1
|
Li L, Liu X, Patel M, Zhang L. Effect of hand-wrist exercises on distal radius fracture healing based on markerless motion capture system. J Biomech 2025; 179:112458. [PMID: 39662262 DOI: 10.1016/j.jbiomech.2024.112458] [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: 03/10/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
With the internal volar locking plate (VLP) technique emerging as a preferred surgical approach, early post-surgery therapeutic exercises have shown promise in promoting wrist functionality after distal radial fractures (DRFs). The biomechanical microenvironment, particularly the role of biomechanical stimuli, plays a crucial role in guiding stem tissue formation at the fracture site. However, much less is known about how various hand exercises interact with the microenvironment and influence fracture healing outcomes. This study employed the Leap Motion Controller for markerless hand motion capture and utilised an enhanced OpenSim hand model to simulate these motions. An advanced DRF healing model, integrating angiogenesis and the mechano-regulated maturation of callus tissue, was applied to simulate the MSCs differentiation and predict the healing outcomes. The effects of various rehabilitation exercises on DRFs' healing outcomes were systematically analysed. The results showed rehabilitation exercises, such as wrist extension/flexion and ulnar deviation, generally had a higher contact force on the distal radius compared with the slack state. Also, the relationship between contact force and muscle activations was not always linear, reflecting the intricate dynamics of the kinematic system. Exercise could induce changes in the bony bridge and cartilage formation, while angiogenesis remained unaffected. In the initial weeks, gripping exercises proved most beneficial, but as time progressed, extension and flexion exercises became more advantageous. The study highlights the importance of tailoring rehabilitation exercises to the dynamic healing process of DRFs. As the healing trajectory progresses, the therapeutic efficacy of specific exercises evolves, necessitating adaptive and patient-specific rehabilitation programs.
Collapse
Affiliation(s)
- Lunjian Li
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Xuanchi Liu
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia.
| | - Minoo Patel
- Centre for Limb Lengthening & Reconstruction, Epworth Hospital Richmond, Richmond, Victoria, Australia
| | - Lihai Zhang
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
2
|
Mozaffari N, Mohammadi R, Delirezh N, Hobbenaghi R, Mohammadi V. Effect of macrophages combined with supernatant of mesenchymal stem cell culture and macrophage culture on wound healing in rats. Tissue Cell 2024; 90:102474. [PMID: 39079451 DOI: 10.1016/j.tice.2024.102474] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 07/11/2024] [Indexed: 09/03/2024]
Abstract
Wound healing is an orderly sequence of events restoring the integrity of the damaged tissue. It consists of inflammatory, proliferation, and remodeling phases. The objective of the current study was to investigate the effect of local transplantation of cultured macrophage loaded with mesenchymal stem cell/macrophage culture supernatants on wound healing. Sixty-four healthy adult male Wistar rats were randomized into 4 groups of sixteen animals each: 1) SHAM group. 2) MAC-MSC/SN group: One-milliliter application of a mixture comprising mesenchymal stem cell and macrophage culture supernatants in a 1:1 ratio was administered locally to the wound bed. 3) MAC group: Local transplantation of macrophage cells cultured in the wound bed. 4) MAC + MAC-MSC/SN group: Local transplantation of cultured macrophage in combination with mesenchymal stem cell/ macrophage culture supernatants in the wound bed. An incisional wound model was used for biomechanical studies, while an excisional wound model was used for biochemical, histopathological, and planimetric assessments. The wound area was significantly reduced in the MAC + MAC-MSC/SN group compared to other groups (P < 0.05). Biomechanical measurements from the MAC + MAC-MSC/SN group were significantly higher compared to other experimental groups (P < 0.05). Biochemical and quantitative histopathological analyses revealed a significant difference between MAC + MAC-MSC/SN and other groups (P < 0.05). MAC + MAC-MSC/SN showed the potential to improve wound healing significantly. This appears to work by angiogenesis stimulation, fibroblast proliferation, inflammation reduction, and granulation tissue formation during the initial stages of the healing process. This accelerated healing leads to earlier wound area reduction and enhanced tensile strength of the damaged area due to the reorganization of granulation tissue and collagen fibers.
Collapse
Affiliation(s)
- Nima Mozaffari
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rahim Mohammadi
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Nowruz Delirezh
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rahim Hobbenaghi
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Vahid Mohammadi
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
3
|
Rota Graziosi E, François S, Nasser F, Gauthier M, Oger M, Favier AL, Drouet M, Jullien N, Riccobono D. Comparison of Three Antagonists of Hedgehog Pathway to Promote Skeletal Muscle Regeneration after High Dose Irradiation. Radiat Res 2024; 201:429-439. [PMID: 38253061 DOI: 10.1667/rade-23-00140.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/24/2024]
Abstract
The current geopolitical context has brought the radiological nuclear risk to the forefront of concerns. High-dose localized radiation exposure leads to the development of a musculocutaneous radiation syndrome affecting the skin and subcutaneous muscles. Despite the implementation of a gold standard treatment based on an invasive surgical procedure coupled with autologous cell therapy, a muscular defect frequently persists. Targeting the modulation of the Hedgehog (Hh) signaling pathway appears to be a promising therapeutic approach. Activation of this pathway enhances cell survival and promotes proliferation after irradiation, while inhibition by Cyclopamine facilitates differentiation. In this study, we compared the effects of three antagonists of Hh, Cyclopamine (CA), Vismodegib (VDG) and Sonidegib (SDG) on differentiation. A stable cell line of murine myoblasts, C2C12, was exposed to X-ray radiation (5 Gy) and treated with CA, VDG or SDG. Analysis of proliferation, survival (apoptosis), morphology, myogenesis genes expression and proteins production were performed. According to the results, VDG does not have a significant impact on C2C12 cells. SDG increases the expression/production of differentiation markers to a similar extent as CA, while morphologically, SDG proves to be more effective than CA. To conclude, SDG can be used in the same way as CA but already has a marketing authorization with an indication against basal cell cancers, facilitating their use in vivo. This proof of concept demonstrates that SDG represents a promising alternative to CA to promotes differentiation of murine myoblasts. Future studies on isolated and cultured satellite cells and in vivo will test this proof of concept.
Collapse
Affiliation(s)
- Emmanuelle Rota Graziosi
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Sabine François
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
| | - Farah Nasser
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Michel Gauthier
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Myriam Oger
- IRBA, French Armed Forces Biomedical Research Institute, Imagery Unit, Department of Platforms and Technology Research, Brétigny-sur-Orge, France
| | - Anne-Laure Favier
- IRBA, French Armed Forces Biomedical Research Institute, Imagery Unit, Department of Platforms and Technology Research, Brétigny-sur-Orge, France
| | - Michel Drouet
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
- IRBA, French Armed Forces Biomedical Research Institute, Radiations Bioeffects Department, Brétigny-sur-Orge, France
| | - Nicolas Jullien
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Diane Riccobono
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
- IRBA, French Armed Forces Biomedical Research Institute, Radiations Bioeffects Department, Brétigny-sur-Orge, France
| |
Collapse
|
4
|
Mattavelli D, Verzeletti V, Deganello A, Fiorentino A, Gualtieri T, Ferrari M, Taboni S, Anfuso W, Ravanelli M, Rampinelli V, Grammatica A, Buffoli B, Maroldi R, Elisabetta C, Rezzani R, Nicolai P, Piazza C. Computer-aided designed 3D-printed polymeric scaffolds for personalized reconstruction of maxillary and mandibular defects: a proof-of-concept study. Eur Arch Otorhinolaryngol 2024; 281:1493-1503. [PMID: 38170208 PMCID: PMC10857968 DOI: 10.1007/s00405-023-08392-0] [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: 11/01/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE To investigate the potential reconstruction of complex maxillofacial defects using computer-aided design 3D-printed polymeric scaffolds by defining the production process, simulating the surgical procedure, and explore the feasibility and reproducibility of the whole algorithm. METHODS This a preclinical study to investigate feasibility, reproducibility and efficacy of the reconstruction algorithm proposed. It encompassed 3 phases: (1) scaffold production (CAD and 3D-printing in polylactic acid); (2) surgical simulation on cadaver heads (navigation-guided osteotomies and scaffold fixation); (3) assessment of reconstruction (bone and occlusal morphological conformance, symmetry, and mechanical stress tests). RESULTS Six cadaver heads were dissected. Six types of defects (3 mandibular and 3 maxillary) with different degree of complexity were tested. In all case the reconstruction algorithm could be successfully completed. Bone morphological conformance was optimal while the occlusal one was slightly higher. Mechanical stress tests were good (mean value, 318.6 and 286.4 N for maxillary and mandibular defects, respectively). CONCLUSIONS Our reconstructive algorithm was feasible and reproducible in a preclinical setting. Functional and aesthetic outcomes were satisfactory independently of the complexity of the defect.
Collapse
Affiliation(s)
- Davide Mattavelli
- Unit of Otorhinolaryngology-Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy.
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, School of Medicine, Brescia, Italy.
| | - Vincenzo Verzeletti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, School of Medicine, Brescia, Italy
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua-Azienda Ospedale Università di Padova, Padua, Italy
| | - Alberto Deganello
- Otolaryngology Head and Neck Surgery Department of IRCCS, National Cancer Institute (INT), Milan, Italy
| | - Antonio Fiorentino
- Department of Mechanical and Industrial Engineering, University of Brescia, Brescia, Italy
| | - Tommaso Gualtieri
- Department of Otorhinolaryngology, Head and Neck Surgery, "Nuovo Santo Stefano" Civil Hospital, Prato, Italy
| | - Marco Ferrari
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua-Azienda Ospedale Università di Padova, Padua, Italy
- Guided Therapeutics (GTx) Program International Scholarship, University Health Network (UHN), Toronto, ON, Canada
| | - Stefano Taboni
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua-Azienda Ospedale Università di Padova, Padua, Italy
- Artificial Intelligence in Medicine and Innovation in Clinical Research and Methodology (PhD Program), Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - William Anfuso
- Otolaryngology Head and Neck Surgery Department of IRCCS, National Cancer Institute (INT), Milan, Italy
| | - Marco Ravanelli
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, School of Medicine, Brescia, Italy
- Unit of Radiology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Vittorio Rampinelli
- Unit of Otorhinolaryngology-Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alberto Grammatica
- Unit of Otorhinolaryngology-Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Barbara Buffoli
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, School of Medicine, Brescia, Italy
| | - Roberto Maroldi
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, School of Medicine, Brescia, Italy
- Unit of Radiology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Ceretti Elisabetta
- Department of Mechanical and Industrial Engineering, University of Brescia, Brescia, Italy
| | - Rita Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, School of Medicine, Brescia, Italy
| | - Piero Nicolai
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padua-Azienda Ospedale Università di Padova, Padua, Italy
| | - Cesare Piazza
- Unit of Otorhinolaryngology-Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, School of Medicine, Brescia, Italy
| |
Collapse
|
5
|
Ozhava D, Bektas C, Lee K, Jackson A, Mao Y. Human Mesenchymal Stem Cells on Size-Sorted Gelatin Hydrogel Microparticles Show Enhanced In Vitro Wound Healing Activities. Gels 2024; 10:97. [PMID: 38391427 PMCID: PMC10887759 DOI: 10.3390/gels10020097] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
The demand for innovative therapeutic interventions to expedite wound healing, particularly in vulnerable populations such as aging and diabetic patients, has prompted the exploration of novel strategies. Mesenchymal stem cell (MSC)-based therapy emerges as a promising avenue for treating acute and chronic wounds. However, its clinical application faces persistent challenges, notably the low survivability and limited retention time of engraftment in wound environments. Addressing this, a strategy to sustain the viability and functionality of human MSCs (hMSCs) in a graft-able format has been identified as crucial for advanced wound care. Hydrogel microparticles (HMPs) emerge as promising entities in the field of wound healing, showcasing versatile capabilities in delivering both cells and bioactive molecules/drugs. In this study, gelatin HMPs (GelMPs) were synthesized via an optimized mild processing method. GelMPs with distinct diameter sizes were sorted and characterized. The growth of hMSCs on GelMPs with various sizes was evaluated. The release of wound healing promoting factors from hMSCs cultured on different GelMPs were assessed using scratch wound assays and gene expression analysis. GelMPs with a size smaller than 100 microns supported better cell growth and cell migration compared to larger sizes (100 microns or 200 microns). While encapsulation of hMSCs in hydrogels has been a common route for delivering viable hMSCs, we hypothesized that hMSCs cultured on GelMPs are more robust than those encapsulated in hydrogels. To test this hypothesis, hMSCs were cultured on GelMPs or in the cross-linked methacrylated gelatin hydrogel (GelMA). Comparative analysis of growth and wound healing effects revealed that hMSCs cultured on GelMPs exhibited higher viability and released more wound healing activities in vitro. This observation highlights the potential of GelMPs, especially those with a size smaller than 100 microns, as a promising carrier for delivering hMSCs in wound healing applications, providing valuable insights for the optimization of advanced therapeutic strategies.
