulfate proteoglycans-mediated cell adhesion. In this study, we characterize cell adhesion-dependent cellular behaviors of human adipose-derived stem cells (hASCs) and human bone marrow stem cells (hBMSCs). We used an MBP-fused basic fibroblast growth factor (MF)-coated surface and fibronectin (FN)-coated surface to restrict and support, respectively, integrin-mediated adhesion. The cells adhered to MF exhibited restricted actin cytoskeleton organization and focal adhesion kinase phosphorylation. The hASCs and hBMSCs exhibited different cytoplasmic projection morphologies on MF. Both hASCs and hBMSCs differentiated more dominantly into osteogenic cells on FN than on MF. In contrast, hASCs differentiated more dominantly into adipogenic cells on MF than on FN, whereas hBMSCs differentiated predominantly into adipogenic cells on FN. The results indicate that hASCs exhibit a competitive differentiation potential (osteogenesis vs. adipogenesis) that depends on the cell adhesion matrix, whereas hBMSCs exhibit both adipogenesis and osteogenesis in integrin-mediated adhesion and thus hBMSCs have noncompetitive differentiation potential. We suggest that comparing differentiation behaviors of hMSCs with the diversity of cell adhesion is an important way to characterize hMSCs for regenerative medicine.
Collapse
Affiliation(s)
- Jung-Mi Kang
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,Department of Biomedical Engineering, Korea University of Science and Technology, Daejeon, South Korea
| | - Thanavel Rajangam
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Jong-Won Rhie
- Department of Plastic Surgery, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,Department of Biomedical Engineering, Korea University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
20
|
Ang J, Lee YA, Raghothaman D, Jayaraman P, Teo KL, Khan FJ, Reuveny S, Chang YT, Kang NY, Oh S. Rapid Detection of Senescent Mesenchymal Stromal Cells by a Fluorescent Probe. Biotechnol J 2019; 14:e1800691. [PMID: 31218816 DOI: 10.1002/biot.201800691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 06/10/2019] [Indexed: 12/16/2022]
Abstract
Despite intense interest in human mesenchymal stromal cells (MSCs), monitoring of the progressive occurrence of senescence has been hindered by the lack of efficient detection tools. Here, the discovery of a novel MSC senescence-specific fluorescent probe (CyBC9) identified by a high-throughput screen is reported. Compared with the prototypical senescence-associated β-galactosidase (SA-β-gal) staining, the CyBC9 assay is rapid (2 h) and nontoxic and can thus be applied to live cells. It is shown that CyBC9 is able to stain early and late senescent populations both in monolayer- and in microcarrier-based cultures. Finally, to investigate the mechanism of CyBC9, colocalization assays are performed and it is found that CyBC9 is accumulated in the mitochondria of senescent MSCs presumably due to the loss of membrane potential. Taken together, it is expected that CyBC9 will be a useful tool to ameliorate cell therapy through rapid and early screening of senescent phenotypes in clinically relevant MSCs.
Collapse
Affiliation(s)
- Joshur Ang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| | - Yong-An Lee
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way#06-01 Centros, Singapore, 138668, Singapore
| | - Deepak Raghothaman
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| | - Premkumar Jayaraman
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| | - Kim L Teo
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| | - Fahima J Khan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| | - Young-Tae Chang
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way#06-01 Centros, Singapore, 138668, Singapore
| | - Nam-Young Kang
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way#06-01 Centros, Singapore, 138668, Singapore
| | - Steve Oh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way #02-02 Helios, Singapore, 138667, Singapore
| |
Collapse
|
21
|
Yang Q, Pinto VMR, Duan W, Paxton EE, Dessauer JH, Ryan W, Lopez MJ. In vitro Characteristics of Heterogeneous Equine Hoof Progenitor Cell Isolates. Front Bioeng Biotechnol 2019; 7:155. [PMID: 31355191 PMCID: PMC6637248 DOI: 10.3389/fbioe.2019.00155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
Damage to an ectodermal-mesodermal interface like that in the equine hoof and human finger nail bed can permanently alter tissue structure and associated function. The purpose of this study was to establish and validate in vitro culture of primary progenitor cell isolates from the ectodermal-mesodermal tissue junction in equine hooves, the stratum internum, with and without chronic inflammation known to contribute to lifelong tissue defects. The following were evaluated in hoof stratum internum cell isolates up to 5 cell passages (P): expansion capacity by cell doublings and doubling time; plasticity with multi-lineage differentiation and colony-forming unit (CFU) frequency percentage; immunophenotype with immunocytochemistry and flow cytometry; gene expression with RT-PCR; and ultrastructure with transmission electron microscopy. The presence of keratin (K)14, 15 and K19 as well as cluster of differentiation (CD)44 and CD29 was determined in situ with immunohistochemistry. To confirm in vivo extracellular matrix (ECM) formation, cell-scaffold (polyethylene glycol/poly-L-lactic acid and tricalcium phosphate/hydroxyapatite) constructs were evaluated with scanning electron microscopy 9 weeks after implantation in athymic mice. Cultured cells had characteristic progenitor cell morphology, expansion, CFU frequency percentage and adipocytic, osteoblastic, and neurocytic differentiation capacity. CD44, CD29, K14, K15 and K19 proteins were present in native hoof stratum internum. Cultured cells also expressed K15, K19 and desmogleins 1 and 3. Gene expression of CD105, CD44, K14, K15, sex determining region Y-box 2 (SOX2) and octamer-binding transcription factor 4 (OCT4) was confirmed in vitro. Cultured cells had large, eccentric nuclei, elongated mitochondria, and intracellular vacuoles. Scaffold implants with cells contained fibrous ECM 9 weeks after implantation compared to little or none on acellular scaffolds. In vitro expansion and plasticity and in vivo ECM deposition of heterogeneous, immature cell isolates from the ectodermal-mesodermal tissue interface of normal and chronically inflamed hooves are typical of primary cell isolates from other adult tissues, and they appear to have both mesodermal and ectodermal qualities in vitro. These results establish a unique cell culture model to target preventative and restorative therapies for ectodermal-mesodermal tissue junctions.
