1
|
Mante N, Undale V, Sanap A, Bhonde R, Tambe P, Bansode M, Gupta RK. Disease microenvironment preconditioning: An evolving approach to improve therapeutic efficacy of human mesenchymal stromal cells. Int Immunopharmacol 2025; 157:114701. [PMID: 40300358 DOI: 10.1016/j.intimp.2025.114701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
Despite the tremendous success in preclinical models, the translation of human mesenchymal stromal cells (hMSCs) as a therapy in the clinic is not up to the expectation. Intrinsic factors (age, sex, health status, life style of the donor, source, cellular senescence, and oxidative stress in hMSCs), extrinsic factors (culture system, batch-to-batch variations, choice of biomaterials, cell processing and preservation protocols), and host microenvironment (inflammatory milieu, oxidative stress, and hypoxia in the recipient) compromise the overall therapeutic efficacy of the transplanted hMSCs. In recent times, the approach of 'Disease Microenvironment Preconditioning (DMP)' has garnered attention to overcome the host-associated attributes involved in compromised hMSCs therapeutic potential. In this review, we discuss various approaches of DMP of hMSCs by employing serum and other body fluids obtained from diseased patients/animals and small molecules, including cytokines such as IFN-γ, IL-6, IL-10, IL- β, TGF-β1, IL-1α, IL-1β, TNF-α, HMGB1, IL-17 A, and IL-8 which are associated with disease conditions. DMP strengthens hMSCs ability to adapt/acclimatize and respond more efficiently to the hostile microenvironment they encounter upon transplantation. DMP modulate hMSCs to withstand inflammation, survive under hypoxic and nutrient-deprived conditions, and resist oxidative stress. Evidence from various disease models ranging from cardiovascular and neurodegenerative disorders to autoimmune diseases and tissue injuries supports the role of DMP in improving hMSC survival, integration, and functional efficacy. While the potential of DMP to revolutionize MSC-based therapies is evident, challenges such as standardizing/optimizing protocols for preconditioning is essential. This review synthesizes current advancements in the approach of DMP aiming to propel the area of regenerative medicine.
Collapse
Affiliation(s)
- Nishant Mante
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India
| | - Vaishali Undale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India.
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India.
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India
| | - Pratima Tambe
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India
| | - Manoj Bansode
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| |
Collapse
|
2
|
Koohi-Hosseinabadi O, Shahriarirad R, Dehghanian A, Amini L, Barzegar S, Daneshparvar A, Alavi O, Khazraei SP, Hosseini S, Arabi Monfared A, Khorram R, Tanideh N, Ashkani-Esfahani S. In-vitro and in-vivo assessment of biocompatibility and efficacy of ostrich eggshell membrane combined with platelet-rich plasma in Achilles tendon regeneration. Sci Rep 2025; 15:841. [PMID: 39755875 PMCID: PMC11700202 DOI: 10.1038/s41598-025-85131-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025] Open
Abstract
Tendon injuries present significant medical, social, and economic challenges globally. Despite advancements in tendon injury repair techniques, outcomes remain suboptimal due to inferior tissue quality and functionality. Tissue engineering offers a promising avenue for tendon regeneration, with biocompatible scaffolds playing a crucial role. Ostrich eggshell membrane (ESM), characterized by a strong preferential orientation of calcite crystals, forms a semipermeable polymer network with excellent mechanical properties compared to membranes from other bird species, emerging as a potential natural scaffold candidate. Coupled with platelet-rich plasma (PRP), known for its regenerative properties, ESM holds promise for improving tendon repair. This study aims to evaluate the biocompatibility and efficacy of an ESM-PRP scaffold in treating Achilles tendon ruptures, employing in vitro and in vivo assessments to gauge its potential in tendon regeneration in living organisms. Ostrich ESM was prepared from pathogen-free ostrich eggs, sterilized with UV radiation and prepared in desired dimensions before implantation (1.5 × 1 cm). High-resolution scanning electron microscopy (HRSEM) was utilized to visualize the sample morphology and fiber bonding. In vitro biocompatibility was assessed using the MTT assay and DAPI staining, while in vivo biocompatibility was evaluated in a rat model. For the in vivo Achilles tendinopathy assay, rats were divided into groups and subjected to AT rupture followed by treatment with ESM, PRP, or a combination. SEM was employed to evaluate tendon morphology, and real-time PCR was conducted to analyze gene expression levels. The in vivo assay indicated that the ESM scaffold was safe for an extended period of 8 weeks, showing no signs of inflammation based on histopathological analysis. In the Achilles tendon rupture model, combining ESM with PRP enhanced tendon healing after 14 weeks post-surgery. This finding was supported by histopathological, morphological, and mechanical evaluations of tendon tissues compared to normal tendons, untreated tendinopathy, and injured tendons treated with the ESM scaffold. Gene expression analysis revealed significantly increased expression of Col1a1, Col3a1, bFGF, Scleraxis (Scx), and tenomodulin in the ESM-PRP groups. The findings of our study demonstrate that the combination of Ostrich ESM with PRP significantly enhances AT repair and is a biocompatible scaffold for the application in living organisms.
Collapse
Affiliation(s)
- Omid Koohi-Hosseinabadi
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Central Research Laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Shahriarirad
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amireza Dehghanian
- Department of Pathology, School of Medicine, Shiraz University, Shiraz, Iran
| | - Laleh Amini
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Sajjad Barzegar
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - Afrooz Daneshparvar
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Alavi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Ali Arabi Monfared
- Department of Medical Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Roya Khorram
- Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, P. O. Box: 7134845794, Shiraz, Iran.