Collapse
Affiliation(s)
- Derya Ozhava
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
- Department of Chemistry and Chemical Processing Technologies, Cumra Vocational School, Selcuk University, 42130 Konya, Turkey
| | - Cemile Bektas
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Kathleen Lee
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Anisha Jackson
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
6
|
Lee G, Han SB, Kim SH, Jeong S, Kim DH. Stretching of porous poly (l-lactide-co-ε-caprolactone) membranes regulates the differentiation of mesenchymal stem cells. Front Cell Dev Biol 2024; 12:1303688. [PMID: 38333594 PMCID: PMC10850303 DOI: 10.3389/fcell.2024.1303688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Background: Among a variety of biomaterials supporting cell growth for therapeutic applications, poly (l-lactide-co-ε-caprolactone) (PLCL) has been considered as one of the most attractive scaffolds for tissue engineering owing to its superior mechanical strength, biocompatibility, and processibility. Although extensive studies have been conducted on the relationship between the microstructure of polymeric materials and their mechanical properties, the use of the fine-tuned morphology and mechanical strength of PLCL membranes in stem cell differentiation has not yet been studied. Methods: PLCL membranes were crystallized in a combination of diverse solvent-nonsolvent mixtures, including methanol (MeOH), isopropanol (IPA), chloroform (CF), and distilled water (DW), with different solvent polarities. A PLCL membrane with high mechanical strength induced by limited pore formation was placed in a custom bioreactor mimicking the reproducible physiological microenvironment of the vascular system to promote the differentiation of mesenchymal stem cells (MSCs) into smooth muscle cells (SMCs). Results: We developed a simple, cost-effective method for fabricating porosity-controlled PLCL membranes based on the crystallization of copolymer chains in a combination of solvents and non-solvents. We confirmed that an increase in the ratio of the non-solvent increased the chain aggregation of PLCL by slow evaporation, leading to improved mechanical properties of the PLCL membrane. Furthermore, we demonstrated that the cyclic stretching of PLCL membranes induced MSC differentiation into SMCs within 10 days of culture. Conclusion: The combination of solvent and non-solvent casting for PLCL solidification can be used to fabricate mechanically durable polymer membranes for use as mechanosensitive scaffolds for stem cell differentiation.
Collapse
Affiliation(s)
- Geonhui Lee
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sangmoo Jeong
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Alipanah-Moghadam R, Khodaei M, Aghamohammadi V, Malekzadeh V, Afrouz M, Nemati A, Zahedian H. Andrographolide induced heme oxygenase-1 expression in MSC-like cells isolated from rat bone marrow exposed to environmental stress. Biochem Biophys Res Commun 2023; 687:149212. [PMID: 37944470 DOI: 10.1016/j.bbrc.2023.149212] [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: 08/12/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Mesenchymal stem cells (MSC-like cells) are the most important stem cells that are used in transplantation clinically in various applications. The survival rate of MSC-like cells is strongly reduced due to adverse conditions in the microenvironment of transplantation, including environmental stress. Heme oxygenase-1 (HO-1) is a member of the heat shock protein, as well as a stress-induced enzyme, present throughout the body. The present study was conducted to investigate the effect of andrographolide, an active derivative from andrographolide paniculate, on HO-1 expression in mesenchymal stem cells derived from rat bone marrow. MATERIALS AND METHODS The rat bone marrow-derived mesenchymal stem cells (BMSC-like cells) were extracted and proliferated in several passages. The identity of MSC-like cells was confirmed by morphological observations and differential tests. The flow cytometry method was used to verify the MSC-specific markers. Isolated MSC-like cells were treated with different concentrations of andrographolide and then exposed to environmental stress. Cell viability was assessed using the MTT colorimetric assay. A real-time PCR technique was employed to evaluate the expression level of HO-1 in the treated MSC-like cells. RESULTS Isolated MSC-like cells demonstrated fibroblast-like morphology. These cells in different culture mediums differentiated into osteocytes and adipocytes and were identified using alizarin red and oil red staining, respectively. As well, MSC-like cells were verified by the detection of CD105 surface antigen and the absence of CD14 and CD45 antigens. The results of the MTT assay showed that the pre-treatment of MSC-like cells with andrographolide concentration independently increased the viability and resistance of these cells to environmental stress caused by hydrogen peroxide and serum deprivation (SD). Real-time PCR findings indicated a significant increase in HO-1 gene expression in the andrographolide-receiving groups (p < 0.01). CONCLUSION Our results suggest that andrographolide creates a promising strategy for enhancing the quality of cell therapy by increasing the resistance of MSC-like cells to environmental stress and inducing the expression of HO-1.
Collapse
Affiliation(s)
- Reza Alipanah-Moghadam
- Department of Clinical Biochemistry, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Khodaei
- Department of Clinical Biochemistry, Ardabil University of Medical Sciences, Ardabil, Iran.
| | | | - Vadoud Malekzadeh
- Department of Anatomical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mehdi Afrouz
- Department of Plant Production and Genetics, University of Mohaghegh Ardabili, Iran.
| | - Ali Nemati
- Department of Clinical Biochemistry, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hoda Zahedian
- Department of Deutsch-Sprachen, Volkshochschule, Gütersloh, Germany
| |
Collapse
|
8
|
Hosseinzadeh M, Kamali A, Baghaban Eslaminejad M, Hosseini S. Higher ratios of chondrocyte to mesenchymal stem cells elevate the therapeutic effects of extracellular vesicles harvested from chondrocyte/mesenchymal stem cell co-culture on osteoarthritis in a rat model. Cell Tissue Res 2023; 394:145-162. [PMID: 37526734 DOI: 10.1007/s00441-023-03819-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023]
Abstract
Extracellular vesicles (EVs) may have a key therapeutic role and offer an innovative treatment for osteoarthritis (OA). Studies have shown that ratio of MSC/chondrocyte could affect their therapeutic outcomes. Here, we investigate the chondrogenic potential and therapeutic effect of EVs derived from MSCs and chondrocytes in the naïve, chondrogenically primed, and co-culture states to treat OA. EVs are isolated from naïve MSCs (M-EV), chondrogenically primed MSCs (cpM-EV), chondrocytes (C-EV), and co-cultures of chondrocytes plus MSCs at ratios of 1:1 (C/M-EV), 2:1 (2C/M-EV), and 4:1 (4C/M-EV). We characterized the isolated EVs in terms of surface markers, morphology, size, and zeta potential, and evaluated their chondrogenic potential in vitro by qRT-PCR and histological analyses. Next, these EVs were intra-articularly injected into osteoarthritic cartilage of a rat model and assessed by radiography, gait parameters, and histological and immunohistochemical analyses. EVs obtained from chondrocytes co-cultured with MSCs resulted in improved matrix production and functional differentiation. Our research showed that close proximity between the two cell types was essential for this response, and improved chondrogenesis and matrix formation were the outcomes of this interaction in vitro. Furthermore, in the in vivo rat OA model induced by a monoiodoacetate (MIA), we observed recovery from OA by increasing ratio of the C/M-derived EV group compared to the other groups. Our findings show that the increasing chondrocyte ratio to MSC leads to high chondrogenic induction and the therapeutic effect of harvested EVs for cartilage repair.
Collapse
Affiliation(s)
- Maryam Hosseinzadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Kamali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Li L, Liu X, Patel M, Zhang L. Depth camera-based model for studying the effects of muscle loading on distal radius fracture healing. Comput Biol Med 2023; 164:107292. [PMID: 37544250 DOI: 10.1016/j.compbiomed.2023.107292] [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: 04/18/2023] [Revised: 06/24/2023] [Accepted: 07/28/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Distal radius fractures (DRFs) treated with volar locking plates (VLPs) allows early rehabilitation exercises favourable to fracture recovery. However, the role of rehabilitation exercises induced muscle forces on the biomechanical microenvironment at the fracture site remains to be fully explored. The purpose of this study is to investigate the effects of muscle forces on DRF healing by developing a depth camera-based fracture healing model. METHOD First, the rehabilitation-related hand motions were captured by a depth camera system. A macro-musculoskeletal model is then developed to analyse the data captured by the system for estimating hand muscle and joint reaction forces which are used as inputs for our previously developed DRF model to predict the tissue differentiation patterns at the fracture site. Finally, the effect of different wrist motions (e.g., from 60° of extension to 60° of flexion) on the DRF healing outcomes will be studied. RESULTS Muscle and joint reaction forces in hands which are highly dependent on hand motions could significantly affect DRF healing through imposed compressive and bending forces at the fracture site. There is an optimal range of wrist motion (i.e., between 40° of extension and 40° of flexion) which could promote mechanical stimuli governed healing while mitigating the risk of bony non-union due to excessive movement at the fracture site. CONCLUSION The developed depth camera-based fracture healing model can accurately predict the influence of muscle loading induced by rehabilitation exercises in distal radius fracture healing outcomes. The outcomes from this study could potentially assist osteopathic surgeons in designing effective post-operative rehabilitation strategies for DRF patients.
Collapse
Affiliation(s)
- Lunjian Li
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Xuanchi Liu
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia.
| | - Minoo Patel
- Centre for Limb Lengthening & Reconstruction, Epworth Hospital Richmond, Richmond, Victoria, Australia
| | - Lihai Zhang
- Department of Infrastructure Engineering, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Turano E, Scambi I, Virla F, Bonetti B, Mariotti R. Extracellular Vesicles from Mesenchymal Stem Cells: Towards Novel Therapeutic Strategies for Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24032917. [PMID: 36769247 PMCID: PMC9917806 DOI: 10.3390/ijms24032917] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Neurodegenerative diseases are fatal disorders of the central nervous system (CNS) which currently lack effective treatments. The application of mesenchymal stem cells (MSCs) represents a new promising approach for treating these incurable disorders. Growing evidence suggest that the therapeutic effects of MSCs are due to the secretion of neurotrophic molecules through extracellular vesicles. The extracellular vesicles produced by MSCs (MSC-EVs) have valuable innate properties deriving from parental cells and could be exploited as cell-free treatments for many neurological diseases. In particular, thanks to their small size, they are able to overcome biological barriers and reach lesion sites inside the CNS. They have a considerable pharmacokinetic and safety profile, avoiding the critical issues related to the fate of cells following transplantation. This review discusses the therapeutic potential of MSC-EVs in the treatment of neurodegenerative diseases, focusing on the strategies to further enhance their beneficial effects such as tracking methods, bioengineering applications, with particular attention to intranasal delivery as a feasible strategy to deliver MSC-EVs directly to the CNS in an effective and minimally invasive way. Current progresses and limiting issues to the extent of the use of MSC-EVs treatment for human neurodegenerative diseases will be also revised.
Collapse
Affiliation(s)
- Ermanna Turano
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Ilaria Scambi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Federica Virla
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Bruno Bonetti
- Neurology Unit, Azienda Ospedaliera Universitaria Integrata Verona, 37124 Verona, Italy
| | - Raffaella Mariotti
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
- Correspondence: ; Tel.: +39-045-802-7164
| |
Collapse
|
11
|
Lv M, Xu Q, He F, Guo J, Zheng Z, Xie J, Wang W. Transosseous-Equivalent/Suture Bridge Technique in Combination With Platelet-Rich Plasma Application Yield Optimal Clinical Outcomes in Arthroscopic Rotator Cuff Repair: A Bayesian Network Analysis of Randomized Controlled Trials. Arthroscopy 2023; 39:425-437.e1. [PMID: 36343767 DOI: 10.1016/j.arthro.2022.10.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE To assess the clinical evidence defining the optimal combination of arthroscopic suture technique and platelet-rich products (PRP), and application for arthroscopic rotator cuff repair (ARCR). METHODS All level of evidence (LOE) I randomized controlled trials (RCT) focusing on arthroscopic suture technique and/or PRP application in ARCR were included. The exclusion criteria were LOE II or worse, studies with other interventions, studies reported none of the predetermined clinical outcomes; studies unable to extract any precise data; studies from the same patient group of included studies. A pair-wise meta-analysis and Bayesian network analysis were performed on each comparison. The intervention options were ranked by Bayesian network analysis. RESULTS 27 studies comprising 1,947 individuals met the inclusion criteria. The application of transosseous equivalent/suture bridge repair (SB) with PRP (SB+PRP) significantly reduced retear rate (risk ratio [RR], 0.29; 95% confidence interval [CI], [0.15, 0.55].) and increased Constant-Murley score (mean difference, 1.90; 95% CI, [0.14, 3.74]), compared to SB repair. Single-row repair (SR) with PRP usage (SR+PRP) significantly reduced retear rate (RR, 0.27; 95% CI, [0.12, 0.55]) and pain visual analog scale (VAS) (mean difference: -0.84; 95% CI [-1.39, -0.46].), increased University of California, Los Angeles (UCLA) shoulder score (mean difference: 1.48; 95% CI [0.50, 2.58]) and Constant-Murley score (mean difference: 4.53; 95% CI [2.65, 6.38]), compared to SR repair. The ranking for outcomes demonstrated SB+PRP resulted in the best retear rate, UCLA shoulder score, with the second-best postoperative pain, Constant-Murley score, while SR+PRP resulted in the best postoperative pain, Constant-Murley score, with the second-best retear rate and UCLA score. CONCLUSION Arthroscopic rotator cuff repair utilizing SB+PRP yields optimal retear rate and UCLA shoulder score, with the second-best postoperative pain and Constant-Murley shoulder outcome score, while SR+PRP yields the best in these two parameters. LEVEL OF EVIDENCE Level I, Bayesian network analysis of level I RCT.