Collapse
Affiliation(s)
- Qingqiu Yang
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Vanessa Marigo Rocha Pinto
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Wei Duan
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Erica E Paxton
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Jenna H Dessauer
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - William Ryan
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
22
|
Parathyroid Hormone-Related Protein (PTHrP) Accelerates Soluble RANKL Signals for Downregulation of Osteogenesis of Bone Mesenchymal Stem Cells. J Clin Med 2019; 8:jcm8060836. [PMID: 31212822 PMCID: PMC6616973 DOI: 10.3390/jcm8060836] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/28/2019] [Accepted: 06/07/2019] [Indexed: 01/17/2023] Open
Abstract
A recent study reported the expression of receptor activator of nuclear factor-κB (RANK) in mesenchymal stem cells (MSCs) surface that negatively regulates osteogenesis of MSCs. Empirical evidence from the previous study confirmed the role of parathyroid hormone-related protein (PTHrP) in osteoblastogenesis. However, it is necessary to understand the paracrine role of PTHrP and RANKL for osteogenesis in order to explore the hidden secrets in bone biology. Considering the above concept, paracrine cues of soluble-receptor activator of nuclear factor-κB ligand (sRANKL) and PTHrP in osteogenic differentiation of MSCs were investigated. Our results confirmed that sRANKL increased the expression of surface-RANK in MSCs at the earlier stage of osteogenesis, which was downregulated later in differentiated MSCs. In contrast, RANKL expression was low at the earlier stage of MSCs proliferation and high at the differentiation stage of MSCs, which may play a fundamental role in osteoclast formation. sRANKL downregulated osteogenesis of MSCs by decreasing progressive ankylosis (ANK) protein expression while PTHrP upregulated the osteogenic exploitive effect of sRANKL. Interestingly, when they were co-cultured with MSCs, T-lymphocytes expressed high membrane-RANKL levels that contribute to osteogenesis inhibition during MSC differentiation. Thus, our results disclose that sRANKL treatment downregulates osteogenesis of MSCs by increasing RANK expression at the earlier stage of differentiation and by inhibiting ANK. Further, we demonstrated that PTHrP accelerated the downregulating osteogenic effect of sRANKL.
Collapse
|
23
|
Duan W, Lopez MJ. Effects of enzyme and cryoprotectant concentrations on yield of equine adipose-derived multipotent stromal cells. Am J Vet Res 2019; 79:1100-1112. [PMID: 30256145 DOI: 10.2460/ajvr.79.10.1100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate effects of various concentrations of collagenase and dimethyl sulfoxide (DMSO) on yield of equine adipose-derived multipotent stromal cells (ASCs) before and after cryopreservation. SAMPLE Supragluteal subcutaneous adipose tissue from 7 Thoroughbreds. PROCEDURES Tissues were incubated with digests containing 0.1%, 0.05%, or 0.025% type I collagenase. Part of each resulting stromal vascular fraction was cryopreserved in 80% fetal bovine serum (FBS), 10% DMSO, and 10% Dulbecco modified Eagle medium F-12 and in 95% FBS and 5% DMSO. Half of each fresh and cryopreserved heterogeneous cell population was not immunophenotyped (unsorted) or was immunophenotyped for CD44+, CD105+, and major histocompatability complex class II (MHCII; CD44+-CD105+-MHCII+ cells and CD44+-CD105+-MHCII- cells). Cell proliferation (cell viability assay), plasticity (CFU frequency), and lineage-specific target gene and oncogene expression (reverse transcriptase PCR assays) were determined in passage 1 cells before and after culture in induction media. RESULTS Digestion with 0.1% collagenase yielded the highest number of nucleated cells. Cell surface marker expression and proliferation rate were not affected by collagenase concentration. Cryopreservation reduced cell expansion rate and CD44+-CD105+-MHCII- CFUs; it also reduced osteogenic plasticity of unsorted cells. However, effects appeared to be unrelated to DMSO concentrations. There were also variable effects on primordial gene expression among cell isolates. CONCLUSIONS AND CLINICAL RELEVANCE Results supported the use of 0.1% collagenase in an adipose tissue digest and 5% DMSO in cryopreservation medium for isolation and cryopreservation, respectively, of equine ASCs. These results may be used as guidelines for standardization of isolation and cryopreservation procedures for equine ASCs.
Collapse
|
24
|
Abstract
Present bioprinting techniques lack the methodology to print with bioactive materials that retain their biological functionalities. This constraint is due to the fact that extrusion-based printing of synthetic polymers is commonly performed at very high temperatures in order to achieve desired mechanical properties and printing resolutions. Consequently, current methodology prevents printing scaffolds embedded with bioactive molecules, such as growth factors. With the wide use of mesenchymal stem cells (MSCs) in regenerative medicine research, the integration of growth factors into 3D printed scaffolds is critical because it can allow for inducible MSC differentiation. We have successfully incorporated growth factors into extrusion printed poly (lactic-co-glycolic acid) (PLGA) scaffolds by introducing dimethyl sulfoxide (DMSO) for low temperature printing. Mechanical testing results demonstrated significantly different compressive and tensile properties for PLGA scaffold printed with or without DMSO. In particular, the PLGA-DMSO scaffold displayed a highly stretchable feature compared to the regular PLGA scaffold. The cellular response of growth factor introduction was evaluated in vitro using human mesenchymal stem cells (hMSCs). By evaporating the DMSO after printing, we ensured that there was no cytotoxic effect on seeded hMSCs. The addition of lineage specific growth factors led to increased expression of corresponding genetic markers for chondrogenesis, osteogenesis, and adipogenesis. We concluded that the use of DMSO for 3D printed scaffold fabrication with bioactive items is a revolutionary methodology in advancing regenerative medicine. The incorporation of bioactive molecules opens pathways to more therapeutic uses for 3D printing in treating damaged or deteriorating native tissue.