- Pharmacology Department, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Soheil Ashkani-Esfahani
- Foot and Ankle Research and Innovation Lab (FARIL), Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Foot and Ankle Division, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Sankova MV, Beeraka NM, Oganesyan MV, Rizaeva NA, Sankov AV, Shelestova OS, Bulygin KV, Vikram PR H, Barinov A, Khalimova A, Padmanabha Reddy Y, Basappa B, Nikolenko VN. Recent developments in Achilles tendon risk-analyzing rupture factors for enhanced injury prevention and clinical guidance: Current implications of regenerative medicine. J Orthop Translat 2024; 49:289-307. [PMID: 39559294 PMCID: PMC11570240 DOI: 10.1016/j.jot.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 11/20/2024] Open
Abstract
Background In recent years, many countries have actively implemented programs and strategies to promote physical education and sports. Despite these efforts, the increase in physical activity has been accompanied by a significant rise in muscle and tendon-ligament injuries, with Achilles tendon rupture being the most prevalent, accounting for 47 % of such injuries. This review aims to summarize all significant factors determining the predisposition of the Achilles tendon to rupture, to develop effective personalized prevention measures. Objective To identify and evaluate the risk factors contributing to Achilles tendon rupture and to develop strategies for personalized prevention. Methods This review utilized data from several databases, including Elsevier, Global Health, PubMed-NCBI, Embase, Medline, Scopus, ResearchGate, RSCI, Cochrane Library, Google Scholar, eLibrary.ru, and CyberLeninka. Both non-modifiable and modifiable risk factors for Achilles tendon injuries and ruptures were analyzed. Results The analysis identified several non-modifiable risk factors, such as genetic predisposition, anatomical and functional features of the Achilles tendon, sex, and age. These factors should be considered when selecting sports activities and designing training programs. Modifiable risk factors included imbalanced nutrition, improper exercise regimens, and inadequate monitoring of Achilles tendon conditions in athletes. Early treatment of musculoskeletal injuries, Achilles tendon diseases, foot deformities, and metabolic disorders is crucial. Long-term drug use and its risk assessment were also highlighted as important considerations. Furthermore, recent clinical advancements in both conventional and surgical methods to treat Achilles tendon injuries were described. The efficacy of these therapies in enhancing functional outcomes in individuals with Achilles injuries was compared. Advancements in cell-based and scaffold-based therapies aimed at enhancing cell regeneration and repairing Achilles injuries were also discussed. Discussion The combination of several established factors significantly increases the risk of Achilles tendon rupture. Addressing these factors through personalized prevention strategies can effectively reduce the incidence of these injuries. Proper nutrition, regular monitoring, timely treatment, and the correction of metabolic disorders are essential components of a comprehensive prevention plan. Conclusion Early identification of Achilles tendon risk factors allows for the timely development of effective personalized prevention strategies. These measures can contribute significantly to public health preservation by reducing the incidence of Achilles tendon ruptures associated with physical activity and sports. Continued research and clinical advancements in treatment methods will further enhance the ability to prevent and manage Achilles tendon injuries. The translational potential of this article This study identifies key modifiable and non-modifiable risk factors for Achilles tendon injuries, paving the way for personalized prevention strategies. Emphasizing nutrition, exercise, and early treatment of musculoskeletal issues, along with advancements in cell-based therapies, offers promising avenues for improving recovery and outcomes. These findings can guide clinical practices in prevention and rehabilitation, ultimately reducing Achilles injuries and enhancing public health.
Collapse
Affiliation(s)
- Maria V. Sankova
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Narasimha M. Beeraka
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA
| | - Marine V. Oganesyan
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Negoriya A. Rizaeva
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Aleksey V. Sankov
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Olga S. Shelestova
- Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill V. Bulygin
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Hemanth Vikram PR
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - A.N. Barinov
- Head of Neurology and Psychotherapy Chair of Medical Academy MEDSI Group, Moscow, Russia
| | - A.K. Khalimova
- International Medical Company “Prime Medical Group”, Almaty, Kazakhstan Asia Halimova Prime Medical Group Medical Center, Republic of Kazakhstan
| | - Y. Padmanabha Reddy
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, Karnataka, 570006, India
| | - Vladimir N. Nikolenko
- Department of Human Anatomy and Histology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
4
|
Gopinatth V, Boghosian T, Perugini JM, Smith MV, Knapik DM. Current Concepts in Orthobiologics for Achilles Tendon Injuries: A Critical Analysis Review. JBJS Rev 2024; 12:01874474-202411000-00003. [PMID: 39499787 DOI: 10.2106/jbjs.rvw.24.00144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
» Platelet-rich plasma and hyaluronic acid are low-risk and potentially high-reward treatments for Achilles tendinopathy, although clinical studies have yielded mixed results with questionable methodological quality» Case series and reports have reported that bone marrow aspirate, stem cells, and amniotic membrane products can improve functional outcomes, alleviate pain, and facilitate return to sport and activities, but high-level evidence studies are lacking» Exosomes are a promising novel biologic with laboratory studies showing improved collagen organization and cell proliferation, greater tendon mechanical properties, and prevention of extracellular matrix breakdown.» Standardization of protocols with clear reporting is necessary for future studies evaluating orthobiologic therapies for Achilles tendon injuries.