Collapse
Affiliation(s)
- Minchao Lv
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China
| | - Qingxin Xu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feixiong He
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China
| | - Jinku Guo
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China
| | - Zhenxin Zheng
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China
| | - Jun Xie
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China
| | - Wei Wang
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, China.
| |
Collapse
|
12
|
Hou W, Duan L, Huang C, Li X, Xu X, Qin P, Hong N, Wang D, Jin W. Cross-Tissue Characterization of Heterogeneities of Mesenchymal Stem Cells and Their Differentiation Potentials. Front Cell Dev Biol 2021; 9:781021. [PMID: 34977025 PMCID: PMC8719164 DOI: 10.3389/fcell.2021.781021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are promising cell sources for regenerative medicine and the treatment of autoimmune disorders. Comparing MSCs from different tissues at the single-cell level is fundamental for optimizing clinical applications. Here we analyzed single-cell RNA-seq data of MSCs from four tissues, namely umbilical cord, bone marrow, synovial tissue, and adipose tissue. We identified three major cell subpopulations, namely osteo-MSCs, chondro-MSCs, and adipo/myo-MSCs, across all MSC samples. MSCs from the umbilical cord exhibited the highest immunosuppression, potentially indicating it is the best immune modulator for autoimmune diseases. MSC subpopulations, with different subtypes and tissue sources, showed pronounced differences in differentiation potentials. After we compared the cell subpopulations and cell status pre-and-post chondrogenesis induction, osteogenesis induction, and adipogenesis induction, respectively, we found MSC subpopulations expanded and differentiated when their subtypes consist with induction directions, while the other subpopulations shrank. We identified the genes and transcription factors underlying each induction at the single-cell level and subpopulation level, providing better targets for improving induction efficiency.
Collapse
Affiliation(s)
- Wenhong Hou
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Li Duan
- Department of Orthopedics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Shenzhen Institute of Geriatircs, Shenzhen, China
| | - Changyuan Huang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xingfu Li
- Department of Orthopedics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xiao Xu
- Department of Orthopedics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Pengfei Qin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Ni Hong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Daping Wang
- Department of Orthopedics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- *Correspondence: Daping Wang, ; Wenfei Jin,
| | - Wenfei Jin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Daping Wang, ; Wenfei Jin,
| |
Collapse
|
13
|
Hidalgo‐Garcia L, Molina‐Tijeras JA, Huertas‐Peña F, Ruiz‐Malagón AJ, Diez‐Echave P, Vezza T, Rodríguez‐Sojo MJ, Morón R, Becerra‐Massare P, Rodríguez‐Nogales A, Gálvez J, Rodríguez‐Cabezas ME, Anderson P. Intestinal mesenchymal cells regulate immune responses and promote epithelial regeneration in vitro and in dextran sulfate sodium-induced experimental colitis in mice. Acta Physiol (Oxf) 2021; 233:e13699. [PMID: 34089568 DOI: 10.1111/apha.13699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022]
Abstract
AIM Disruption of the intestinal mucosal tolerance, that is, the immunological unresponsiveness to innocuous food antigens and the commensal microbiota, in the colon is associated with several chronic diseases including inflammatory bowel disease (IBD). Understanding the mechanisms responsible for intestinal mucosal tolerance has potential translational value for its therapy and management. Human intestinal mesenchymal cells (iMCs) play important roles in colonic mucosal tolerance, but further studies on their tissue regenerative and immunomodulatory capacities are necessary in order to fully understand their function in health and disease. METHODS In this study, we have isolated and analysed the capacity of human iMCs to promote wound healing and modulate immune responses in vitro and in vivo, using the dextran sulfate sodium (DSS)-induced colitis model. RESULTS Cultured iMCs were CD45- CD73+ CD90+ CD105+ and accelerated the wound closure in a normal colon mucosa (NCM) 356 human epithelial cell wound healing assay. Furthermore, iMCs blocked the LPS-mediated induction of TNF-α in THP-1 macrophages and inhibited the proliferation of peripheral blood mononuclear cells, partly through the induction of indoleamine-2,3-dioxygenase. In DSS colitic mice, iMCs administration reduced the disease activity index and ameliorated intestinal tissue damage and permeability. Furthermore, iMCs reduced intestinal inflammation, evidenced by a decreased mRNA expression of pro-inflammatory cytokines, reduced IL-1β secretion by intestinal explants and inhibited colonic iNOS protein expression. CONCLUSIONS Our data show that human iMCs isolated from the noninflamed intestine possess tissue-regenerative and immunomodulatory capacities that could potentially be harnessed/restored in order to reduce IBD severity.
Collapse
Affiliation(s)
- Laura Hidalgo‐Garcia
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - José Alberto Molina‐Tijeras
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Francisco Huertas‐Peña
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio de Cirugía Hospital Universitario Virgen de las Nieves Granada Spain
| | - Antonio Jesús Ruiz‐Malagón
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Patricia Diez‐Echave
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Teresa Vezza
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - María Jesús Rodríguez‐Sojo
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Rocío Morón
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio Farmacia Hospitalaria Hospital Universitario Clínico San Cecilio Granada Spain
| | | | - Alba Rodríguez‐Nogales
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio de Digestivo Hospital Universitario Virgen de las Nieves Granada Spain
| | - Julio Gálvez
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBER‐EHD) University of Granada Granada Spain
| | - María Elena Rodríguez‐Cabezas
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Per Anderson
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio de Análisis Clínicos e Inmunología Hospital Universitario Virgen de las Nieves Granada Spain
| |
Collapse
|
14
|
Pan Q, Li Y, Li Y, Wang H, Kong L, Yang Z, Zhang X, Bai S, Zong Z, Chen G, Lin S, Li G. Local administration of allogeneic or autologous bone marrow-derived mesenchymal stromal cells enhances bone formation similarly in distraction osteogenesis. Cytotherapy 2021; 23:590-598. [PMID: 33546925 DOI: 10.1016/j.jcyt.2020.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AIMS Distraction osteogenesis (DO) is a surgical technique to promote bone regeneration that requires a long time for bone healing. Bone marrow-derived mesenchymal stromal cells (MSCs) have been applied to accelerate bone formation in DO. Allogeneic MSCs are attractive, as they could be ready to use in clinics. Whether allogeneic MSCs would have an effect similar to autologous MSCs with regard to promoting bone formation in DO is still unknown. This study compares the effect of autologous MSCs versus allogeneic MSCs on bone formation in a rat DO model. METHODS Rat bone marrow-derived MSCs were isolated, characterized and expanded in vitro. Adult rats were subjected to right tibia transverse osteotomy. On the third day of distraction, each rat received one injection of phosphate-buffered saline (PBS), autologous MSCs or allogeneic MSCs at the distraction site. Tibiae were harvested after 28 days of consolidation for micro-computed tomography examination, mechanical test and histological analysis. RESULTS Results showed that treatment with both allogeneic and autologous MSCs promoted bone formation, with significantly higher bone mass, mechanical properties and mineral apposition rate as well as expression of angiogenic and bone formation markers at the regeneration sites compared with the PBS-treated group. No statistical difference in bone formation was found between the allogeneic and autologous MSC treatment groups. CONCLUSIONS This study indicates that allogeneic and autologous MSCs have a similar effect on promoting bone consolidation in DO. MSCs from an allogeneic source could be used off-the-shelf with DO to achieve early bone healing.
Collapse
Affiliation(s)
- Qi Pan
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Ye Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Yucong Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Lingchi Kong
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Zhengmeng Yang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Xiaoting Zhang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Shanshan Bai
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Zhixian Zong
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Guanghua Chen
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Sien Lin
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China; Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, California, USA.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China; Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China; Department of Orthopaedics and Traumatology, Affiliated Baoan Hospital of Shenzhen, Southern Medical University, People's Hospital of Baoan District, Shenzhen, China..
| |
Collapse
|
15
|
Liu B, Jeong HJ, Yeo JH, Oh JH. Efficacy of Intraoperative Platelet-Rich Plasma Augmentation and Postoperative Platelet-Rich Plasma Booster Injection for Rotator Cuff Healing: A Randomized Controlled Clinical Trial. Orthop J Sports Med 2021; 9:23259671211006100. [PMID: 34159208 PMCID: PMC8182201 DOI: 10.1177/23259671211006100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/05/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Platelet-rich plasma (PRP) has been applied as an adjuvant treatment for
arthroscopic rotator cuff repair (ARCR) to enhance rotator cuff healing.
However, it remains debatable whether PRP enhances tendon-to-bone
healing. Purpose: To assess the efficacy of intraoperative augmentation and postoperative
injection of PRP that was prepared using the double-spin method and calcium
activation without thrombin in patients with ARCR. Study Design: Randomized controlled trial; Level of evidence, 1; and cohort study; Level of
evidence, 3. Methods: A total of 58 patients underwent ARCR using intraoperative PRP augmentation.
Half of the patients were randomly assigned to receive an additional
ultrasound-guided PRP injection at the repair site at 2 weeks
postoperatively (PRP-booster group); the other half did not receive the
booster injection (PRP-only group). A control group that did not receive any
PRP treatment was retrospectively matched using propensity score matching.
Structural integrity was assessed using magnetic resonance imaging at 1 year
postoperatively, and healing rates were compared between patients with tear
sizes ≤2 cm versus >2 cm. Functional outcomes were assessed using the
visual analog scale (VAS) for pain; VAS for satisfaction; shoulder range of
motion; and Constant, American Shoulder and Elbow Surgeons, and Simple
Shoulder Test scores at minimum 2-year follow-up. Results: In patients with tears >2 cm, the rate of healing failure at 1-year
follow-up was significantly less in the overall PRP group than in the
control group (12.9% vs 35.7%, respectively; P = .040),
however, the PRP-booster group did not present a better healing rate than
did the PRP-only group. The overall PRP group had lower VAS for pain scores
compared with the control group (0.5 ± 1.1 vs 1.3 ± 1.8, respectively;
P = .016) and higher VAS for satisfaction scores (9.2 ±
1.2 vs 8.6 ± 1.7; P = .023) at the final follow-up, whereas
no statistical difference was found between the PRP-only and PRP-booster
groups in functional outcomes. Conclusion: Intraoperative PRP augmentation during ARCR demonstrated superior anatomic
healing results in patients with rotator cuff tears >2 cm as well as
reduced pain and increased subjective satisfaction. PRP booster injection
provided no additional benefit to tendon integrity or functional
recovery.
Collapse
Affiliation(s)
- Bei Liu
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Hyeon Jang Jeong
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Ji Hyun Yeo
- Department of Orthopaedic Surgery, Dongguk University College of Medicine, Dongguk University Ilsan Hospital, Goyangsi, Republic of Korea
| | - Joo Han Oh
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| |
Collapse
|
16
|
Alarcin E, Bal-Öztürk A, Avci H, Ghorbanpoor H, Dogan Guzel F, Akpek A, Yesiltas G, Canak-Ipek T, Avci-Adali M. Current Strategies for the Regeneration of Skeletal Muscle Tissue. Int J Mol Sci 2021; 22:5929. [PMID: 34072959 PMCID: PMC8198586 DOI: 10.3390/ijms22115929] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic injuries, tumor resections, and degenerative diseases can damage skeletal muscle and lead to functional impairment and severe disability. Skeletal muscle regeneration is a complex process that depends on various cell types, signaling molecules, architectural cues, and physicochemical properties to be successful. To promote muscle repair and regeneration, various strategies for skeletal muscle tissue engineering have been developed in the last decades. However, there is still a high demand for the development of new methods and materials that promote skeletal muscle repair and functional regeneration to bring approaches closer to therapies in the clinic that structurally and functionally repair muscle. The combination of stem cells, biomaterials, and biomolecules is used to induce skeletal muscle regeneration. In this review, we provide an overview of different cell types used to treat skeletal muscle injury, highlight current strategies in biomaterial-based approaches, the importance of topography for the successful creation of functional striated muscle fibers, and discuss novel methods for muscle regeneration and challenges for their future clinical implementation.
Collapse
Affiliation(s)
- Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34854 Istanbul, Turkey;
| | - Ayca Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, 34010 Istanbul, Turkey;
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Hüseyin Avci
- Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Cellular Therapy and Stem Cell Research Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Translational Medicine Research and Clinical Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Hamed Ghorbanpoor
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
- Department of Biomedical Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Fatma Dogan Guzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
| | - Ali Akpek
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Gözde Yesiltas
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Tuba Canak-Ipek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| |
Collapse
|
17
|
Nonsurgical Management of Ulnar Collateral Ligament Injuries. JOURNAL OF THE AMERICAN ACADEMY OF ORTHOPAEDIC SURGEONS GLOBAL RESEARCH AND REVIEWS 2021; 5:01979360-202104000-00003. [PMID: 33835949 DOI: 10.5435/jaaosglobal-d-20-00257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/30/2021] [Indexed: 12/14/2022]
Abstract
Ulnar collateral ligament (UCL) injuries are a common source of pain and disability in the overhead athlete and often result in notable loss of time from competition. Over the past 10 to 15 years, the prevalence of UCL injury and reconstruction has undergone a dramatic increase, making it imperative to determine which patients may benefit from a nonsurgical regimen. Nonsurgical treatment involves a multidisciplinary approach of rehabilitation with tailored physical therapy programs and, in certain cases, biologic adjuncts. Physical therapy protocols should focus on strengthening the periscapular muscles, rotator cuff, core musculature, and flexor pronator mass to help stabilize the injured elbow and prevent injury recurrence before the initiation of a progressive throwing program. The implementation of injury prevention programs has shifted the focus from just the elbow and have included the shoulder, legs, and core in an effort to help decrease the stress on the upper extremity. In addition, biologic therapies such as platelet-rich therapy are promising modalities to augment the conservative treatment of UCL injuries but remain under investigation. The purpose of this study is to review available strategies and outcomes for conservatively treating UCL injuries.