Collapse
|
25
|
Gugjoo MB, Amarpal, Makhdoomi DM, Sharma GT. Equine Mesenchymal Stem Cells: Properties, Sources, Characterization, and Potential Therapeutic Applications. J Equine Vet Sci 2018; 72:16-27. [PMID: 30929778 DOI: 10.1016/j.jevs.2018.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 09/06/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Properties like sustained multiplication and self-renewal, and homing and multilineage differentiation to undertake repair of the damaged tissues make stem cells the lifeline for any living system. Therefore, stem cell therapy is regarded to carry immense therapeutic potential. Though the dearth of understanding about the basic biological properties and pathways involved in therapeutic benefits currently limit the application of stem cells in humans as well as animals, there are innumerable reports that suggest clinical benefits of stem cell therapy in equine. Among various stem cell sources, currently adult mesenchymal stem cells (MSCs) are preferred for therapeutic application in horse owing to their easy availability, capacity to modulate inflammation, and promote healing. Also the cells carry very limited teratogenic risk compared to the pluripotent stem cells. Mesenchymal stem cells were earlier considered mainly for musculoskeletal tissues, but now may also be utilized in other diverse clinical problems in horse, and the results may be extrapolated even for human medicine. The current review highlights biological properties, sources, mechanisms, and potential therapeutic applications of stem cells in equine practice.
Collapse
Affiliation(s)
- Mudasir Bashir Gugjoo
- Division of Surgery, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India.
| | - Amarpal
- Division of Surgery, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India
| | - Dil Mohammad Makhdoomi
- Division of Surgery, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar, J&K, India
| | - Gutulla Taru Sharma
- Division of Physiology and Climatology, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India
| |
Collapse
|
26
|
Barrachina L, Remacha AR, Romero A, Zaragoza P, Vázquez FJ, Rodellar C. Differentiation of equine bone marrow derived mesenchymal stem cells increases the expression of immunogenic genes. Vet Immunol Immunopathol 2018; 200:1-6. [DOI: 10.1016/j.vetimm.2018.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/14/2018] [Accepted: 04/09/2018] [Indexed: 12/27/2022]
|
27
|
Scheinpflug J, Pfeiffenberger M, Damerau A, Schwarz F, Textor M, Lang A, Schulze F. Journey into Bone Models: A Review. Genes (Basel) 2018; 9:E247. [PMID: 29748516 PMCID: PMC5977187 DOI: 10.3390/genes9050247] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/24/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
Bone is a complex tissue with a variety of functions, such as providing mechanical stability for locomotion, protection of the inner organs, mineral homeostasis and haematopoiesis. To fulfil these diverse roles in the human body, bone consists of a multitude of different cells and an extracellular matrix that is mechanically stable, yet flexible at the same time. Unlike most tissues, bone is under constant renewal facilitated by a coordinated interaction of bone-forming and bone-resorbing cells. It is thus challenging to recreate bone in its complexity in vitro and most current models rather focus on certain aspects of bone biology that are of relevance for the research question addressed. In addition, animal models are still regarded as the gold-standard in the context of bone biology and pathology, especially for the development of novel treatment strategies. However, species-specific differences impede the translation of findings from animal models to humans. The current review summarizes and discusses the latest developments in bone tissue engineering and organoid culture including suitable cell sources, extracellular matrices and microfluidic bioreactor systems. With available technology in mind, a best possible bone model will be hypothesized. Furthermore, the future need and application of such a complex model will be discussed.
Collapse
Affiliation(s)
- Julia Scheinpflug
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| | - Moritz Pfeiffenberger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany.
| | - Alexandra Damerau
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany.
| | - Franziska Schwarz
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| | - Martin Textor
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| | - Annemarie Lang
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany.
| | - Frank Schulze
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| |
Collapse
|
28
|
Duan W, Chen C, Haque M, Hayes D, Lopez MJ. Polymer-mineral scaffold augments in vivo equine multipotent stromal cell osteogenesis. Stem Cell Res Ther 2018. [PMID: 29523214 PMCID: PMC5845133 DOI: 10.1186/s13287-018-0790-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Use of bioscaffolds to direct osteogenic differentiation of adult multipotent stromal cells (MSCs) without exogenous proteins is a contemporary approach to bone regeneration. Identification of in vivo osteogenic contributions of exogenous MSCs on bioscaffolds after long-term implantation is vital to understanding cell persistence and effect duration. Methods This study was designed to quantify in vivo equine MSC osteogenesis on synthetic polymer scaffolds with distinct mineral combinations 9 weeks after implantation in a murine model. Cryopreserved, passage (P)1, equine bone marrow-derived MSCs (BMSC) and adipose tissue-derived MSCs (ASC) were culture expanded to P3 and immunophenotyped with flow cytometry. They were then loaded by spinner flask on to scaffolds composed of tricalcium phosphate (TCP)/hydroxyapatite (HA) (40:60; HT), polyethylene glycol (PEG)/poly-l-lactic acid (PLLA) (60:40; GA), or PEG/PLLA/TCP/HA (36:24:24:16; GT). Scaffolds with and without cells were maintained in static culture for up to 21 days or implanted subcutaneously in athymic mice that were radiographed every 3 weeks up to 9 weeks. In vitro cell viability and proliferation were determined. Explant composition (double-stranded (ds)DNA, collagen, sulfated glycosaminoglycan (sGAG), protein), equine and murine osteogenic target gene expression, microcomputed tomography (μCT) mineralization, and light microscopic structure were assessed. Results The ASC and BMSC number increased significantly in HT constructs between 7 and 21 days of culture, and BMSCs increased similarly in GT constructs. Radiographic opacity increased with time in GT-BMSC constructs. Extracellular matrix (ECM) components and dsDNA increased significantly in GT compared to HT constructs. Equine and murine osteogenic gene expression was highest in BMSC constructs with mineral-containing scaffolds. The HT constructs with either cell type had the highest mineral deposition based on μCT. Regardless of composition, scaffolds with cells had more ECM than those without, and osteoid was apparent in all BMSC constructs. Conclusions In this study, both exogenous and host MSCs appear to contribute to in vivo osteogenesis. Addition of mineral to polymer scaffolds enhances equine MSC osteogenesis over polymer alone, but pure mineral scaffold provides superior osteogenic support. These results emphasize the need for bioscaffolds that provide customized osteogenic direction of both exo- and endogenous MSCs for the best regenerative potential.