Collapse
Affiliation(s)
- Varun Gopinatth
- Saint Louis University School of Medicine, St. Louis, Missouri
| | - Tanya Boghosian
- Washington University School of Medicine, St. Louis, Missouri
| | | | - Matthew V Smith
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Derrick M Knapik
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
5
|
Strecanska M, Sekelova T, Csobonyeiova M, Danisovic L, Cehakova M. Therapeutic applications of mesenchymal/medicinal stem/signaling cells preconditioned with external factors: Are there more efficient approaches to utilize their regenerative potential? Life Sci 2024; 346:122647. [PMID: 38614298 DOI: 10.1016/j.lfs.2024.122647] [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: 02/21/2024] [Revised: 03/25/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Mesenchymal/medicinal stem/signaling cells (MSCs) have emerged as a promising treatment option for various disorders. However, the donor's age, advanced stage of disease, and prolonged in vitro expansion often diminish the innate regenerative potential of MSCs. Besides that, the absence of MSCs' comprehensive "pre-admission testing" can result in the injection of cells with reduced viability and function, which may negatively affect the overall outcome of MSC-based therapies. It is, therefore, essential to develop effective strategies to improve the impaired biological performance of MSCs. This review focuses on the comprehensive characterization of various methods of external MSCs stimulation (hypoxia, heat shock, caloric restriction, acidosis, 3D culture, and application of extracellular matrix) that augment their medicinal potential. To emphasize the significance of MSCs priming, we summarize the effects of individual and combined preconditioning approaches, highlighting their impact on MSCs' response to either physiological or pathological conditions. We further investigate the synergic action of exogenous factors to maximize MSCs' therapeutic potential. Not to omit the field of tissue engineering, the application of pretreated MSCs seeded on scaffolds is discussed as well.
Collapse
Affiliation(s)
- Magdalena Strecanska
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Tatiana Sekelova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| | - Michaela Cehakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia.
| |
Collapse
|
6
|
Menjivar NG, Oropallo J, Gebremedhn S, Souza LA, Gad A, Puttlitz CM, Tesfaye D. MicroRNA Nano-Shuttles: Engineering Extracellular Vesicles as a Cutting-Edge Biotechnology Platform for Clinical Use in Therapeutics. Biol Proced Online 2024; 26:14. [PMID: 38773366 PMCID: PMC11106895 DOI: 10.1186/s12575-024-00241-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous transporters of various active biomolecules with inflicting phenotypic capabilities, that are naturally secreted by almost all cells with a promising vantage point as a potential leading drug delivery platform. The intrinsic characteristics of their low toxicity, superior structural stability, and cargo loading capacity continue to fuel a multitude of research avenues dedicated to loading EVs with therapeutic and diagnostic cargos (pharmaceutical compounds, nucleic acids, proteins, and nanomaterials) in attempts to generate superior natural nanoscale delivery systems for clinical application in therapeutics. In addition to their well-known role in intercellular communication, EVs harbor microRNAs (miRNAs), which can alter the translational potential of receiving cells and thus act as important mediators in numerous biological and pathological processes. To leverage this potential, EVs can be structurally engineered to shuttle therapeutic miRNAs to diseased recipient cells as a potential targeted 'treatment' or 'therapy'. Herein, this review focuses on the therapeutic potential of EV-coupled miRNAs; summarizing the biogenesis, contents, and function of EVs, as well as providing both a comprehensive discussion of current EV loading techniques and an update on miRNA-engineered EVs as a next-generation platform piloting benchtop studies to propel potential clinical translation on the forefront of nanomedicine.
Collapse
Affiliation(s)
- Nico G Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jaiden Oropallo
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
- Orthopaedic Research Center (ORC), Translational Medicine Institute (TMI), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- J.R. Simplot Company, 1099 W. Front St, Boise, ID, 83702, USA
| | - Luca A Souza
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, 225 Av. Duque de Caxias Norte, Pirassununga, SP, 13635-900, Brazil
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Christian M Puttlitz
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
7
|
Farzamfar S, Garcia LM, Rahmani M, Bolduc S. Navigating the Immunological Crossroads: Mesenchymal Stem/Stromal Cells as Architects of Inflammatory Harmony in Tissue-Engineered Constructs. Bioengineering (Basel) 2024; 11:494. [PMID: 38790361 PMCID: PMC11118848 DOI: 10.3390/bioengineering11050494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic landscape of tissue engineering, the integration of tissue-engineered constructs (TECs) faces a dual challenge-initiating beneficial inflammation for regeneration while avoiding the perils of prolonged immune activation. As TECs encounter the immediate reaction of the immune system upon implantation, the unique immunomodulatory properties of mesenchymal stem/stromal cells (MSCs) emerge as key navigators. Harnessing the paracrine effects of MSCs, researchers aim to craft a localized microenvironment that not only enhances TEC integration but also holds therapeutic promise for inflammatory-driven pathologies. This review unravels the latest advancements, applications, obstacles, and future prospects surrounding the strategic alliance between MSCs and TECs, shedding light on the immunological symphony that guides the course of regenerative medicine.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Luciana Melo Garcia
- Department of Medicine, Université Laval, Québec, QC G1V 0A6, Canada;
- Hematology-Oncology Service, CHU de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Mahya Rahmani
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Stephane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
8
|
Yu YH, Lee CH, Hsu YH, Chou YC, Yu PC, Huang CT, Liu SJ. Anti-Adhesive Resorbable Indomethacin/Bupivacaine-Eluting Nanofibers for Tendon Rupture Repair: In Vitro and In Vivo Studies. Int J Mol Sci 2023; 24:16235. [PMID: 38003425 PMCID: PMC10671766 DOI: 10.3390/ijms242216235] [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: 10/20/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The treatment and surgical repair of torn Achilles tendons seldom return the wounded tendon to its original elasticity and stiffness. This study explored the in vitro and in vivo simultaneous release of indomethacin and bupivacaine from electrospun polylactide-polyglycolide composite membranes for their capacity to repair torn Achilles tendons. These membranes were fabricated by mixing polylactide-polyglycolide/indomethacin, polylactide-polyglycolide/collagen, and polylactide-polyglycolide/bupivacaine with 1,1,1,3,3,3-hexafluoro-2-propanol into sandwich-structured composites. Subsequently, the in vitro pharmaceutic release rates over 30 days were determined, and the in vivo release behavior and effectiveness of the loaded drugs were assessed using an animal surgical model. High concentrations of indomethacin and bupivacaine were released for over four weeks. The released pharmaceutics resulted in complete recovery of rat tendons, and the nanofibrous composite membranes exhibited exceptional mechanical strength. Additionally, the anti-adhesion capacity of the developed membrane was confirmed. Using the electrospinning technique developed in this study, we plan on manufacturing degradable composite membranes for tendon healing, which can deliver sustained pharmaceutical release and provide a collagenous habitat.