Collapse
|
18
|
Glaeser JD, Behrens P, Stefanovic T, Salehi K, Papalamprou A, Tawackoli W, Metzger MF, Eberlein S, Nelson T, Arabi Y, Kim K, Baloh RH, Ben-David S, Cohn-Schwartz D, Ryu R, Bae HW, Gazit Z, Sheyn D. Neural crest-derived mesenchymal progenitor cells enhance cranial allograft integration. Stem Cells Transl Med 2021; 10:797-809. [PMID: 33512772 PMCID: PMC8046069 DOI: 10.1002/sctm.20-0364] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/10/2020] [Accepted: 11/09/2020] [Indexed: 01/17/2023] Open
Abstract
Replacement of lost cranial bone (partly mesodermal and partly neural crest‐derived) is challenging and includes the use of nonviable allografts. To revitalize allografts, bone marrow‐derived mesenchymal stromal cells (mesoderm‐derived BM‐MSCs) have been used with limited success. We hypothesize that coating of allografts with induced neural crest cell‐mesenchymal progenitor cells (iNCC‐MPCs) improves implant‐to‐bone integration in mouse cranial defects. Human induced pluripotent stem cells were reprogramed from dermal fibroblasts, differentiated to iNCCs and then to iNCC‐MPCs. BM‐MSCs were used as reference. Cells were labeled with luciferase (Luc2) and characterized for MSC consensus markers expression, differentiation, and risk of cellular transformation. A calvarial defect was created in non‐obese diabetic/severe combined immunodeficiency (NOD/SCID) mice and allografts were implanted, with or without cell coating. Bioluminescence imaging (BLI), microcomputed tomography (μCT), histology, immunofluorescence, and biomechanical tests were performed. Characterization of iNCC‐MPC‐Luc2 vs BM‐MSC‐Luc2 showed no difference in MSC markers expression and differentiation in vitro. In vivo, BLI indicated survival of both cell types for at least 8 weeks. At week 8, μCT analysis showed enhanced structural parameters in the iNCC‐MPC‐Luc2 group and increased bone volume in the BM‐MSC‐Luc2 group compared to controls. Histology demonstrated improved integration of iNCC‐MPC‐Luc2 allografts compared to BM‐MSC‐Luc2 group and controls. Human osteocalcin and collagen type 1 were detected at the allograft‐host interphase in cell‐seeded groups. The iNCC‐MPC‐Luc2 group also demonstrated improved biomechanical properties compared to BM‐MSC‐Luc2 implants and cell‐free controls. Our results show an improved integration of iNCC‐MPC‐Luc2‐coated allografts compared to BM‐MSC‐Luc2 and controls, suggesting the use of iNCC‐MPCs as potential cell source for cranial bone repair.
Collapse
Affiliation(s)
- Juliane D Glaeser
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Phillip Behrens
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tina Stefanovic
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Khosrowdad Salehi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Angela Papalamprou
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Wafa Tawackoli
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Melodie F Metzger
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Orthopaedic Biomechanics Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Samuel Eberlein
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Trevor Nelson
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yasaman Arabi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kevin Kim
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Orthopaedic Biomechanics Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert H Baloh
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shiran Ben-David
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Doron Cohn-Schwartz
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Division of Internal Medicine, Rambam Health Care Campus, Haifa, Israel
| | - Robert Ryu
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hyun W Bae
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zulma Gazit
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
19
|
Deng Z, Jin J, Wang S, Qi F, Chen X, Liu C, Li Y, Ma Y, Lyu F, Zheng Q. Narrative review of the choices of stem cell sources and hydrogels for cartilage tissue engineering. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1598. [PMID: 33437797 PMCID: PMC7791208 DOI: 10.21037/atm-20-2342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Stem cell-based therapy is a promising treatment for cartilage defects due to the pluripotency, abundant sources and low immunogenicity of stem cells. Hydrogels are a promising class of biomaterials for cartilage engineering and are characterized by bioactivity, degradability and elasticity as well as provide water content and mechanical support. The combination of stem cells and hydrogels opens new possibilities for cartilage tissue engineering. However, the selection of suitable types of stem cells and hydrogels is difficult. Currently, various types of stem cells, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and peripheral blood mononuclear cells (PBMSCs), and various types of hydrogels, including natural polymers, chemically modified natural polymers and synthetic polymers, have been explored based on their potential for cartilage tissue engineering. These materials are used independently or in combination; however, there is no clear understanding of their merits and disadvantages with regard to their suitability for cartilage repair. In this article, we aim to review recent progress in the use of stem cell-hydrogel hybrid constructs for cartilage tissue engineering. We focus on the effects of stem cell types and hydrogel types on efficient chondrogenesis from cellular, preclinical and clinical perspectives. We compare and analyze the advantages and disadvantages of these cells and hydrogels with the hope of increasing discussion of their suitability for cartilage repair and present our perspective on their use for the improvement of physical and biological properties for cartilage tissue engineering.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiewen Jin
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fangjie Qi
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xuepan Chen
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chang Liu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fengjuan Lyu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,South China University of Technology-the University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
20
|
Yeung DA, Kelly NH. The Role of Collagen-Based Biomaterials in Chronic Wound Healing and Sports Medicine Applications. Bioengineering (Basel) 2021; 8:bioengineering8010008. [PMID: 33429996 PMCID: PMC7827215 DOI: 10.3390/bioengineering8010008] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/24/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Advancements in tissue engineering have taken aim at treating tissue types that have difficulty healing naturally. In order to achieve improved healing conditions, the balance of exogenous matrix, cells, and different factors must be carefully controlled. This review seeks to explore the aspects of tissue engineering in specific tissue types treated in sports medicine and advanced wound management from the perspective of the matrix component. While the predominant material to be discussed is collagen I, it would be remiss not to mention its relation to the other contributing factors to tissue engineered healing. The main categories of materials summarized here are (1) reconstituted collagen scaffolds, (2) decellularized matrix tissue, and (3) non-decellularized tissue. These three groups are ordered by their increase in additional components beyond simply collagen.
Collapse
|
21
|
BMSC-derived exosomes from congenital polydactyly tissue alleviate osteoarthritis by promoting chondrocyte proliferation. Cell Death Discov 2020; 6:142. [PMID: 33303743 PMCID: PMC7730395 DOI: 10.1038/s41420-020-00374-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
In the past decade, mesenchymal stem cells (MSCs) have been widely used for the treatment of osteoarthritis (OA), and exosomes may play a major role. Here, we acquired a special kind of MSCs from the bone marrow of surgically resected tissue from the hand of a patient with polydactyly. Experiments were focused on the role of polydactyly bone marrow-derived MSCs (pBMSCs) in osteoarthritis. The results showed that the pBMSCs had a greater ability than the BMSCs to differentiate into chondrocytes. Mechanistically, the expression of BMP4 was significantly higher in the pBMSCs than it was in the BMSCs. Furthermore, we showed that the migration and proliferation of chondrocytes were stimulated by exosomes secreted by pBMSC (pBMSC-EXOs). Notably, the downregulation of BMP4 in pBMSCs by siRNA inhibited both the chondrogenic differentiation potential of the MSCs and the function of the chondrocytes. In addition, the injection of pBMSC-EXOs and BMSC-EXOs attenuated OA in an OA mouse model, but the pBMSC-EXOs had a superior therapeutic effect compared with that of the BMSC-EXOs. Taken together, the data indicate that pBMSCs have greater ability to differentiate into chondrocytes and regulate chondrocyte formation through BMP4 signaling. Therefore, pBMSC-EXOs may represent a novel treatment for OA.
Collapse
|
22
|
Guo Q, Guo Q, Xiao Y, Li C, Huang Y, Luo X. Regulation of bone marrow mesenchymal stem cell fate by long non-coding RNA. Bone 2020; 141:115617. [PMID: 32853852 DOI: 10.1016/j.bone.2020.115617] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
Abstract
Bone mesenchymal stem cells (BMSCs) are progenitor cells isolated from bone marrow, which keep potential to differentiate into several kinds of cells including osteoblasts and adipocytes. A dynamic mutual regulation exists between osteogenesis and adipogenesis processes. Long non-coding RNA (lncRNA) performs diverse functions in biological activities including regulation of BMSCs commitment. Evidence has shown that lncRNA regulates key signaling pathways including TGFβ/BMP, Wnt and Notch pathways, and several transcription factors in BMSCs differention. Dysregulation of lncRNA in BMSCs leads to disruption of osteo-adipogenesis difffrentiation and results in impairment of bone homeostasis. In this review, we focus on the role of lncRNA in several critical signaling pathways that involved in regulation of osteo-adipogenesis of BMSC and prospects the potential clinical application of lncRNA.
Collapse
Affiliation(s)
- Qiaoyue Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan, PR China.
| |
Collapse
|
23
|
Cha H, Hong S, Park JH, Park HH. Stem Cell-Derived Exosomes and Nanovesicles: Promotion of Cell Proliferation, Migration, and Anti-Senescence for Treatment of Wound Damage and Skin Ageing. Pharmaceutics 2020; 12:E1135. [PMID: 33255430 PMCID: PMC7761250 DOI: 10.3390/pharmaceutics12121135] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs), such as exosomes, are nano-sized vesicles derived from endocytic membranes and contain biomolecules such as proteins, lipids, RNAs, and DNAs for the transfer of signals to recipient cells, playing significant roles in cell-to-cell communication. Discovery of exosomes has attracted attention for possible use as next generation therapies in clinical applications; however, several studies suggest that cells secrete exosomes that perform as mediators in the tumor niche and play several roles in tumorigenesis, angiogenesis, and metastasis. Recently, stem cell-derived exosomes have been suggested as a desirable source for regenerative medicine due to their roles in the promotion of angiogenesis via migratory and proliferative mechanisms. This review is aimed at demonstrating the present knowledge of stem cell-derived exosomes and cell-engineered nanovesicles (CNVs) as proliferative, migratory, and anti-senescent therapeutic biomaterial for use in tissue regeneration; wound healing and anti-ageing are explained. We conclude this review by discussing the future perspectives of stem cell-derived exosomes and CNVs as a platform in therapeutic strategies for treatment of wound damage and skin aging.
Collapse
Affiliation(s)
- Hyeonjin Cha
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
| | - Seyoung Hong
- Interdisciplinary Program in Biohealth-Machinery Convergence Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
| | - Ju Hyun Park
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
| | - Hee Ho Park
- Interdisciplinary Program in Biohealth-Machinery Convergence Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
- Department of Biotechnology and Bioengineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| |
Collapse
|
24
|
Radmanesh F, Mahmoudi M, Yazdanpanah E, Keyvani V, Kia N, Nikpoor AR, Zafari P, Esmaeili SA. The immunomodulatory effects of mesenchymal stromal cell-based therapy in human and animal models of systemic lupus erythematosus. IUBMB Life 2020; 72:2366-2381. [PMID: 33006813 DOI: 10.1002/iub.2387] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune inflammatory disease with no absolute cure. Although the exact etiopathogenesis of SLE is still enigmatic, it has been well demonstrated that a combination of genetic predisposition and environmental factors trigger a disturbance in immune responses and thereby participate in the development of this condition. Almost all available therapeutic strategies in SLE are primarily based on the administration of immunosuppressive drugs and are not curative. Mesenchymal stromal cells (MSCs) are a subset of non-hematopoietic adult stem cells that can be isolated from many adult tissues and are increasingly recognized as immune response modulating agents. MSC-mediated inhibition of immune responses is a complex mechanism that involves almost every aspect of the immune response. MSCs suppress the maturation of antigen-presenting cells (DC and MQ), proliferation of T cells (Th1, T17, and Th2), proliferation and immunoglobulin production of B cells, the cytotoxic activity of CTL and NK cells in addition to increasing regulatory cytokines (TGF-β and IL10), and decreasing inflammatory cytokines (IL17, INF-ϒ, TNF-α, and IL12) levels. MSCs have shown encouraging results in the treatment of several autoimmune diseases, in particular SLE. This report aims to review the beneficial and therapeutic properties of MSCs; it also focuses on the results of animal model studies, preclinical studies, and clinical trials of MSC therapy in SLE from the immunoregulatory aspect.