Collapse
Affiliation(s)
- Wei Duan
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Cong Chen
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Masudul Haque
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Daniel Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
29
|
Duan W, Lopez MJ. Canine Adult Adipose Tissue-Derived Multipotent Stromal Cell Isolation and Characterization. Methods Mol Biol 2018; 1773:189-202. [PMID: 29687391 DOI: 10.1007/978-1-4939-7799-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adult multipotent stromal cells (MSCs) are increasingly popular for direct therapeutic applications and bioengineering. Canine patients constitute a major component of veterinary practice, and the dog is an established preclinical animal model for numerous traumatic, degenerative, and disease conditions. Current information supports the presence and relative abundance of adipose tissue-derived multipotent stromal cells (ASCs) in various canine adipose tissue depots. Refined isolation and characterization techniques contribute to collective knowledge of ASC phenotypes and subpopulations for customized, targeted applications. Continued efforts to augment understanding of canine ASCs is critical to progressive treatment advances and high-impact study outcomes. This chapter contains a description of techniques to isolate and characterize canine ASCs.
Collapse
Affiliation(s)
- Wei Duan
- Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
30
|
Duan W, Haque M, Kearney MT, Lopez MJ. Collagen and Hydroxyapatite Scaffolds Activate Distinct Osteogenesis Signaling Pathways in Adult Adipose-Derived Multipotent Stromal Cells. Tissue Eng Part C Methods 2017; 23:592-603. [PMID: 28877641 PMCID: PMC5653142 DOI: 10.1089/ten.tec.2017.0078] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022] Open
Abstract
Osteogenic cell signaling pathway disruption varies among bone diseases. This investigation was designed to identify adipose-derived multipotent stromal cell (ASC) and bone graft scaffold combinations for local, targeted restoration of gene expression and extracellular matrix (ECM) deposition. Human ASC osteogenesis on bone graft materials was quantified following culture in stromal (S), osteogenic (O), or osteogenic for 48 h followed by stromal medium (OS) to test the two-part hypothesis: (1) identical ASC isolates on distinct bone graft scaffolds demonstrate unique viability, differentiation, ECM production, and gene expression in the same culture conditions; (2) identical ASC-bone graft scaffold combinations have different cell viability, differentiation, ECM production, and gene expression when cultured in S, O, or OS medium. Three commercially available bone graft scaffold materials, type I bovine collagen (C), hydroxyapatite + β-tricalcium phosphate + type I bovine collagen (HT), and β-tricalcium phosphate + type I bovine collagen (CT) were evaluated. Passage 3 ASCs were loaded onto scaffold blocks with a spinner flask bioreactor, and constructs were cultured up to 28 days. Cell viability, gene expression (alkaline phosphatase [ALPL], osteoprotegerin [TNFRSF11B], osteocalcin [BGLAP], cannabinoid receptors type I [CNR1] and II [CNR2], receptor activator of nuclear factor kappa β ligand [TNFSF11]), as well as ECM DNA, collagen, sulfated glycosaminoglycan, and protein content were quantified. Matrix organization was evaluated with scanning electron microscopy. Effects of scaffold, medium, or culture duration on cell viability were minimal. Significantly higher initial ALPL expression decreased with time, while BGLAP expression increased in HT constructs in O medium, and the constructs had the most abundant ECM components and ultrastructural organization. There was a similar, although delayed, pattern of gene expression and greater ECM collagen with less organization in C constructs in O medium. Higher CNR1 expression in C versus higher TNFRSF11B/TNFSF11 expression in HT constructs throughout the study support stimulation of unique osteogenic signaling pathways by identical cell isolates. These results suggest that bone scaffold composition may be used to selectively target specific osteogenic cell signaling pathways in ASC constructs to stimulate ECM deposition based on therapeutic needs.