Collapse
Affiliation(s)
- Yi-Hsun Yu
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.)
| | - Chen-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Yung-Heng Hsu
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.)
| | - Ying-Chao Chou
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.)
| | - Ping-Chun Yu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chao-Tsai Huang
- Department of Chemical and Materials Engineering, Tamkang University, New Taipei City 25137, Taiwan;
| | - Shih-Jung Liu
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (Y.-H.Y.)
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
9
|
Tai L, Saffery NS, Chin SP, Cheong SK. Secretome profile of TNF-α-induced human umbilical cord mesenchymal stem cells unveils biological processes relevant to skin wound healing. Regen Med 2023; 18:839-856. [PMID: 37671699 DOI: 10.2217/rme-2023-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Aim: To profile and study the proteins responsible for the beneficial effect of the TNF-α-induced human umbilical cord mesenchymal stem cells (hUCMSCs) secretome in wound healing. Methods: The hUCMSCs secretome was generated with (induced) or without (uninduced) TNF-α and was subsequently analyzed by liquid chromatography-mass spectrometry, immunoassay and in vitro scratch assay. Results: Proteomic analysis revealed approximately 260 proteins, including 51 and 55 unique proteins in the induced and uninduced secretomes, respectively. Gene ontology analysis disclosed that differential proteins in the induced secretome mainly involved inflammation-related terms. The induced secretome, consisting of higher levels of FGFb, VEGF, PDGF and IL-6, significantly accelerated wound closure and enhanced MMP-13 secretion in HaCaT keratinocytes. Conclusion: The secretome from induced hUCMSCs includes factors that promote wound closure.
Collapse
Affiliation(s)
- Lihui Tai
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Nik Syazana Saffery
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Sze Piaw Chin
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Soon Keng Cheong
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
- M. Kandiah Faculty of Medicine & Health Sciences (MK FMHS), Universiti Tunku Abdul Rahman Sungai Long City Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
10
|
Wang S, Yao Z, Chen L, Li J, Chen S, Fan C. Preclinical assessment of IL-1β primed human umbilical cord mesenchymal stem cells for tendon functional repair through TGF-β/IL-10 signaling. Heliyon 2023; 9:e21411. [PMID: 37954299 PMCID: PMC10638607 DOI: 10.1016/j.heliyon.2023.e21411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
Background Inadequate repair capacity and disturbed immune compartments are the main pathological causes of tendinopathy. Transplantation of mesenchymal stem cells (MSCs) become an effective clinic option to alleviate tendinopathy. Interleukin-1β (IL-1β) could confer on MSCs enhanced immunoregulatory capability to remodel the repair microenvironment favoring tissue repair. Therefore, IL-1β activated UC-MSCs (1βUC-MSCs) may exert favorable efficacy in promoting tendon repair in a preclinical tendinopathy rat model. Methods Tendon-derived stem cells (TDSCs) were isolated and characterized. In vitro, the levels of immunoregulatory-related cytokines such as IL-1β, IL-6, IL-10, and TGF-β secreted by 1βUC-MSCs and unprimed UC-MSCs was measured. And tendon-specific markers expressed by TDSCs cultured with primed cultured medium (CM) or unprimed CM were detected. In vivo, Achilles tendinopathy was induced by 30 μL collagenase I injection in Sprague Dawley rats. One week later, the rats were randomly injected with UC-MSCs primed with IL-1β (106 cells per tendon), UC-MSCs, or PBS. After rats were sacrificed, histological evaluation, electron microscopy, biomechanical tests, gait performance were conducted to evaluate the structural and functional recovery of Achilles tendons. The inflammation and metabolic state of the extracellular matrix, and the potential mechanism were assessed by immunohistochemical staining and Western blot. Results UC-MSCs were activated by IL-1β to secrete higher levels of IL-10 and TGF-β while the secretion levels of IL-6 and IL-1β were not changed significantly, promoting a higher expression level of COL I and TNMD in TDSCs under proinflammatory environment. In vivo, the transplanted 1βUC-MSCs could survive up to 5 weeks after injection with tenogenic differentiation and improved tendon healing histologically semi-quantified by modified Bonar scores. This structural regeneration was further confirmed by observation of ultrastructural morphology, and led to good functional recovery including improved biomechanical properties and gait performance. During this process, the inflammatory response and metabolism of the extracellular matrix was improved through TGF-β/IL-10 pathway. Conclusion This study demonstrated that the transplantation of UC-MSCs activated by IL-1β exhibited satisfactory ability for promoting tendon functional repair in a tendinopathy rat model. During this process, the balance of inflammatory response and extracellular matrix metabolism was remodeled, and the TGF-β/Smad2/3 and IL-10 signaling pathways were activated simultaneously. We cautiously conclude that the IL-1β primed UC-MSCs could be a promising strategy for enhancing the ability of MSCs to treat tendinopathy.