Collapse
Affiliation(s)
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahideh Keyvani
- Molecular Genetics, Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nadia Kia
- Skin Cancer Prevention Research Center, Torvergata University of Medical Sciences, Rome, Italy
| | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parisa Zafari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Science, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Science, Sari, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Yu F, Wu F, Li F, Liao X, Wang Y, Li X, Wang C, Shi Y, Ye L. Wnt7b-induced Sox11 functions enhance self-renewal and osteogenic commitment of bone marrow mesenchymal stem cells. Stem Cells 2020; 38:1020-1033. [PMID: 32346881 DOI: 10.1002/stem.3192] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/05/2023]
Abstract
As a profoundly anabolic regulator of bone, Wnt7b is well acknowledged to enhance osteoblast activities. Here, we report that bone marrow mesenchymal stem cells (BMSCs) are another important population responding to Wnt7b. In this study, we systematically investigated the in vivo role of Wnt7b in BMSCs using transgenic mice, high-throughput RNA-seq, immunohistochemistry, RT-qPCR, and in situ hybridization. These methods led us to uncover that Sox11 is induced via Wnt7b in BMSCs. Colony formation assay, flow cytometry, EdU incorporation labeling, RT-qPCR, and Western blot were conducted to detect the self-renewal capacity of BMSCs. Alkaline phosphatase staining, alizarin red staining, and ex vivo BMSCs transplantation were utilized to detect the osteogenic ability of BMSCs. ChIP-qPCR, shRNAs, and immunofluorescence staining were utilized to investigate the underlying mechanisms. Consequently, bone-derived Wnt7b was found to decrease in osteoporosis and elevate in bone fracture healing. During bone fracture healing, Wnt7b was particularly expressed in the mesenchymal cells residing within healing frontiers. RNA-seq data of Wnt7b-overexpressed bones uncovered the significant upregulation of Sox11. Histological results further unveiled that Sox11 is specifically increased in BMSCs. Wnt7b-induced Sox11 was demonstrated to reinforce both self-renewal and osteogenic differentiation of BMSCs. Mechanistically, Wnt7b activates the Ca2+ -dependent Nfatc1 signaling to directly induce Sox11 transcription, which in turn activates the transcriptions of both proliferation-related transcription factors (Ccnb1 and Sox2) and osteogenesis-related factors (Runx2, Sp7) in BMSCs. It is intriguing that this Wnt7b-Sox11 signaling in BMSCs is β-Catenin-independent. Overall, this study provides brand new insights of Wnt7b in bone formation, namely, Wnt7b can enhance both self-renewal and osteogenic differentiation of BMSCs via inducing Sox11. These findings present a new crosstalk between Wnt and Sox signaling in BMSCs.
Collapse
Affiliation(s)
- Fanyuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Feifei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Xueyang Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Yitian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Xin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
26
|
Cao L, Liu W, Zhong Y, Zhang Y, Gao D, He T, Liu Y, Zou Z, Mo Y, Peng S, Shuai C. Linc02349 promotes osteogenesis of human umbilical cord-derived stem cells by acting as a competing endogenous RNA for miR-25-3p and miR-33b-5p. Cell Prolif 2020; 53:e12814. [PMID: 32346990 PMCID: PMC7260076 DOI: 10.1111/cpr.12814] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 12/18/2022] Open
Abstract
Objectives Increasing evidences suggest that inducing mesenchymal stem cells to differentiate into osteoblasts has been as an especially important component in the prevention and therapy for degenerative bone disease. Here, we identify a novel lncRNA, linc02349, which increases significantly during osteogenic differentiation. Materials and methods Human umbilical cord‐derived stem cells (hUC‐MSCs) and dental pulp mesenchymal stem cells were used. Overexpression and knockdown of linc02349 in cell lines were generated using lentiviral‐mediated gene delivery method. Bioinformatics prediction, Ago2‐RIP assay and dual‐luciferase reporter system were employed to examine miRNA which interacts with linc02349. The RNA FISH assay was performed to identify the subcelluar location of linc02349. Alizarin Red S staining, ALP staining and qPCR were applied to identify the osteogenic differentiation. The potential linc02349‐regulated genes, miR‐25‐3p and miR‐33b‐5p, were explored by ChIP, RIP and Western blotting assays. Micro‐CT was used to measure the osteogenic content in bone formation assay in vivo. Results Linc02349 overexpression improves osteogenic differentiation by in vitro and in vivo analysis. Mechanistically, linc02349 acts as a molecular sponge for miR‐25‐3p and miR‐33b‐5p to control expression abundance of SMAD5 and Wnt10b, respectively, which eventually activated Dlx5/OSX pathway and hence promoted osteogenic differentiation. In addition, we revealed that STAT3 interacts with linc02349 promoter region and positively regulates the linc02349 transcriptional activity. Conclusion These findings identify that linc02349 modulates the osteogenic differentiation through acting as a sponge RNA of miR‐25‐3p and miR‐33b‐5p and regulating SMAD5 and Wnt10b, and proposed a new interaction between STAT3 and linc02349, which could be a potential target in the process the osteogenesis of hUC‐MSCs for future clinical application.
Collapse
Affiliation(s)
- Lihua Cao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wei Liu
- Institute of Metabolism and Endocrinology, Nation Clinical Research Center for Metabolic Diseases, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Yancheng Zhong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Yanru Zhang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Dan Gao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Tiantian He
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Ying Liu
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Zi Zou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Yuqing Mo
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China
| | - Shuping Peng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,School of basic Medical Science, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Cijun Shuai
- Institute of Bioadditive Manufacturing, Jiangxi University of Science and Technology, Nanchang, China.,State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha, China
| |
Collapse
|
27
|
Sun C, Wang L, Wang H, Huang T, Yao W, Li J, Zhang X. Single-cell RNA-seq highlights heterogeneity in human primary Wharton's jelly mesenchymal stem/stromal cells cultured in vitro. Stem Cell Res Ther 2020; 11:149. [PMID: 32252818 PMCID: PMC7132901 DOI: 10.1186/s13287-020-01660-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/10/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSCs) are multipotent cells with a promising application potential in regenerative medicine and immunomodulation. However, MSCs cultured in vitro exhibit functional heterogeneity. The underlying molecular mechanisms that define MSC heterogeneity remain unclear. METHODS We investigated the gene expression profile via single-cell RNA sequencing (scRNA-seq) of human primary Wharton's jelly-derived MSCs (WJMSCs) cultured in vitro from three donors. We also isolated CD142+ and CD142- WJMSCs based on scRNA-seq data and compared their proliferation capacity and "wound healing" potential in vitro. Meanwhile, we analyzed publicly available adipose-derived MSC (ADMSCs) scRNA-seq data and performed transcriptome comparison between WJMSCs and ADMSCs at the single-cell level. RESULTS GO enrichment analysis of highly variable genes (HVGs) obtained from WJMSCs revealed that these genes are significantly enriched in extracellular region with binding function, involved in developmental process, signal transduction, cell proliferation, etc. Pathway analysis showed that these HVGs are associated with functional characteristics of classic MSCs, such as inflammation mediated by chemokine and cytokine signaling, integrin signaling, and angiogenesis. After regressing out the batch and cell cycle effects, these HVGs were used for dimension reduction and clustering analysis to identify candidate subpopulations. Differentially expressed gene analysis revealed the existence of several distinct subpopulations of MSCs that exhibit diverse functional characteristics related to proliferation, development, and inflammation response. In line with our data, sorted CD142+ and CD142- WJMSCs showed distinct proliferation capacity as well as "wound healing" potential. Although WJMSCs and ADMSCs were derived from different tissues and were displaying different differentiation potencies, their HVGs were largely overlapped and had similar functional enrichment. CONCLUSION HVGs identified in MSCs are associated with classic MSC function. Regarding therapeutic potential, these genes are associated with functional characteristics, on which the MSC clinical application were theoretically based, such as development and inflammation response. Altogether, these HVGs hold the potential to be used as candidate markers for further potency association studies.
Collapse
Affiliation(s)
- Changbin Sun
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
- James D. Watson Institute of Genome Science, Hangzhou, 310008, China
| | - Lei Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
- James D. Watson Institute of Genome Science, Hangzhou, 310008, China
| | - Hailun Wang
- Department of Radiation Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Tingrun Huang
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
- James D. Watson Institute of Genome Science, Hangzhou, 310008, China
| | - Wenwen Yao
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jing Li
- BGI-Shenzhen, Shenzhen, 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Xi Zhang
- BGI-Shenzhen, Shenzhen, 518083, China.
- China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.
- James D. Watson Institute of Genome Science, Hangzhou, 310008, China.
| |
Collapse
|
28
|
Yun WS, Aryal S, Ahn YJ, Seo YJ, Key J. Engineered iron oxide nanoparticles to improve regenerative effects of mesenchymal stem cells. Biomed Eng Lett 2020; 10:259-273. [PMID: 32477611 DOI: 10.1007/s13534-020-00153-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/14/2020] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Abstract Mesenchymal stem cells (MSCs) based therapies are a major field of regenerative medicine. However, the success of MSC therapy relies on the efficiency of its delivery and retention, differentiation, and secreting paracrine factors at the target sites. Recent studies show that superparamagnetic iron oxide nanoparticles (SPIONs) modulate the regenerative effects of MSCs. After interacting with the cell membrane of MSCs, SPIONs can enter the cells via the endocytic pathway. The physicochemical properties of nanoparticles, including size, surface charge (zeta-potential), and surface ligand, influence their interactions with MSC, such as cellular uptake, cytotoxicity, homing factors, and regenerative related factors (VEGF, TGF-β1). Therefore, in-depth knowledge of the physicochemical properties of SPIONs might be a promising lead in regenerative and anti-inflammation research using SPIONs mediated MSCs. In this review, recent research on SPIONs with MSCs and the various designs of SPIONs are examined and summarized. Graphic abstract A graphical abstract describes important parameters in the design of superparamagnetic iron oxide nanoparticles, affecting mesenchymal stem cells. These physicochemical properties are closely related to the mesenchymal stem cells to achieve improved cellular responses such as homing factors and cell uptake.
Collapse
Affiliation(s)
- Wan Su Yun
- 1Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do South Korea
| | - Susmita Aryal
- 1Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do South Korea
| | - Ye Ji Ahn
- 2Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,3Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Young Joon Seo
- 2Research Institute of Hearing Enhancement, Yonsei University Wonju College of Medicine, Wonju, South Korea.,3Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Jaehong Key
- 1Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do South Korea
| |
Collapse
|
29
|
Characterization and therapeutic applications of mesenchymal stem cells for regenerative medicine. Tissue Cell 2020; 64:101330. [PMID: 32473704 DOI: 10.1016/j.tice.2020.101330] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/04/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent, genomic stable, self-renewable, and culturally expandable adult stem cells. MSCs facilitate tissue development, maintenance and repair, and produce secretory factors that support engraftment and trophic functions, marking them an attractive option in cell therapy, regenerative medicine and tissue engineering. METHOD In this review, we summarize the recent researches regarding the isolation and characterization of MSCs, therapeutic applications and advanced engineering techniques. We also discuss the advantages and limitations that remain to be overcome for MSCs based therapy. RESULTS It has been demonstrated that MSCs are able to modulate endogenous tissue and immune cells. Preclinical studies and early phase clinical trials have shown their great potential for tissue engineering of bone, cartilage, marrow stroma, muscle, fat, and other connective tissues. CONCLUSIONS MSC-based therapy show considerable promise to rebuild damaged or diseased tissues, which could be a promising therapeutic method for regeneration medicine.
Collapse
|
30
|
De D, Karmakar P, Bhattacharya D. Stem Cell Aging and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1326:11-37. [PMID: 32910426 DOI: 10.1007/5584_2020_577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stem cells are a promising source for regenerative medicine to cure a plethora of diseases that are currently treated based on either palliative or symptomatic relief or by preventing their onset and progression. Aging-associated degenerative changes in stem cells, stem cell niches, and signaling pathways bring a step by step decline in the regenerative and functional potential of tissues. Clinical studies and experiments on model organisms have pointed out checkpoints that aging will inevitably impose on stem cell aiming for transplantation and hence questions are raised about the age of the donor. In the following discourse, we review the fundamental molecular pathways that are implicated in stem cell aging and the current progress in tissue engineering and transplantation of each type of stem cells in regenerative medicine. We further focus on the consequences of stem cell aging on their clinical uses and the development of novel strategies to bypass those pitfalls and improve tissue replenishment.
Collapse
Affiliation(s)
- Debojyoti De
- Department of Life science and Biotechnology, Jadavpur University, Kolkata, India
| | - Parimal Karmakar
- Department of Life science and Biotechnology, Jadavpur University, Kolkata, India
| | | |
Collapse
|
31
|
Khademi-Shirvan M, Ghorbaninejad M, Hosseini S, Baghaban Eslaminejad M. The Importance of Stem Cell Senescence in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1288:87-102. [PMID: 32026416 DOI: 10.1007/5584_2020_489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are an interesting tool in regenerative medicine and a unique cell-based therapy to treat aging-associated diseases. Successful MSC therapy needs a large-scale cell culture, and requires a prolonged in vitro cell culture that subsequently leads to cell senescence. Administration of senescent MSCs results in inefficient cell differentiation in the clinical setting. Therefore, it is of utmost importance to enhance our knowledge about the aging process and methods to detect cell senescence in order to overcome this challenge. Numerous studies have addressed senescence in various aspects. Here, we review the characteristics of MSCs, how aging affects their features, mechanisms involved in aging of MSCs, and potential approaches to detect MSC senescence in vitro.