Collapse
Affiliation(s)
- Wei Duan
- 1 Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| | - Masudul Haque
- 1 Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| | - Michael T Kearney
- 2 Department of Pathobiological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Mandi J Lopez
- 1 Laboratory for Equine and Comparative Orthopedic Research, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
31
|
Raut A, Khanna A. High-throughput sequencing to identify microRNA signatures during hepatic differentiation of human umbilical cord Wharton's jelly-derived mesenchymal stem cells. Hepatol Res 2017; 47:910-927. [PMID: 27653181 DOI: 10.1111/hepr.12820] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/10/2016] [Accepted: 09/17/2016] [Indexed: 12/27/2022]
Abstract
AIM MicroRNAs (miRNAs) constitute a class of small non-coding RNAs involved in regulation of cognate mRNAs post-transcriptionally. MicroRNAs have been implicated in regulating the stem cell differentiation process. Limited regulatory miRNAs have been reported to date during hepatic differentiation of stem cells. The present study was designed to identify the signature miRNAs implicated in hepatic differentiation of stem cells using next-generation sequencing methods. METHODS We undertook sequencing of miRNAs isolated from three different time points during hepatic differentiation of human umbilical cord Wharton's jelly-derived mesenchymal stem cells (hUC-MSCs) from two biological replicates. RESULTS Out of a total known 2588 miRNAs (according to miRBase version 21), 880 miRNAs were identified in our study. A total of 63 significantly expressed miRNAs during hepatic differentiation, with at least 2-fold change and a false discovery rate value <0.05, were considered for further analysis. The putative target genes of significantly downregulated miRNAs during hepatic differentiation appeared to be mostly associated with biological processes that are essential for hepatic differentiation and maintenance of mature hepatic phenotype-like liver development, stem cell differentiation, Wnt receptor signaling pathway, and drug and cholesterol metabolic processes. Putative target genes of significantly upregulated miRNAs are highly enriched in regulating processes that block hepatic differentiation of hUC-MSCs like epithelial-mesenchymal transition, transforming growth factor-β receptor signaling pathway, and stem cell maintenance. CONCLUSION The study provides a new insight for investigation of miRNA-regulated pathways during the differentiation process.
Collapse
Affiliation(s)
- Akshata Raut
- Department of Biological Sciences, Sunandan Divatia School of Science, Narsee Monjee Institute of Management Studies University, Mumbai, India
| | - Aparna Khanna
- Department of Biological Sciences, Sunandan Divatia School of Science, Narsee Monjee Institute of Management Studies University, Mumbai, India
| |
Collapse
|
32
|
Colbath AC, Frisbie DD, Dow SW, Kisiday JD, McIlwraith CW, Goodrich LR. Equine Models for the Investigation of Mesenchymal Stem Cell Therapies in Orthopaedic Disease. OPER TECHN SPORT MED 2017. [DOI: 10.1053/j.otsm.2016.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
33
|
Liu S, Zhou J, Zhang X, Liu Y, Chen J, Hu B, Song J, Zhang Y. Strategies to Optimize Adult Stem Cell Therapy for Tissue Regeneration. Int J Mol Sci 2016; 17:ijms17060982. [PMID: 27338364 PMCID: PMC4926512 DOI: 10.3390/ijms17060982] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 12/13/2022] Open
Abstract
Stem cell therapy aims to replace damaged or aged cells with healthy functioning cells in congenital defects, tissue injuries, autoimmune disorders, and neurogenic degenerative diseases. Among various types of stem cells, adult stem cells (i.e., tissue-specific stem cells) commit to becoming the functional cells from their tissue of origin. These cells are the most commonly used in cell-based therapy since they do not confer risk of teratomas, do not require fetal stem cell maneuvers and thus are free of ethical concerns, and they confer low immunogenicity (even if allogenous). The goal of this review is to summarize the current state of the art and advances in using stem cell therapy for tissue repair in solid organs. Here we address key factors in cell preparation, such as the source of adult stem cells, optimal cell types for implantation (universal mesenchymal stem cells vs. tissue-specific stem cells, or induced vs. non-induced stem cells), early or late passages of stem cells, stem cells with endogenous or exogenous growth factors, preconditioning of stem cells (hypoxia, growth factors, or conditioned medium), using various controlled release systems to deliver growth factors with hydrogels or microspheres to provide apposite interactions of stem cells and their niche. We also review several approaches of cell delivery that affect the outcomes of cell therapy, including the appropriate routes of cell administration (systemic, intravenous, or intraperitoneal vs. local administration), timing for cell therapy (immediate vs. a few days after injury), single injection of a large number of cells vs. multiple smaller injections, a single site for injection vs. multiple sites and use of rodents vs. larger animal models. Future directions of stem cell-based therapies are also discussed to guide potential clinical applications.
Collapse
Affiliation(s)
- Shan Liu
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Jingli Zhou
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Xuan Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Yang Liu
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Jin Chen
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Bo Hu
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Jinlin Song
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing 401147, China.
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| |
Collapse
|
34
|
Pustlauk W, Paul B, Gelinsky M, Bernhardt A. Jellyfish collagen and alginate: Combined marine materials for superior chondrogenesis of hMSC. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 64:190-198. [PMID: 27127044 DOI: 10.1016/j.msec.2016.03.081] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/28/2016] [Accepted: 03/22/2016] [Indexed: 11/15/2022]
Abstract
Marine, hybrid constructs of porous scaffolds from fibrillized jellyfish collagen and alginate hydrogel are mimicking both of the main tissue components of cartilage, thus being a promising approach for chondrogenic differentiation of human mesenchymal stem cells (hMSC). Investigating their potential for articular cartilage repair, the present study examined scaffolds being either infiltrated with an alginate-cell-suspension (ACS) or seeded with hMSC and embedded in alginate after cell adhesion (EAS). Hybrid constructs with 2×10(5) and 4.5×10(5)hMSC/scaffold were compared to hMSC encapsulated in pure alginate discs, both chondrogenically stimulated for 21days. Typical round, chondrocyte-like morphology was observed in pure alginate gels and ACS scaffolds, while cells in EAS were elongated and tightly attached to the collagen pores. Col 2 gene expression was comparable in all scaffold types examined. However, the Col 2/Col 1 ratio was higher for pure alginate discs and ACS scaffolds compared to EAS. In contrast, cells in EAS scaffolds displayed higher gene expression of Sox 9, Col 11 and ACAN compared to ACS and pure alginate. Secretion of sulfated glycosaminoglycans (sGAG) was comparable for ACS and EAS scaffolds. In conclusion hybrid constructs of jellyfish collagen and alginate support hMSC chondrogenic differentiation and provide more stable and constructs compared to pure hydrogels.