Collapse
Affiliation(s)
- Shikun Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Zhixiao Yao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Lei Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine Tongji University, Shanghai, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Shuai Chen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| |
Collapse
|
11
|
Rehman A, Nigam A, Laino L, Russo D, Todisco C, Esposito G, Svolacchia F, Giuzio F, Desiderio V, Ferraro G. Mesenchymal Stem Cells in Soft Tissue Regenerative Medicine: A Comprehensive Review. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1449. [PMID: 37629738 PMCID: PMC10456353 DOI: 10.3390/medicina59081449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023]
Abstract
Soft tissue regeneration holds significant promise for addressing various clinical challenges, ranging from craniofacial and oral tissue defects to blood vessels, muscle, and fibrous tissue regeneration. Mesenchymal stem cells (MSCs) have emerged as a promising tool in regenerative medicine due to their unique characteristics and potential to differentiate into multiple cell lineages. This comprehensive review explores the role of MSCs in different aspects of soft tissue regeneration, including their application in craniofacial and oral soft tissue regeneration, nerve regeneration, blood vessel regeneration, muscle regeneration, and fibrous tissue regeneration. By examining the latest research findings and clinical advancements, this article aims to provide insights into the current state of MSC-based therapies in soft tissue regenerative medicine.
Collapse
Affiliation(s)
- Ayesha Rehman
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (A.R.); (A.N.)
| | - Aditya Nigam
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (A.R.); (A.N.)
| | - Luigi Laino
- Multidisciplinary Department of Medicine for Surgery and Orthodontics, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (L.L.); (D.R.); (G.F.)
| | - Diana Russo
- Multidisciplinary Department of Medicine for Surgery and Orthodontics, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (L.L.); (D.R.); (G.F.)
| | | | | | - Fabiano Svolacchia
- Departments of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00118 Rome, Italy;
| | - Federica Giuzio
- Department of Sciences, University of Basilicata, Via Nazario Sauro 85, 85100 Potenza, Italy;
- U.O.S.D. of Plastic Surgery A.O.R “San Carlo”, 85100 Potenza, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (A.R.); (A.N.)
| | - Giuseppe Ferraro
- Multidisciplinary Department of Medicine for Surgery and Orthodontics, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (L.L.); (D.R.); (G.F.)
| |
Collapse
|
12
|
Abdulmalik S, Gallo J, Nip J, Katebifar S, Arul M, Lebaschi A, Munch LN, Bartly JM, Choudhary S, Kalajzic I, Banasavadi-Siddegowdae YK, Nukavarapu SP, Kumbar SG. Nanofiber matrix formulations for the delivery of Exendin-4 for tendon regeneration: In vitro and in vivo assessment. Bioact Mater 2023; 25:42-60. [PMID: 36733930 PMCID: PMC9876843 DOI: 10.1016/j.bioactmat.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Tendon and ligament injuries are the most common musculoskeletal injuries, which not only impact the quality of life but result in a massive economic burden. Surgical interventions for tendon/ligament injuries utilize biological and/or engineered grafts to reconstruct damaged tissue, but these have limitations. Engineered matrices confer superior physicochemical properties over biological grafts but lack desirable bioactivity to promote tissue healing. While incorporating drugs can enhance bioactivity, large matrix surface areas and hydrophobicity can lead to uncontrolled burst release and/or incomplete release due to binding. To overcome these limitations, we evaluated the delivery of a peptide growth factor (exendin-4; Ex-4) using an enhanced nanofiber matrix in a tendon injury model. To overcome drug surface binding due to matrix hydrophobicity of poly(caprolactone) (PCL)-which would be expected to enhance cell-material interactions-we blended PCL and cellulose acetate (CA) and electrospun nanofiber matrices with fiber diameters ranging from 600 to 1000 nm. To avoid burst release and protect the drug, we encapsulated Ex-4 in the open lumen of halloysite nanotubes (HNTs), sealed the HNT tube endings with a polymer blend, and mixed Ex-4-loaded HNTs into the polymer mixture before electrospinning. This reduced burst release from ∼75% to ∼40%, but did not alter matrix morphology, fiber diameter, or tensile properties. We evaluated the bioactivity of the Ex-4 nanofiber formulation by culturing human mesenchymal stem cells (hMSCs) on matrix surfaces for 21 days and measuring tenogenic differentiation, compared with nanofiber matrices in basal media alone. Strikingly, we observed that Ex-4 nanofiber matrices accelerated the hMSC proliferation rate and elevated levels of sulfated glycosaminoglycan, tendon-related genes (Scx, Mkx, and Tnmd), and ECM-related genes (Col-I, Col-III, and Dcn), compared to control. We then assessed the safety and efficacy of Ex-4 nanofiber matrices in a full-thickness rat Achilles tendon defect with histology, marker expression, functional walking track analysis, and mechanical testing. Our analysis confirmed that Ex-4 nanofiber matrices enhanced tendon healing and reduced fibrocartilage formation versus nanofiber matrices alone. These findings implicate Ex-4 as a potentially valuable tool for tendon tissue engineering.