Collapse
Affiliation(s)
- Maliheh Khademi-Shirvan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
32
|
Bone regeneration of canine artificial alveolar clefts using bone-marrow-derived mesenchymal stromal cells and β-tricalcium phosphate: A preliminary study. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.odw.2012.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
33
|
Araque-Monrós MC, García-Cruz DM, Escobar-Ivirico JL, Gil-Santos L, Monleón-Pradas M, Más-Estellés J. Regenerative and Resorbable PLA/HA Hybrid Construct for Tendon/Ligament Tissue Engineering. Ann Biomed Eng 2019; 48:757-767. [DOI: 10.1007/s10439-019-02403-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022]
|
34
|
Long EG, Buluk M, Gallagher MB, Schneider JM, Brown JL. Human mesenchymal stem cell morphology, migration, and differentiation on micro and nano-textured titanium. Bioact Mater 2019; 4:249-255. [PMID: 31667441 PMCID: PMC6812408 DOI: 10.1016/j.bioactmat.2019.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/24/2019] [Accepted: 08/26/2019] [Indexed: 01/25/2023] Open
Abstract
Orthopedic implants rely on facilitating a robust interaction between the implant material surface and the surrounding bone tissue. Ideally, the interface will encourage osseointegration with the host bone, resulting in strong fixation and implant stability. However, implant failure can occur due to the lack of integration with bone tissue or bacterial infection. The chosen material and surface topography of orthopedic implants are key factors that influence the early events following implantation and may ultimately define the success of a device. Early attachment, rapid migration and improved differentiation of stem cells to osteoblasts are necessary to populate the surface of biomedical implants, potentially preventing biofilm formation and implant-associated infection. This article explores these early stem cell specific events by seeding human mesenchymal stem cells (MSCs) on four clinically relevant materials: polyether ether ketone (PEEK), Ti6Al4V (smooth Ti), macro-micro rough Ti6Al4V (Endoskeleton®), and macro-micro-nano rough Ti6Al4V (nanoLOCK®). The results demonstrate the incorporation of a hierarchical macro-micro-nano roughness on titanium produces a stellate morphology typical of mature osteoblasts/osteocytes, rapid and random migration, and improved osteogenic differentiation in seeded MSCs. Literature suggests rapid coverage of a surface by stem cells coupled with stimulation of bone differentiation minimizes the opportunity for biofilm formation while increasing the rate of device integration with the surrounding bone tissue.
Collapse
Affiliation(s)
- Emily G Long
- Department of Biomedical Engineering, The Pennsylvania State University, 122 CBEB Building, University Park, PA, 16802, USA
| | - Merve Buluk
- Department of Biomedical Engineering, The Pennsylvania State University, 122 CBEB Building, University Park, PA, 16802, USA
| | - Michelle B Gallagher
- Titan Spine, Inc., Mequon Research Center, 6140 W. Executive Drive, Suite A, Mequon, WI, 53092, USA
| | - Jennifer M Schneider
- Titan Spine, Inc., Mequon Research Center, 6140 W. Executive Drive, Suite A, Mequon, WI, 53092, USA
| | - Justin L Brown
- Department of Biomedical Engineering, The Pennsylvania State University, 122 CBEB Building, University Park, PA, 16802, USA
| |
Collapse
|
35
|
Kraft DL, Walck ER, Carrasco A, Crocker MD, Song L, Long MG, Mosse MA, Nadeem B, Imanbayev GT, Czechowicz AD, McCullough MJ. The MarrowMiner: A Novel Minimally Invasive and Effective Device for the Harvest of Bone Marrow. Biol Blood Marrow Transplant 2019; 26:219-229. [PMID: 31491487 DOI: 10.1016/j.bbmt.2019.08.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023]
Abstract
Bone marrow (BM) is a rich source of hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), and other important stem/progenitor cells. It is the traditional source of cells used in hematopoietic cell transplantation, which is a proven curative treatment for many blood and immune diseases. BM-derived cells have also been shown to have other diverse clinical uses and are increasingly being used in orthopedic medicine, regenerative medicine, and gene therapy applications. Traditional methods for harvesting BM are crude, tedious, time-consuming, and expensive, requiring multiple bone punctures under general anesthesia with serial small-volume aspirates often diluted with peripheral blood. The MarrowMiner (MM) is a novel device designed for rapid and minimally invasive BM harvest. Here we show the safety and efficacy of the MM in both preclinical and clinical settings. In a large-animal porcine model, the MM enabled effective BM collection with similar total nucleated cell collection and increased colony formation compared with standard methods. The MM was subsequently evaluated in a clinical study showing effective and complication-free anterior and posterior BM collection of 20 patients under only local anesthesia or light sedation. Increased total nucleated and mononucleated cell collection was achieved with the MM compared with standard methods in the same patients. Importantly, stem cell content was high with trends toward increased HSC, MSC, and endothelial progenitor cells with similar T cell content. Given the MM is a novel device approved by the US Food and Drug Administration, enabling safe, effective, and minimally invasive harvest of BM, we anticipate rapid adoption for various applications.
Collapse
Affiliation(s)
| | - Emily R Walck
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | | | | | - Lin Song
- Stryker Orthopaedics, Mahwah, New Jersey
| | | | - Maia A Mosse
- School of Medicine, Stanford University, Stanford, California
| | | | | | - Agnieszka D Czechowicz
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | | |
Collapse
|
36
|
Hladik D, Höfig I, Oestreicher U, Beckers J, Matjanovski M, Bao X, Scherthan H, Atkinson MJ, Rosemann M. Long-term culture of mesenchymal stem cells impairs ATM-dependent recognition of DNA breaks and increases genetic instability. Stem Cell Res Ther 2019; 10:218. [PMID: 31358047 PMCID: PMC6664790 DOI: 10.1186/s13287-019-1334-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are attracting increasing interest for cell-based therapies, making use of both their immuno-modulating and regenerative potential. For such therapeutic applications, a massive in vitro expansion of donor cells is usually necessary to furnish sufficient material for transplantation. It is not established to what extent the long-term genomic stability and potency of MSCs can be compromised as a result of this rapid ex vivo expansion. In this study, we investigated the DNA damage response and chromosomal stability (indicated by micronuclei induction) after sub-lethal doses of gamma irradiation in murine MSCs at different stages of their in vitro expansion. METHODS Bone-marrow-derived tri-potent MSCs were explanted from 3-month-old female FVB/N mice and expanded in vitro for up to 12 weeks. DNA damage response and repair kinetics after gamma irradiation were quantified by the induction of γH2AX/53BP1 DSB repair foci. Micronuclei were counted in post-mitotic, binucleated cells using an automated image analyzer Metafer4. Involvement of DNA damage response pathways was tested using chemical ATM and DNA-PK inhibitors. RESULTS Murine bone-marrow-derived MSCs in long-term expansion culture gradually lose their ability to recognize endogenous and radiation-induced DNA double-strand breaks. This impaired DNA damage response, indicated by a decrease in the number of γH2AX/53BP1 DSB repair foci, was associated with reduced ATM dependency of foci formation, a slower DNA repair kinetics, and an increased number of residual DNA double-strand breaks 7 h post irradiation. In parallel with this impaired efficiency of DNA break recognition and repair in older MSCs, chromosomal instability after mitosis increased significantly as shown by a higher number of micronuclei, both spontaneously and induced by γ-irradiation. Multifactorial regression analysis demonstrates that in vitro aging reduced DNA damage recognition in MSCs after irradiation by a multiplicative interaction with dose (p < 0.0001), whereas the increased frequency of micronuclei was caused by an additive interaction between in vitro aging and radiation dose. CONCLUSION The detrimental impact of long-term in vitro expansion on DNA damage response of MSCs warrants a regular monitoring of this process during the ex vivo growth of these cells to improve therapeutic safety and efficiency.
Collapse
Affiliation(s)
- Daniela Hladik
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany
| | - Ines Höfig
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany.,Present Address: BioNTech IMFS, Vollmersbachstr. 66, 55743, Idar-Oberstein, Germany
| | - Ursula Oestreicher
- BfS Federal Office for Radiation Protection, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Chair of Experimental Genetics, Technische Universität München, Wissenschaftszentrum Weihenstephan, 85354, Freising, Germany
| | - Martina Matjanovski
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany
| | - Xuanwen Bao
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany
| | - Harry Scherthan
- Bundeswehr Institute of Radiobiology, University of Ulm, Neuherbergstr. 11, 80937, Munich, Germany
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany.,Chair of Radiation Biology, Technical University of Munich, 81675, Munich, Germany
| | - Michael Rosemann
- Institute of Radiation Biology, Helmholtz Zentrum München GmbH, 85764, Neuherberg, Germany.
| |
Collapse
|
37
|
Allogeneic Adipose-Derived Mesenchymal Stem Cell Transplantation Enhances the Expression of Angiogenic Factors in a Mouse Acute Hindlimb Ischemic Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1083:1-17. [PMID: 28687961 DOI: 10.1007/5584_2017_63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell migration and molecular mechanisms during healing of damaged vascular or muscle tissues are emerging fields of interest worldwide. The study herein focuses on evaluating the role of allogenic adipose-derived mesenchymal stem cells (ADMSCs) in restoring damaged tissues. Using a hindlimb ischemic mouse model, ADMSC-mediated induction of cell migration and gene expression related to myocyte regeneration and angiogenesis were evaluated. ADMSCs were labeled with GFP (ADMSC-GFP). The proximal end of the femoral blood vessel of mice (over 6 months of age) are ligated at two positions then cut between the two ties. Hindlimb ischemic mice were randomly divided into two groups: Group I (n = 30) which was injected with PBS (100 μL) and Group II (n = 30) which was transplanted with ADMSC-GFP (106 cells/100 μL PBS) at the rectus femoris muscle. The migration of ADMSC-GFP in hindlimb was analyzed by UV-Vis system. The expression of genes related to angiogenesis and muscle tissue repair was quantified by real-time RT-PCR. The results showed that ADMSCs existed in the grafted hindlimb for 7 days. Grafted cells migrated to other damaged areas such as thigh and heel. In both groups the ischemic hindlimb showed an increased expression of several angiogenic genes, including Flt-1, Flk-1, and Ang-2. In particular, the expression of Ang-2 and myogenic-related gene MyoD was significantly increased in the ADMSC-treated group compared to the PBS-treated (control) group; the expression increased at day 28 compared to day 3. The other factors, such as VE-Cadherin, HGF, CD31, Myf5, and TGF-β, were also more highly expressed in the ADMSC-treated group than in the control group. Thus, grafted ADMSCs were able to migrate to other areas in the injured hindlimb, persist for approximately 7 days, and have a significantly positive impact on stimulating expression of myogenic- and angiogenesis-related genes.
Collapse
|
38
|
Fu J, Zhang Y, Chu J, Wang X, Yan W, Zhang Q, Liu H. Reduced Graphene Oxide Incorporated Acellular Dermal Composite Scaffold Enables Efficient Local Delivery of Mesenchymal Stem Cells for Accelerating Diabetic Wound Healing. ACS Biomater Sci Eng 2019; 5:4054-4066. [PMID: 33448807 DOI: 10.1021/acsbiomaterials.9b00485] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic skin wounds caused by diabetes mellitus (DM) have been acknowledged as one of the most intractable complications. Local transplantation of mesenchymal stem cells (MSCs) is a promising method, but strategies for stabilizing and efficiently delivering active MSCs according to the wound circumstance with high proteolysis remain the main barrier. Hereon, the study demonstrates the feasibility of incorporating reduced graphene oxide (RGO) nanoparticles with an acellular dermal matrix (ADM) to improve physicochemical characteristics of natural scaffold material and fabricate a highly efficient local transplantation system for MSCs in diabetic wound healing. Under the influence of RGO nanoparticles, the ADM-RGO composite scaffolds achieved high stability and strong mechanical behaviors. In vitro, conductive ADM-RGO scaffolds demonstrated an admirable milieu for stem cells adhesion and proliferation. After having been cocultured with MSCs, the ADM-RGO-MSC composite scaffolds were transplanted into the full-thickness wound of a diabetic model that was induced by streptozotocin (STZ) to evaluate its effects. As a result, the ADM-RGO composite scaffold delivered with MSCs supported robust vascularization and collagen deposition as well as rapid re-epithelialization during diabetic wound healing. Overall, the versatile nature of the ADM-RGO composite scaffold makes it an efficient transplanting mediator for pluripotent stem cells in tissue engineering applications. The composite scaffold delivered with MSCs presents a promising approach for nonhealing diabetic wounds.
Collapse
Affiliation(s)
- JinPing Fu
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| | - Yue Zhang
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| | - Jing Chu
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| | - Xiao Wang
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| | - WenXia Yan
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| | - Qiong Zhang
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| | - HanPing Liu
- Department of College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, Guangdong 510631, China
| |
Collapse
|
39
|
Yan Y, Cheng B, Chen K, Cui W, Qi J, Li X, Deng L. Enhanced Osteogenesis of Bone Marrow-Derived Mesenchymal Stem Cells by a Functionalized Silk Fibroin Hydrogel for Bone Defect Repair. Adv Healthc Mater 2019; 8:e1801043. [PMID: 30485718 DOI: 10.1002/adhm.201801043] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/05/2018] [Indexed: 12/25/2022]
Abstract
Silk fibroin (SF) from Bombyx mori is a promising natural material for the synthesis of biocompatible and biodegradable hydrogels for use in biomedical applications from tissue engineering to drug delivery. However, weak gelation performance and the lack of biochemical cues to trigger cell proliferation and differentiation currently significantly limit its application in these areas. Herein, a biofunctional hydrogel containing SF (2.0%) and a small peptide gelator (e.g., NapFFRGD = 1.0 wt%) is generated via cooperative molecular self-assembly. The introduction of NapFFRGD to SF is shown to significantly improve its gelation properties by lowering both its threshold gelation concentration to 2.0% and gelation time to 20 min under physiological conditions (pH = 7.4, 37 °C), as well as functionalizing the SF hydrogel with cell-adhesive motifs (e.g., RGD). Besides mediating cell adhesion, the RGD ligands incorporated within the SF-RGD gel promote the osteogenic differentiation of bone marrow-derived mesenchymal stem cells encapsulated within the gel matrix, leading to bone regeneration in a mouse calvarial defect model, compared with a blank SF gel (2.0%, pH = 7.4). This work suggests that SF could be easily tailored with bioactive peptide gelators to afford bioactive hydrogels with favorable microenvironments for tissue regeneration applications.