Collapse
Affiliation(s)
- W Pustlauk
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty Carl Gustav Carus of Technische Universität Dresden, Fetscher Str. 74, 01307 Dresden, Germany
| | - B Paul
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty Carl Gustav Carus of Technische Universität Dresden, Fetscher Str. 74, 01307 Dresden, Germany
| | - M Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty Carl Gustav Carus of Technische Universität Dresden, Fetscher Str. 74, 01307 Dresden, Germany
| | - A Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty Carl Gustav Carus of Technische Universität Dresden, Fetscher Str. 74, 01307 Dresden, Germany.
| |
Collapse
|
35
|
Panadero J, Lanceros-Mendez S, Ribelles JG. Differentiation of mesenchymal stem cells for cartilage tissue engineering: Individual and synergetic effects of three-dimensional environment and mechanical loading. Acta Biomater 2016; 33:1-12. [PMID: 26826532 DOI: 10.1016/j.actbio.2016.01.037] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 12/17/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022]
Abstract
Chondrogenesis of dedifferentiated chondrocytes and mesenchymal stem cells is influenced not only by soluble molecules like growth factors, but also by the cell environment itself. The latter is achieved through both mechanical cues - which act as stimulation factor and influences nutrient transport - and adhesion to extracellular matrix cues - which determine cell shape. Although the effects of soluble molecules and cell environment have been intensively addressed, few observations and conclusions about the interaction between the two have been achieved. In this work, we review the state of the art on the single effects between mechanical and biochemical cues, as well as on the combination of the two. Furthermore, we provide a discussion on the techniques currently used to determine the mechanical properties of materials and tissues generated in vitro, their limitations and the future research needs to properly address the identified problems. STATEMENT OF SIGNIFICANCE The importance of biomechanical cues in chondrogenesis is well known. This paper reviews the existing literature on the effect of mechanical stimulation on chondrogenic differentiation of mesenchymal stem cells in order to regenerate hyaline cartilage. Contradictory results found with respect to the effect of different modes of external loading can be explained by the different properties of the scaffolding system that holds the cells, which determine cell adhesion and morphology and spatial distribution of cells, as well as the stress transmission to the cells. Thus, this review seeks to provide an insight into the interplay between external loading program and scaffold properties during chondrogenic differentiation. The review of the literature reveals an important gap in the knowledge in this field and encourages new experimental studies. The main issue is that in each of the few cases in which the interplay is investigated, just two groups of scaffolds are compared, leaving intermediate adhesion conditions out of study. The authors propose broader studies implementing new high-throughput techniques for mechanical characterization of tissue engineering constructs and the inclusion of fatigue analysis as support methodology to more exhaustive mechanical characterization.
Collapse
|
36
|
Pizzute T, Lynch K, Pei M. Impact of tissue-specific stem cells on lineage-specific differentiation: a focus on the musculoskeletal system. Stem Cell Rev Rep 2015; 11:119-32. [PMID: 25113801 DOI: 10.1007/s12015-014-9546-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tissue-specific stem cells are found throughout the body and, with proper intervention and environmental cues, these stem cells exercise their capabilities for differentiation into several lineages to form cartilage, bone, muscle, and adipose tissue in vitro and in vivo. Interestingly, it has been widely demonstrated that they do not differentiate with the same efficacy during lineage-specific differentiation studies, as the tissue-specific stem cells are generally more effective when differentiating toward the tissues from which they were derived. This review focuses on four mesodermal lineages for tissue-specific stem cell differentiation: adipogenesis, chondrogenesis, myogenesis, and osteogenesis. It is intended to give insight into current multilineage differentiation and comparative research, highlight and contrast known trends regarding differentiation, and introduce supporting evidence which demonstrates particular tissue-specific stem cells' superiority in lineage-specific differentiation, along with their resident tissue origins and natural roles. In addition, some epigenetic and transcriptomic differences between stem cells which may explain the observed trends are discussed.
Collapse
Affiliation(s)
- Tyler Pizzute
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | | | | |
Collapse
|
37
|
Pustlauk W, Paul B, Brueggemeier S, Gelinsky M, Bernhardt A. Modulation of chondrogenic differentiation of human mesenchymal stem cells in jellyfish collagen scaffolds by cell density and culture medium. J Tissue Eng Regen Med 2015; 11:1710-1722. [PMID: 26178016 DOI: 10.1002/term.2065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 05/05/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
Abstract
Studies on tissue-engineering approaches for the regeneration of traumatized cartilage focus increasingly on multipotent human mesenchymal stem cells (hMSCs) as an alternative to autologous chondrocytes. The present study applied porous scaffolds made of collagen from the jellyfish Rhopilema esculentum for the in vitro chondrogenic differentiation of hMSCs. Culture conditions in those scaffolds differ from conditions in high-density pellet cultures, making a re-examination of these data necessary. We systematically investigated the influence of seeding density, basic culture media [Dulbecco's modified Eagle's medium (DMEM), α-minimum essential medium (α-MEM)] with varying glucose content and supplementation with fetal calf serum (FCS) or bovine serum albumin (BSA) on the chondrogenic differentiation of hMSCs. Gene expression analyses of selected markers for chondrogenic differentiation and hypertrophic development were conducted. Furthermore, the production of cartilage extracellular matrix (ECM) was analysed by quantification of sulphated glycosaminoglycan and collagen type II contents. The strongest upregulation of chondrogenic markers, along with the highest ECM deposition was observed in scaffolds seeded with 2.4 × 106 cells/cm3 after cultivation in high-glucose DMEM and 0.125% BSA. Lower seeding densities compared to high-density pellet cultures were sufficient to induce in vitro chondrogenic differentiation of hMSCs in collagen scaffolds, which reduces the amount of cells required for the seeding of scaffolds and thus the monolayer expansion period. Furthermore, examination of the impact of FCS and α-MEM on chondrogenic MSC differentiation is an important prerequisite for the development of an osteochondral medium for simultaneous osteogenic and chondrogenic differentiation in biphasic scaffolds for osteochondral tissue regeneration. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- W Pustlauk
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - B Paul
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - S Brueggemeier
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - M Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - A Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| |
Collapse
|
38
|
Correia V, Panadero JA, Ribeiro C, Sencadas V, Rocha JG, Gomez Ribelles JL, Lanceros-Méndez S. Design and validation of a biomechanical bioreactor for cartilage tissue culture. Biomech Model Mechanobiol 2015; 15:471-8. [PMID: 26153426 DOI: 10.1007/s10237-015-0698-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/26/2015] [Indexed: 11/28/2022]
Abstract
Specific tissues, such as cartilage, undergo mechanical solicitation under their normal performance in human body. In this sense, it seems necessary that proper tissue engineering strategies of these tissues should incorporate mechanical solicitations during cell culture, in order to properly evaluate the influence of the mechanical stimulus. This work reports on a user-friendly bioreactor suitable for applying controlled mechanical stimulation--amplitude and frequency--to three-dimensional scaffolds. Its design and main components are described, as well as its operation characteristics. The modular design allows easy cleaning and operating under laminar hood. Different protocols for the sterilization of the hermetic enclosure are tested and ensure lack of observable contaminations, complying with the requirements to be used for cell culture. The cell viability study was performed with KUM5 cells.