Collapse
Affiliation(s)
- Sama Abdulmalik
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Jack Gallo
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Jonathan Nip
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Sara Katebifar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Michael Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Amir Lebaschi
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Lucas N. Munch
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Jenna M. Bartly
- Department of Immunology, Center on Aging, University of Connecticut Health, Farmington, CT, USA
| | - Shilpa Choudhary
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health, Farmington, CT, USA
| | | | - Syam P. Nukavarapu
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Sangamesh G. Kumbar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
13
|
von Stade D, Meyers M, Johnson J, Schlegel TT, Romeo A, Regan D, McGilvray K. Exosome Cell Origin Affects In Vitro Markers of Tendon Repair in Ovine Macrophages and Tenocytes. Tissue Eng Part A 2023; 29:282-291. [PMID: 36792933 PMCID: PMC10178933 DOI: 10.1089/ten.tea.2022.0185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
Tendon injuries and disease are resistant to surgical repair; thus, adjunct therapies are widely investigated, especially mesenchymal stromal cells (MSCs) and, more recently, their extracellular vesicles (MSCdEVs), for example, exosomes. Thought to act on resident and infiltrating immune cells, the role of MSCdEVs in paracrine signaling is of great interest. This study investigated how MSCdEVs differ from analogs derived from resident (tenocyte) populations (TdEV). As macrophages play a significant role in tendon maintenance and repair, macrophage signaling was compared by cytokine quantification using a multiplexed immunoassay and tenocyte migration by in vitro scratch-wound analysis. TdEV-treated macrophages decreased IL-1 and increased MIP-1 and CXCL8 expression. In addition, macrophage signaling favored collagen synthesis and tenocyte bioactivity, while reducing proangiogenic signaling when TdEVs were used in place of MSCdEVs. These in vitro data demonstrate a differential influence of exosomes on macrophage signaling, according to cell source, supporting that local cell-derived exosomes may preferentially drive healing by different means with possible different outcomes compared to MSCdEVs. Impact Statement Adipose-derived mesenchymal stromal cell (AdMSC) exosomes (EVs) can improve tendon mechanical resilience, tissue organization, and M2 macrophage phenotype predominance in response to tendon injury. This active area of investigation drives great interest in the function of these exosomes as adjunct therapies for tendon disease, particularly rotator cuff tendinopathy. However, little is known about the effects of EVs as a function of cell source, nor regarding their efficacy in preclinical translational ovine models. Herein we demonstrate a differential effect of exosomes as a function of cell source, tenocyte compared to AdMSCs, on macrophage signaling and tenocyte migration of ovine cells.
Collapse
Affiliation(s)
- Devin von Stade
- Department of Mechanical Engineering, Orthopaedic Bioengineering Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | - Melinda Meyers
- Department of Clinical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | - James Johnson
- Department of Mechanical Engineering, Orthopaedic Bioengineering Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | | | - Anthony Romeo
- Shoulder Elbow Sports Medicine, Chicago, Illinois, USA
| | - Daniel Regan
- Department of Microbiology, Immunology, and Pathology, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Kirk McGilvray
- Department of Mechanical Engineering, Orthopaedic Bioengineering Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
14
|
Xu H, Zhu Y, Hsiao AWT, Xu J, Tong W, Chang L, Zhang X, Chen YF, Li J, Chen W, Zhang Y, Chan HF, Lee CW. Bioactive glass-elicited stem cell-derived extracellular vesicles regulate M2 macrophage polarization and angiogenesis to improve tendon regeneration and functional recovery. Biomaterials 2023; 294:121998. [PMID: 36641814 DOI: 10.1016/j.biomaterials.2023.121998] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/31/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Effective countermeasures for tendon injury remains unsatisfactory. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs)-based therapy via regulation of Mφ-mediated angiogenesis has emerged as a promising strategy for tissue regeneration. Still, approaches to tailor the functions of EVs to treat tendon injuries have been limited. We reported a novel strategy by applying MSC-EVs boosted with bioactive glasses (BG). BG-elicited EVs (EVB) showed up-regulation of medicinal miRNAs, including miR-199b-3p and miR-125a-5p, which play a pivotal role in M2 Mφ-mediated angiogenesis. EVB accelerated angiogenesis via the reprogrammed anti-inflammatory M2 Mφs compared with naïve MSC-EVs (EVN). In rodent Achilles tendon rupture model, EVB local administration activated anti-inflammatory responses via M2 polarization and led to a spatial correlation between M2 Mφs and newly formed blood vessels. Our results showed that EVB outperformed EVN in promoting tenogenesis and in reducing detrimental morphological changes without causing heterotopic ossification. Biomechanical test revealed that EVB significantly improved ultimate load, stiffness, and tensile modulus of the repaired tendon, along with a positive correlation between M2/M1 ratio and biomechanical properties. On the basis of the boosted nature to reprogram regenerative microenvironment, EVB holds considerable potential to be developed as a next-generation therapeutic modality for enhancing functional regeneration to achieve satisfying tendon regeneration.
Collapse
Affiliation(s)
- Hongtao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Yanlun Zhu
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Allen Wei-Ting Hsiao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Xuerao Zhang
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Jie Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Wei Chen
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Yingze Zhang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong SAR, China.