Collapse
Affiliation(s)
- Yufei Yan
- Shanghai Key Laboratory for Bone and Joint Diseases; Shanghai Institute of Orthopaedics and Traumatology; Shanghai Ruijin Hospital; Shanghai Jiaotong University; Shanghai 200025 China
| | - Baochang Cheng
- College of Chemistry; Chemical Engineering and Materials Science; Soochow University; Suzhou 215123 China
| | - Kaizhe Chen
- Shanghai Key Laboratory for Bone and Joint Diseases; Shanghai Institute of Orthopaedics and Traumatology; Shanghai Ruijin Hospital; Shanghai Jiaotong University; Shanghai 200025 China
| | - Wenguo Cui
- Shanghai Key Laboratory for Bone and Joint Diseases; Shanghai Institute of Orthopaedics and Traumatology; Shanghai Ruijin Hospital; Shanghai Jiaotong University; Shanghai 200025 China
| | - Jin Qi
- Shanghai Key Laboratory for Bone and Joint Diseases; Shanghai Institute of Orthopaedics and Traumatology; Shanghai Ruijin Hospital; Shanghai Jiaotong University; Shanghai 200025 China
| | - Xinming Li
- College of Chemistry; Chemical Engineering and Materials Science; Soochow University; Suzhou 215123 China
| | - Lianfu Deng
- Shanghai Key Laboratory for Bone and Joint Diseases; Shanghai Institute of Orthopaedics and Traumatology; Shanghai Ruijin Hospital; Shanghai Jiaotong University; Shanghai 200025 China
| |
Collapse
|
40
|
Yasunaga M, Kajiya H, Toshimitsu T, Nakashima H, Tamaoki S, Ishikawa H, Maeda H, Ohno J. The Early Autophagic Pathway Contributes to Osteogenic Differentiation of Human Periodontal Ligament Stem Cells. J HARD TISSUE BIOL 2019. [DOI: 10.2485/jhtb.28.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Madoka Yasunaga
- Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College
- Research Center for Regenerative Medicine, Fukuoka Dental College
| | - Hiroshi Kajiya
- Research Center for Regenerative Medicine, Fukuoka Dental College
- Section of Cellular Physiology, Department of Physiological Science and Molecular Biology, Fukuoka Dental College
| | - Takuya Toshimitsu
- Research Center for Regenerative Medicine, Fukuoka Dental College
- Dentistry for the Disabled, Department of Oral Growth and Development, Fukuoka Dental College
| | - Hiroki Nakashima
- Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College
- Research Center for Regenerative Medicine, Fukuoka Dental College
| | - Sachio Tamaoki
- Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College
| | - Hiroyuki Ishikawa
- Former Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College
| | - Hidefumi Maeda
- Division of Oral Rehabilitation, Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University
| | - Jun Ohno
- Research Center for Regenerative Medicine, Fukuoka Dental College
| |
Collapse
|
41
|
Bae EB, Yoo JH, Jeong SI, Kim MS, Lim YM, Ahn JJ, Lee JJ, Lee SH, Kim HJ, Huh JB. Effect of Titanium Implants Coated with Radiation-Crosslinked Collagen on Stability and Osseointegration in Rat Tibia. MATERIALS 2018; 11:ma11122520. [PMID: 30545019 PMCID: PMC6316992 DOI: 10.3390/ma11122520] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022]
Abstract
This study aimed to evaluate the titanium (Ti) implants coated with collagen type Ⅰ crosslinked using gamma-irrigation or glutaraldehyde (GA). The in vitro surface observations, quantification assay, and cell studies using human mesenchymal stem cells (hMSCs) were conducted. For in vivo experiments, the implants were divided into three groups and inserted into the rat tibias: control group (non-treated Ti implant), GA group (Ti implants coated with GA-crosslinked collagen) and 25 kGy group (Ti implants coated with gamma-radiation-crosslinked collagen at dose of 25 kGy). The animals were sacrificed at 4 weeks after implantation and the tissue sections were obtained. New bone volume (mm³) and bone-to-implant contact (BIC, %) within the region of interest (ROI) was measured. The in vitro results showed the highest osteogenic differentiation and levels of osteogenesis-related gene expressions in the 25 kGy group without cytotoxicity. The new bone volume of GA group was significantly higher than the control (p < 0.05). In the result of the BIC, the 25 kGy group was significantly higher than the control (p < 0.05). However, there was no significant difference between the experimental groups. Within the limitations of this study, Ti implant coated with gamma-radiation-crosslinked collagen has potential utility without side effects from chemical agents.
Collapse
Affiliation(s)
- Eun-Bin Bae
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Ji-Hyun Yoo
- Department of Oral Physiology, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Sung-In Jeong
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 1266 Sinjeong-dong, Jeongeup-si, Jeollabuk-do 56212, Korea.
| | - Min-Su Kim
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Youn-Mook Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 1266 Sinjeong-dong, Jeongeup-si, Jeollabuk-do 56212, Korea.
| | - Jong-Ju Ahn
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Jin-Ju Lee
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - So-Hyoun Lee
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Hyung-Joon Kim
- Department of Oral Physiology, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Jung-Bo Huh
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
42
|
Sane MS, Misra N, Mousa OM, Czop S, Tang H, Khoo LT, Jones CD, Mustafi SB. Cytokines in umbilical cord blood-derived cellular product: a mechanistic insight into bone repair. Regen Med 2018; 13:881-898. [PMID: 30346891 PMCID: PMC6439518 DOI: 10.2217/rme-2018-0102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022] Open
Abstract
AIM Umbilical cord blood (UCB) finds frequent applications in regenerative medicine. We evaluated the role of cytokines present in a uniquely processed, UCB-derived cellular allograft product (UCBp). MATERIALS & METHODS Luminex multiplex assay and standard cell biology methods were employed. RESULTS Study with allografts from 33 donors identified 44 quantifiable cytokines in the UCBp derived conditioned media (CM). The UCBp-CM elevated proliferation and migration rates of mesenchymal stem cells (MSCs) and bone marrow stromal cells. Moreover, UCBp-CM induced secretion of VEGF-A and osteoprotegerin, which promoted angiogenesis of endothelial cells and positively influenced the osteogenic differentiation of MSCs, respectively. CONCLUSION Cytokines in UCBp stimulate cellular processes important for bone regeneration, making UCBp an excellent candidate for potential applications in orthopedic procedures like bone non-union and spinal fusion.
Collapse
Affiliation(s)
- Mukta S Sane
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
| | - Neha Misra
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
| | - Omid Mohammad Mousa
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
- Department of Regulatory & Medical Affairs, Burst Biologics, Boise, ID 83705, USA
- Biomolecular Research Center, Boise State University, Boise, ID, 83725, USA
| | - Steve Czop
- Department of Regulatory & Medical Affairs, Burst Biologics, Boise, ID 83705, USA
| | - Huiyuan Tang
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
| | - Larry T Khoo
- The Spine Clinic of Los Angeles, Good Samaritan Hospital, University of Southern California, Los Angeles, CA 90017, USA
| | - Christopher D Jones
- Department of Research & Development, Burst Biologics, Boise, ID 83705, USA
- Department of Regulatory & Medical Affairs, Burst Biologics, Boise, ID 83705, USA
| | | |
Collapse
|
43
|
Alshareeda AT, Rakha E, Alghwainem A, Alrfaei B, Alsowayan B, Albugami A, Alsubayyil AM, Abomraee M, Mohd Zin NK. The effect of human placental chorionic villi derived mesenchymal stem cell on triple-negative breast cancer hallmarks. PLoS One 2018; 13:e0207593. [PMID: 30458011 PMCID: PMC6245746 DOI: 10.1371/journal.pone.0207593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/03/2018] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can influence the tumour microenvironment (TEM) and play a major role in tumourigenesis. Triple-negative [Ostrogen receptor (ER-), Progesterone receptor (PgR-), and HER2/neu receptor (HER2-)] breast cancer (TNBC) is an aggressive class of BC characterized by poor prognosis and lacks the benefit of routinely available targeted therapies. This study aims to investigate the effect of human placental chorionic villi derived MSCs (CVMSCs) on the behavior of TNBC in vitro. This was done by assaying different cancer hallmarks including proliferation, migration and angiogenesis. Cell proliferation rate of TNBC cell line (MDA-MB231) was monitored in real time using the xCELLigence system. Whereas, Boyden chamber migration assay was used to measure MDA-MB231 motility and invasiveness toward CVMSCs. Finally, a three-dimensional (3D) model using a co-culture system of CVMSCs with MDA-MB231 with or without the addition of human umbilical vein endothelial cells (HUVECs) was created to assess tumour angiogenesis in vitro. CVMSCs were able to significantly reduce the proliferative and migratory capacity of MDA-MB231 cells. Co-culturing of MDA-MB231 with CVMSCs, not only inhibited the tube formation ability of HUVECs but also reduced the expression of the BC characteristic cytokines; IL-10, IL-12, CXCL9 and CXCL10 of CVMSCs. These results support the hypothesis that CVMSCs can influence the behavior of TNBC cells and provides a basic for a potential therapeutic approach in a pre-clinical settings. The data from this study also highlight the complexity of the in vitro cancer angiogenesis model settings and regulations.
Collapse
Affiliation(s)
- Alaa T Alshareeda
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Emad Rakha
- University of Nottingham and Nottingham University Hospitals NHS Trust, City Hospital, Department of Cellular Pathology, UK, Nottingham, United Kingdom
| | - Ayidah Alghwainem
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Bahauddeen Alrfaei
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Batla Alsowayan
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Abdullah Albugami
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Abdullah M Alsubayyil
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | - Mohmed Abomraee
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Saudi Arabia
| | | |
Collapse
|
44
|
Linard C, Brachet M, L’homme B, Strup-Perrot C, Busson E, Bonneau M, Lataillade JJ, Bey E, Benderitter M. Long-term effectiveness of local BM-MSCs for skeletal muscle regeneration: a proof of concept obtained on a pig model of severe radiation burn. Stem Cell Res Ther 2018; 9:299. [PMID: 30409227 PMCID: PMC6225585 DOI: 10.1186/s13287-018-1051-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Medical management of the severe musculocutaneous radiation syndrome involves surgical intervention with debridement of necrotic tissue. Even when skin excision is replaced by specific plastic surgery, treatment of the muscle radiation injury nonetheless remains difficult, for it involves a massive muscle defect in an unpredictable environment, subject to inflammatory waves weeks to months after irradiation, which delay healing and predispose the patient to the development of fibrous scar tissue. In this study, we investigated the long-term effect of local injections of bone marrow-derived mesenchymal stromal cells (BM-MSCs), combined with plastic surgery, to treat muscle necrosis in a large animal model. METHODS Three months after irradiation to the rump, minipigs were treated by excision of necrotic muscle tissue, vascularized flap surgery, and four injections with or without local autologous BM-MSCs, performed weekly. The quality of the muscle wound healing was examined 1 year post-surgery. RESULTS The skeletal muscle surgery without MSC treatment led to permanent deposition of collagen 1 and 3, decreased myofiber diameter, failed muscle fiber regeneration, a reduced number of capillaries, and the accumulation of high calcium and fat. In animals treated by surgery and MSC injections, these indicators were substantially better and demonstrated established regeneration. MSC therapy acts at several levels by stimulating growth factors such as VEGF, which is involved in angiogenesis and satellite cell pool maintenance, and creating a macrophage M1/M2 balance. CONCLUSION Thus, cell therapy using BM-MSCs is an effective and safe way to improve recovery of irradiation-induced skeletal muscle damage without signs of long-term degeneration.