Collapse
Affiliation(s)
- V Correia
- Centro/Departamento de Física da Universidade do Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,Algoritmi Research Centre, Universidade do Minho, Campus de Azurém, 4800-058, Guimarães, Portugal
| | - J A Panadero
- Centro/Departamento de Física da Universidade do Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - C Ribeiro
- Centro/Departamento de Física da Universidade do Minho, Campus de Gualtar, 4710-057, Braga, Portugal.
| | - V Sencadas
- Centro/Departamento de Física da Universidade do Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - J G Rocha
- Algoritmi Research Centre, Universidade do Minho, Campus de Azurém, 4800-058, Guimarães, Portugal
| | - J L Gomez Ribelles
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain.,Ciber en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valencia, Spain
| | - S Lanceros-Méndez
- Centro/Departamento de Física da Universidade do Minho, Campus de Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
39
|
Marx C, Silveira MD, Beyer Nardi N. Adipose-derived stem cells in veterinary medicine: characterization and therapeutic applications. Stem Cells Dev 2015; 24:803-13. [PMID: 25556829 DOI: 10.1089/scd.2014.0407] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells, considered one of the most promising cell types for therapeutic applications due to their capacity to secrete regenerative bioactive molecules, are present in all tissues. Stem cells derived from the adipose tissue have been increasingly used for cell therapy in humans and animals, both as freshly isolated, stromal vascular fraction (SVF) cells, or as cultivated adipose-derived stem cells (ASCs). ASCs have been characterized in different animal species for proliferation, differentiation potential, immunophenotype, gene expression, and potential for tissue engineering. Whereas canine and equine ASCs are well studied, feline cells are still poorly known. Many companies around the world offer ASC therapy for dogs, cats, and horses, although in most countries these activities are not yet controlled by regulatory agencies. This is the first study to review the characterization and clinical use of SVF and ASCs in spontaneously occurring diseases in veterinary patients. Although a relatively large number of studies investigating ASC therapy in induced lesions are available in the literature, a surprisingly small number of reports describe ASC therapy for naturally affected dogs, cats, and horses. A total of seven studies were found with dogs, only two studies in cats, and four in horses. Taken as a whole, the results do not allow a conclusion on the effect of this therapy, due to the generally small number of patients included, diversity of cell populations used, and lack of adequate controls. Further controlled studies are clearly needed to establish the real potential of ASC in veterinary medicine.
Collapse
Affiliation(s)
- Camila Marx
- 1 Laboratory of Stem Cells and Tissue Engineering, Universidade Luterana do Brasil , Canoas, RS, Brazil
| | | | | |
Collapse
|
40
|
Lopez MJ, Jarazo J. State of the art: stem cells in equine regenerative medicine. Equine Vet J 2014; 47:145-54. [PMID: 24957845 DOI: 10.1111/evj.12311] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 06/01/2014] [Indexed: 12/20/2022]
Abstract
According to Greek mythology, Prometheus' liver grew back nightly after it was removed each day by an eagle as punishment for giving mankind fire. Hence, contrary to popular belief, the concept of tissue and organ regeneration is not new. In the early 20th century, cell culture and ex vivo organ preservation studies by Alexis Carrel, some with famed aviator Charles Lindbergh, established a foundation for much of modern regenerative medicine. While early beliefs and discoveries foreshadowed significant accomplishments in regenerative medicine, advances in knowledge within numerous scientific disciplines, as well as nano- and micromolecular level imaging and detection technologies, have contributed to explosive advances over the last 20 years. Virtually limitless preparations, combinations and applications of the 3 major components of regenerative medicine, namely cells, biomaterials and bioactive molecules, have created a new paradigm of future therapeutic options for most species. It is increasingly clear, however, that despite significant parallels among and within species, there is no 'one-size-fits-all' regenerative therapy. Likewise, a panacea has yet to be discovered that completely reverses the consequences of time, trauma and disease. Nonetheless, there is no question that the promise and potential of regenerative medicine have forever altered medical practices. The horse is a relative newcomer to regenerative medicine applications, yet there is already a large body of work to incorporate novel regenerative therapies into standard care. This review focuses on the current state and potential future of stem cells in equine regenerative medicine.