| | - Chien-Wei Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
15
|
Quintero D, Perucca Orfei C, Kaplan LD, de Girolamo L, Best TM, Kouroupis D. The roles and therapeutic potentialof mesenchymal stem/stromal cells and their extracellular vesicles in tendinopathies. Front Bioeng Biotechnol 2023; 11:1040762. [PMID: 36741745 PMCID: PMC9892947 DOI: 10.3389/fbioe.2023.1040762] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Tendinopathies encompass a highly prevalent, multi-faceted spectrum of disorders, characterized by activity-related pain, compromised function, and propensity for an extended absence from sport and the workplace. The pathophysiology of tendinopathy continues to evolve. For decades, it has been related primarily to repetitive overload trauma but more recently, the onset of tendinopathy has been attributed to the tissue's failed attempt to heal after subclinical inflammatory and immune challenges (failed healing model). Conventional tendinopathy management produces only short-term symptomatic relief and often results in incomplete repair or healing leading to compromised tendon function. For this reason, there has been increased effort to develop therapeutics to overcome the tissue's failed healing response by targeting the cellular metaplasia and pro-inflammatory extra-cellular environment. On this basis, stem cell-based therapies have been proposed as an alternative therapeutic approach designed to modify the course of the various tendon pathologies. Mesenchymal stem/stromal cells (MSCs) are multipotent stem cells often referred to as "medicinal signaling cells" due to their immunomodulatory and anti-inflammatory properties that can produce a pro-regenerative microenvironment in pathological tendons. However, the adoption of MSCs into clinical practice has been limited by FDA regulations and perceived risk of adverse events upon infusion in vivo. The introduction of cell-free approaches, such as the extracellular vesicles of MSCs, has encouraged new perspectives for the treatment of tendinopathies, showing promising short-term results. In this article, we review the most recent advances in MSC-based and MSC-derived therapies for tendinopathies. Preclinical and clinical studies are included with comment on future directions of this rapidly developing therapeutic modality, including the importance of understanding tissue loading and its relationship to any treatment regimen.
Collapse
Affiliation(s)
- Daniel Quintero
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Lee D. Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Thomas M. Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States,Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States,*Correspondence: Dimitrios Kouroupis,
| |
Collapse
|
16
|
Xu H, Zhu Y, Xu J, Tong W, Hu S, Chen Y, Deng S, Yao H, Li J, Lee C, Chan HF. Injectable bioactive glass/sodium alginate hydrogel with immunomodulatory and angiogenic properties for enhanced tendon healing. Bioeng Transl Med 2023; 8:e10345. [PMID: 36684098 PMCID: PMC9842034 DOI: 10.1002/btm2.10345] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/17/2022] [Accepted: 04/29/2022] [Indexed: 01/25/2023] Open
Abstract
Tendon healing is a complex process involving inflammation, proliferation, and remodeling, eventually achieving a state of hypocellularity and hypovascularity. Currently, few treatments can satisfactorily restore the structure and function of native tendon. Bioactive glass (BG) has been shown to possess immunomodulatory and angiogenic properties. In this study, we investigated whether an injectable hydrogel fabricated of BG and sodium alginate (SA) could be applied to enhance tenogenesis following suture repair of injured tendon. We demonstrated that BG/SA hydrogel significantly accelerated tenogenesis without inducing heterotopic ossification based on histological analysis. The therapeutic effect could attribute to increased angiogenesis and M1 to M2 phenotypic switch of macrophages within 7 days post-surgery. Morphological characterization demonstrated that BG/SA hydrogel partially reverted the pathological changes of Achilles tendon, including increased length and cross-sectional area (CSA). Finally, biomechanical test showed that BG/SA hydrogel significantly improved ultimate load, failure stress, and tensile modulus of the repaired tendon. In conclusion, administration of an injectable BG/SA hydrogel can be a novel and promising therapeutic approach to augment Achilles tendon healing in conjunction with surgical intervention.
Collapse
Affiliation(s)
- Hongtao Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
- Department of OrthopedicsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yanlun Zhu
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Shiwen Hu
- Musculoskeletal Research Laboratory, Department of Orthopedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
- School of Materials Science and EngineeringLanzhou University of TechnologyLanzhouChina
| | - Yi‐Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Ph.D. Program for Translational Science, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Master Program in Clinical Genomics and Proteomics, School of PharmacyTaipei Medical UniversityTaipeiTaiwan
| | - Shuai Deng
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Jie Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Chien‐Wei Lee
- Center for Translational Genomics ResearchChina Medical University Hospital, China Medical UniversityTaichungTaiwan
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and GeneticsThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
| |
Collapse
|
17
|
Arabiyat AS, Yeisley DJ, Güiza-Argüello VR, Qureshi F, Culibrk RA, Hahn J, Hahn MS. Effects of Stromal Cell Conditioned Medium and Antipurinergic Treatment on Macrophage Phenotype. Tissue Eng Part C Methods 2022; 28:656-671. [PMID: 36329666 PMCID: PMC9807257 DOI: 10.1089/ten.tec.2022.0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
The immunomodulatory capacity of the human mesenchymal stromal cell (MSC) secretome has been a critical driver for the development of cell-free MSC products, such as conditioned medium (CM), for regenerative medicine applications. This is particularly true as cell-free MSC products present several advantages over direct autologous or allogeneic MSC delivery with respect to safety, manufacturability, and defined potency. Recently, significant effort has been placed into creating novel MSC CM formulations with an immunomodulatory capacity tailored for specific regenerative contexts. For instance, the immunoregulatory nature of MSC CM has previously been tuned through a number of cytokine-priming strategies. Herein, we propose an alternate method to tailor the immunomodulatory "phenotype" of cytokine-primed MSC CM through coupling with the pharmacological agent, suramin. Suramin interferes with the signaling of purines including extracellular adenosine triphosphate (ATP), which plays a critical role in the activation of the innate immune system after injury. Toward this end, human THP-1-derived macrophages were activated to a proinflammatory phenotype and treated with (1) unprimed/native MSC CM, (2) interferon-γ/tumor necrosis factor α-primed MSC CM (primed CM), (3) suramin alone, or (4) primed MSC CM and suramin (primed CM/suramin). Markers of key macrophage functions-cytokine secretion, autophagy, oxidative stress modulation, and activation/migration-were assessed. Consistent with previous literature, primed CM elevated macrophage secretion of several proinflammatory and pleiotropic cytokines relative to native CM; whereas addition of suramin imparted consistent shifts in terms of TNFα (↓), interleukin-10 (↓), and hepatocyte growth factor (↑) irrespective of CM. In addition, both primed CM and suramin, individually and combined, increased reactive oxygen species production relative to native CM, and addition of suramin to primed CM shifted levels of CX3CL1, a factor involved in ATP-associated macrophage regulation. Varimax rotation assessment of the secreted cytokine profiles confirmed that primed CM/suramin resulted in a THP-1 phenotypic shift away from the lipopolysaccharide-activated proinflammatory state that was distinct from that of primed CM or native CM alone. This altered primed CM/suramin-associated phenotype may prove beneficial for healing in certain regenerative contexts. These results may inform future work coupling antipurinergic treatments with MSC-derived therapies in regenerative medicine applications.