Collapse
Affiliation(s)
- Christine Linard
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
- Unité de Thérapie Tissulaire et Traumatologie de Guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Michel Brachet
- Department of Plastic Surgery, Military Hospital of Percy, Clamart, France
| | - Bruno L’homme
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
| | - Carine Strup-Perrot
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
| | - Elodie Busson
- Unité des Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Michel Bonneau
- Centre of Research in Interventional Imaging, National Institut of Agronomic Research, Jouy-en-Josas, France
| | - Jean-Jacques Lataillade
- Unité de Thérapie Tissulaire et Traumatologie de Guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Eric Bey
- Department of Plastic Surgery, Military Hospital of Percy, Clamart, France
| | - Marc Benderitter
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
| |
Collapse
|
45
|
O'Valle F, de Buitrago JG, Hernández-Cortés P, Padial-Molina M, Crespo-Lora V, Cobo M, Aguilar D, Galindo-Moreno P. Increased Expression of Musashi-1 Evidences Mesenchymal Repair in Maxillary Sinus Floor Elevation. Sci Rep 2018; 8:12243. [PMID: 30116022 PMCID: PMC6095840 DOI: 10.1038/s41598-018-29908-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/17/2018] [Indexed: 11/21/2022] Open
Abstract
This study aimed to analyze the expression of Musashi-1 (MSI1) in maxillary native bone and grafted bone after maxillary sinus floor elevation. To do so, fifty-seven bone biopsies from 45 participants were studied. Eighteen samples were collected from native bone while 39 were obtained 6 months after maxillary sinus grafting procedures. Musashi-1 was analyzed by immunohistochemistry and RT-PCR. MSI1 was detected in osteoblasts and osteocytes in 97.4% (38/39) of grafted areas. In native bone, MSI1 was detected in only 66.6% (12/18) of the biopsies, mainly in osteocytes. Detection of MSI1 was significantly higher in osteoprogenitor mesenchymal cells of grafted biopsies (p < 0.001) but minor in smooth muscle and endothelial cells; no expression was detected in adipocytes. The mesenchymal cells of the non-mineralized tissue of native bone showed very low nuclear expression of MSI1, in comparison to fusiform cells in grafted areas (0.28(0.13) vs. 2.10(0.14), respectively; p < 0.001). Additionally, the detection of MSI1 mRNA was significantly higher in biopsies from grafted areas than those from native bone (1.00(0.51) vs. 60.34(35.2), respectively; p = 0.029). Thus, our results regardig the significantly higher detection of Musashi-1 in grafted sites than in native bone reflects its importance in the remodeling/repair events that occur after maxillary sinus floor elevation in humans.
Collapse
Affiliation(s)
- Francisco O'Valle
- Department of Pathology & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary (Ibs-Granada), University of Granada, Granada, Spain
| | - Juan G de Buitrago
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
| | - Pedro Hernández-Cortés
- Department of Orthopedic Surgery, San Cecilio University Hospital of Granada, Granada, Spain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
| | - Vicente Crespo-Lora
- Department of Pathology & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Marien Cobo
- Department of Genomic Medicine & GENYO, Centre for Genomics and Oncological Research, Pfizer - University of Granada - Andalusian Regional Government, Granada, Spain
| | - David Aguilar
- Department of Pathology & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain.
| |
Collapse
|
46
|
Oh M, Lee J, Kim YJ, Rhee WJ, Park JH. Exosomes Derived from Human Induced Pluripotent Stem Cells Ameliorate the Aging of Skin Fibroblasts. Int J Mol Sci 2018; 19:ijms19061715. [PMID: 29890746 PMCID: PMC6032439 DOI: 10.3390/ijms19061715] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022] Open
Abstract
Stem cells and their paracrine factors have emerged as a resource for regenerative medicine. Many studies have shown the beneficial effects of paracrine factors secreted from adult stem cells, such as exosomes, on skin aging. However, to date, few reports have demonstrated the use of exosomes derived from human pluripotent stem cells for the treatment of skin aging. In this study, we collected exosomes from the conditioned medium of human induced pluripotent stem cells (iPSCs) and investigated the effect on aged human dermal fibroblasts (HDFs). Cell proliferation and viability were determined by an MTT assay and cell migration capacity was shown by a scratch wound assay and a transwell migration assay. To induce photoaging and natural senescence, HDFs were irradiated by UVB (315 nm) and subcultured for over 30 passages, respectively. The expression level of certain mRNAs was evaluated by quantitative real-time PCR (qPCR). Senescence-associated-β-galactosidase (SA-β-Gal) activity was assessed as a marker of natural senescence. As a result, we found that exosomes derived from human iPSCs (iPSCs-Exo) stimulated the proliferation and migration of HDFs under normal conditions. Pretreatment with iPSCs-Exo inhibited the damages of HDFs and overexpression of matrix-degrading enzymes (MMP-1/3) caused by UVB irradiation. The iPSCs-Exo also increased the expression level of collagen type I in the photo-aged HDFs. In addition, we demonstrated that iPSCs-Exo significantly reduced the expression level of SA-β-Gal and MMP-1/3 and restored the collagen type I expression in senescent HDFs. Taken together, it is anticipated that these results suggest a therapeutic potential of iPSCs-Exo for the treatment of skin aging.
Collapse
Affiliation(s)
- Myeongsik Oh
- Department of Medical Biomaterials Engineering, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341, Korea.
| | - Jinhee Lee
- Division of Bioengineering, Incheon National University, Incheon 22012, Korea.
| | - Yu Jin Kim
- Department of Medical Biomaterials Engineering, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341, Korea.
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon 22012, Korea.
| | - Ju Hyun Park
- Department of Medical Biomaterials Engineering, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341, Korea.
- Institute of Bioscience and Biotechnology, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341, Korea.
| |
Collapse
|
47
|
Abumaree MH, Bahattab E, Alsadoun A, Al Dosaimani A, Abomaray FM, Khatlani T, Kalionis B, El-Muzaini MF, Alawad AO, AlAskar AS. Characterization of the interaction between human decidua parietalis mesenchymal stem/stromal cells and natural killer cells. Stem Cell Res Ther 2018; 9:102. [PMID: 29650045 PMCID: PMC5898063 DOI: 10.1186/s13287-018-0844-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Background Human decidua parietalis mesenchymal stem/multipotent stromal cells (DPMSCs) have unique phenotypic and functional properties that make them promising candidates for cell-based therapy. Here, we investigated DPMSC interaction with natural killer (NK) cells, and the effects of this interaction on NK cell phenotypic characteristics and functional activities. Methods DPMSCs isolated from the decidua parietalis of human fetal membranes were cultured with interleukin (IL)-2-activated and IL-2-unactivated NK cells isolated from healthy human peripheral blood. NK cell proliferation and cytolytic activities were then examined using functional assays. NK cell expression of receptors mediating the cytolytic activity against DPMSCs, and the mechanism underlying this effect on DPMSCs, were also examined using flow cytometry and light microscopy, respectively. Results DPMSCs stimulated IL-2-induced proliferation of resting NK cells and the proliferation of activated NK cells. Moreover, IL-2-activated NK cells, but not freshly isolated NK cells, efficiently lysed DPMSCs. The induction of this NK cell cytolytic activity against DPMSCs was mediated by the activating NK cell receptors NKG2D, CD69, NKp30, and NKp44. However, DPMSCs showed a direct induction of NK cell cytolytic activity through CD69. We also found that DPMSCs expressed the ligands for these activating NK cell receptors including Nectin-2, ULBP-2, MICA, and MICB. Although DPMSCs expressed HLA class I molecules, they were susceptible to lysis by NK cells, suggesting that HLA class I antigens do not play a significant role in NK cell cytolytic action. In addition, DPMSCs did not inhibit NK cell cytolytic activity against cancer cells. Importantly, DPMSCs significantly increased NK expression of inflammatory molecules with anticancer activities. Conclusions We conclude that DPMSCs have potential for therapeutic application in cancer therapy, but not in transplantation or immunological diseases.
Collapse
Affiliation(s)
- M H Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia. .,College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia.
| | - E Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - A Alsadoun
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - A Al Dosaimani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia
| | - F M Abomaray
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, 14186, Stockholm, Sweden.,Center for Hematology and Regenerative Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - T Khatlani
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia
| | - B Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, VIC, 3052, Australia
| | - M F El-Muzaini
- Department of Obstetrics and Gynaecology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia
| | - A O Alawad
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - A S AlAskar
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia.,College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia.,Adult Hematology and Stem Cell Transplantation, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia
| |
Collapse
|
48
|
|
49
|
Sevivas N, Teixeira FG, Portugal R, Direito-Santos B, Espregueira-Mendes J, Oliveira FJ, Silva RF, Sousa N, Sow WT, Nguyen LTH, Ng KW, Salgado AJ. Mesenchymal Stem Cell Secretome Improves Tendon Cell Viability In Vitro and Tendon-Bone Healing In Vivo When a Tissue Engineering Strategy Is Used in a Rat Model of Chronic Massive Rotator Cuff Tear. Am J Sports Med 2018; 46:449-459. [PMID: 29053925 DOI: 10.1177/0363546517735850] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Massive rotator cuff tears (MRCTs) represent a major clinical concern, especially when degeneration and chronicity are involved, which highly compromise healing capacity. PURPOSE To study the effect of the secretome of mesenchymal stem cells (MSCs) on tendon cells (TCs) followed by the combination of these activated TCs with an electrospun keratin-based scaffold to develop a tissue engineering strategy to improve tendon-bone interface (TBi) healing in a chronic MRCT rat model. STUDY DESIGN Controlled laboratory study. METHODS Human TCs (hTCs) cultured with the human MSCs (hMSCs) secretome (as conditioned media [CM]) were combined with keratin electrospun scaffolds and further implanted in a chronic MRCT rat model. Wistar-Han rats (N = 15) were randomly assigned to 1 of 3 groups: untreated lesion (MRCT group, n = 5), lesion treated with a scaffold only (scaffold-only group, n = 5), and lesion treated with a scaffold seeded with hTCs preconditioned with hMSCs-CM (STC_hMSC_CM group, n = 5). After sacrifice, 16 weeks after surgery, the rotator cuff TBi was harvested for histological analysis and biomechanical testing. RESULTS The hMSCs secretome increased hTCs viability and density in vitro. In vivo, a significant improvement of the tendon maturing score was observed in the STC_hMSC_CM group (mean ± standard error of the mean, 15.6 ± 1.08) compared with the MRCT group (11.0 ± 1.38; P < .05). Biomechanical tests revealed a significant increase in the total elongation to rupture (STC_hMSC_CM, 11.99 ± 3.30 mm; scaffold-only, 9.89 ± 3.47 mm; MRCT, 5.86 ± 3.16 mm; P < .05) as well as a lower stiffness (STC_hMSC_CM, 6.25 ± 1.74 N/mm; scaffold-only, 6.72 ± 1.28 N/mm; MRCT, 11.54 ± 2.99 N/mm; P < .01). CONCLUSION The results demonstrated that hMSCs-CM increased hTCs viability and density in vitro. Clear benefits also were observed when these primed cells were integrated into a tissue engineering strategy with an electrospun keratin scaffold, as evidenced by improved histological and biomechanical properties for the STC_hMSC_CM group compared with the MRCT group. CLINICAL RELEVANCE This work supports further investigation into the use of MSC secretome for priming TCs toward a more differentiated phenotype, and it promotes the tissue engineering strategy as a promising modality to help improve treatment outcomes for chronic MRCTs.
Collapse
Affiliation(s)
- Nuno Sevivas
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Orthopaedics Department, Hospital de Braga and Hospital Privado de Braga, Braga, Portugal.,Clínica Espregueira-Mendes, FIFA Medical Centre of Excellence, Estádio do Dragão, Porto, Portugal
| | - Fábio Gabriel Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Bruno Direito-Santos
- Orthopaedics Department, Hospital de Braga and Hospital Privado de Braga, Braga, Portugal
| | - João Espregueira-Mendes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Clínica Espregueira-Mendes, FIFA Medical Centre of Excellence, Estádio do Dragão, Porto, Portugal.,3B's Research Group, Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
| | - Filipe J Oliveira
- CICECO, Department of Materials and Ceramic Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Rui F Silva
- CICECO, Department of Materials and Ceramic Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Wan Ting Sow
- School of Materials Science and Engineering, Nanyang Technological University, Singapore
| | - Luong T H Nguyen
- School of Materials Science and Engineering, Nanyang Technological University, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
50
|
Generating Chondromimetic Mesenchymal Stem Cell Spheroids by Regulating Media Composition and Surface Coating. Cell Mol Bioeng 2017; 11:99-115. [PMID: 29623134 DOI: 10.1007/s12195-017-0517-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Spheroids of mesenchymal stem cells (MSCs) in cartilage tissue engineering have been shown to enhance regenerative potential owing to their 3D structure. In this study, we explored the possibility of priming spheroids under different media to replace the use of inductive surface coatings for chondrogenic differentiation. METHODS Rat bone marrow-derived MSCs were organized into cell spheroids by the hanging drop technique and subsequently cultured on hyaluronic acid (HA) coated or non-coated well plates under different cell media conditions. Endpoint analysis included cell viability, DNA and Glycosaminoglycan (GAG) and collagen content, gene expression and immunohistochemistry. RESULTS For chondrogenic applications, MSC spheroids derived on non-coated surfaces outperformed the spheroids derived from HA-coated surfaces in matrix synthesis and collagen II gene expression. Spheroids on non-coated surfaces gave rise to the highest collagen and GAG when primed with medium containing insulin-like growth factor (IGF) for 1 week during spheroid formation. Spheroids that were grown in chondroinductive raw material-inclusive media such as aggrecan or chondroitin sulfate exhibited the highest Collagen II gene expression in the non-coated surface at 1 week. CONCLUSION Media priming by growth factors and raw materials might be a more predictive influencer of chondrogenesis compared to inductive-surfaces. Such tailored bioactivity of the stem cell spheroids in the stage of the spheroid formation may give rise to a platform technology that may eventually produce spheroids capable of chondrogenesis achieved by mere media manipulation, skipping the need for additional culture on a modified surface, that paves the way for cost-effective technologies.
Collapse
|