Collapse
Affiliation(s)
- M J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Equine Health Studies Program, Department of Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, USA
| | | |
Collapse
|
41
|
Multiscale fluid-structure interaction modelling to determine the mechanical stimulation of bone cells in a tissue engineered scaffold. Biomech Model Mechanobiol 2014; 14:231-43. [PMID: 24903125 DOI: 10.1007/s10237-014-0599-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 05/22/2014] [Indexed: 10/25/2022]
Abstract
Recent studies have shown that mechanical stimulation, by means of flow perfusion and mechanical compression (or stretching), enhances osteogenic differentiation of mesenchymal stem cells and bone cells within biomaterial scaffolds in vitro. However, the precise mechanisms by which such stimulation enhances bone regeneration is not yet fully understood. Previous computational studies have sought to characterise the mechanical stimulation on cells within biomaterial scaffolds using either computational fluid dynamics or finite element (FE) approaches. However, the physical environment within a scaffold under perfusion is extremely complex and requires a multiscale and multiphysics approach to study the mechanical stimulation of cells. In this study, we seek to determine the mechanical stimulation of osteoblasts seeded in a biomaterial scaffold under flow perfusion and mechanical compression using multiscale modelling by two-way fluid-structure interaction and FE approaches. The mechanical stimulation, in terms of wall shear stress (WSS) and strain in osteoblasts, is quantified at different locations within the scaffold for cells of different attachment morphologies (attached, bridged). The results show that 75.4 % of scaffold surface has a WSS of 0.1-10 mPa, which indicates the likelihood of bone cell differentiation at these locations. For attached and bridged osteoblasts, the maximum strains are 397 and 177,200 με, respectively. Additionally, the results from mechanical compression show that attached cells are more stimulated (maximum strain = 22,600 με) than bridged cells (maximum strain = 10.000 με)Such information is important for understanding the biological response of osteoblasts under in vitro stimulation. Finally, a combination of perfusion and compression of a tissue engineering scaffold is suggested for osteogenic differentiation.
Collapse
|
42
|
In vivo construction of tissue-engineered cartilage using adipose-derived stem cells and bioreactor technology. Cell Tissue Bank 2014; 16:123-33. [DOI: 10.1007/s10561-014-9448-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 04/10/2014] [Indexed: 01/22/2023]
|
43
|
Barberini DJ, Freitas NPP, Magnoni MS, Maia L, Listoni AJ, Heckler MC, Sudano MJ, Golim MA, da Cruz Landim-Alvarenga F, Amorim RM. Equine mesenchymal stem cells from bone marrow, adipose tissue and umbilical cord: immunophenotypic characterization and differentiation potential. Stem Cell Res Ther 2014; 5:25. [PMID: 24559797 PMCID: PMC4055040 DOI: 10.1186/scrt414] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 02/13/2014] [Indexed: 01/01/2023] Open
Abstract
Introduction Studies with mesenchymal stem cells (MSCs) are increasing due to their immunomodulatory, anti-inflammatory and tissue regenerative properties. However, there is still no agreement about the best source of equine MSCs for a bank for allogeneic therapy. The aim of this study was to evaluate the cell culture and immunophenotypic characteristics and differentiation potential of equine MSCs from bone marrow (BM-MSCs), adipose tissue (AT-MSCs) and umbilical cord (UC-MSCs) under identical in vitro conditions, to compare these sources for research or an allogeneic therapy cell bank. Methods The BM-MSCs, AT-MSCs and UC-MSCs were cultured and evaluated in vitro for their osteogenic, adipogenic and chondrogenic differentiation potential. Additionally, MSCs were assessed for CD105, CD44, CD34, CD90 and MHC-II markers by flow cytometry, and MHC-II was also assessed by immunocytochemistry. To interpret the flow cytometry results, statistical analysis was performed using ANOVA. Results The harvesting and culturing procedures of BM-MSCs, AT-MSCs and UC-MSCs were feasible, with an average cell growth until the third passage of 25 days for BM-MSCs, 15 days for AT-MSCs and 26 days for UC-MSCs. MSCs from all sources were able to differentiate into osteogenic (after 10 days for BM-MSCs and AT-MSCs and 15 days for UC-MSCs), adipogenic (after 8 days for BM-MSCs and AT-MSCs and 15 days for UC-MSCs) and chondrogenic (after 21 days for BM-MSCs, AT-MSCs and UC-MSCs) lineages. MSCs showed high expression of CD105, CD44 and CD90 and low or negative expression of CD34 and MHC-II. The MHC-II was not detected by immunocytochemistry techniques in any of the MSCs studied. Conclusions The BM, AT and UC are feasible sources for harvesting equine MSCs, and their immunophenotypic and multipotency characteristics attained minimal criteria for defining MSCs. Due to the low expression of MHC-II by MSCs, all of the sources could be used in clinical trials involving allogeneic therapy in horses. However, the BM-MSCs and AT-MSCs showed fastest ‘‘in vitro’’ differentiation and AT-MSCs showed highest cell growth until third passage. These findings suggest that BM and AT may be preferable for cell banking purposes.
Collapse
|
44
|
Zhang Q, Yin L, Li B, Meng R, Dao R, Hu S, Qiu X. Pulmonary adenocarcinoma with osseous metaplasia: a rare occurrence possibly associated with early stage? Onco Targets Ther 2013; 6:1631-4. [PMID: 24250229 PMCID: PMC3829678 DOI: 10.2147/ott.s48195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Adenocarcinoma is the most common type of malignant pulmonary tumor, but osseous metaplasia of this tumor is extremely rare. To date, only 21 cases have been reported in the literature worldwide. Here, we report a case of primary pulmonary adenocarcinoma with benign osseous stromal metaplasia in a 60-year-old woman and discuss the pathogenesis of intratumoral ossification and review the relevant literature. We found that pulmonary adenocarcinoma with osseous metaplasia may be more likely to occur in early tumor stages.
Collapse
Affiliation(s)
- Qingfu Zhang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|