Collapse
Affiliation(s)
- Ahmad S. Arabiyat
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Daniel J. Yeisley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Viviana R. Güiza-Argüello
- Department of Metallurgical Engineering and Materials Science, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Fatir Qureshi
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Robert A. Culibrk
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| |
Collapse
|
18
|
Koch DW, Berglund AK, Messenger KM, Gilbertie JM, Ellis IM, Schnabel LV. Interleukin-1β in tendon injury enhances reparative gene and protein expression in mesenchymal stem cells. Front Vet Sci 2022; 9:963759. [PMID: 36032300 PMCID: PMC9410625 DOI: 10.3389/fvets.2022.963759] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Tendon injury in the horse carries a high morbidity and monetary burden. Despite appropriate therapy, reinjury is estimated to occur in 50–65% of cases. Although intralesional mesenchymal stem cell (MSC) therapy has improved tissue architecture and reinjury rates, the mechanisms by which they promote repair are still being investigated. Additionally, reevaluating our application of MSCs in tendon injury is necessary given recent evidence that suggests MSCs exposed to inflammation (deemed MSC licensing) have an enhanced reparative effect. However, applying MSC therapy in this context is limited by the inadequate quantification of the temporal cytokine profile in tendon injury, which hinders our ability to administer MSCs into an environment that could potentiate their effect. Therefore, the objectives of this study were to define the temporal cytokine microenvironment in a surgically induced model of equine tendon injury using ultrafiltration probes and subsequently evaluate changes in MSC gene and protein expression following in vitro inflammatory licensing with cytokines of similar concentration as identified in vivo. In our in vivo surgically induced tendon injury model, IL-1β and IL-6 were the predominant pro-inflammatory cytokines present in tendon ultrafiltrate where a discrete peak in cytokine concentration occurred within 48 h following injury. Thereafter, MSCs were licensed in vitro with IL-1β and IL-6 at a concentration identified from the in vivo study; however, only IL-1β induced upregulation of multiple genes beneficial to tendon healing as identified by RNA-sequencing. Specifically, vascular development, ECM synthesis and remodeling, chemokine and growth factor function alteration, and immunomodulation and tissue reparative genes were significantly upregulated. A significant increase in the protein expression of IL-6, VEGF, and PGE2 was confirmed in IL-1β-licensed MSCs compared to naïve MSCs. This study improves our knowledge of the temporal tendon cytokine microenvironment following injury, which could be beneficial for the development and determining optimal timing of administration of regenerative therapies. Furthermore, these data support the need to further study the benefit of MSCs administered within the inflamed tendon microenvironment or exogenously licensed with IL-1β in vitro prior to treatment as licensed MSCs could enhance their therapeutic benefit in the healing tendon.
Collapse
Affiliation(s)
- Drew W. Koch
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Alix K. Berglund
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Kristen M. Messenger
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Jessica M. Gilbertie
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Ilene M. Ellis
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- *Correspondence: Lauren V. Schnabel
| |
Collapse
|
19
|
Ramires LC, Jeyaraman M, Muthu S, Shankar A N, Santos GS, da Fonseca LF, Lana JF, Rajendran RL, Gangadaran P, Jogalekar MP, Cardoso AA, Eickhoff A. Application of Orthobiologics in Achilles Tendinopathy: A Review. Life (Basel) 2022; 12:399. [PMID: 35330150 PMCID: PMC8954398 DOI: 10.3390/life12030399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Orthobiologics are biological materials that are intended for the regeneration of bone, cartilage, and soft tissues. In this review, we discuss the application of orthobiologics in Achilles tendinopathy, more specifically. We explain the concepts and definitions of each orthobiologic and the literature regarding its use in tendon disorders. The biological potential of these materials can be harnessed and administered into injured tissues, particularly in areas where standard healing is disrupted, a typical feature of Achilles tendinopathy. These products contain a wide variety of cell populations, cytokines, and growth factors, which have been shown to modulate many other cells at local and distal sites in the body. Collectively, they can shift the state of escalated inflammation and degeneration to reestablish tissue homeostasis. The typical features of Achilles tendinopathy are failed healing responses, persistent inflammation, and predominant catabolic reactions. Therefore, the application of orthobiologic tools represents a viable solution, considering their demonstrated efficacy, safety, and relatively easy manipulation. Perhaps a synergistic approach regarding the combination of these orthobiologics may promote more significant clinical outcomes rather than individual application. Although numerous optimistic results have been registered in the literature, additional studies and clinical trials are still highly desired to further illuminate the clinical utility and efficacy of these therapeutic strategies in the management of tendinopathies.
Collapse
Affiliation(s)
- Luciano C. Ramires
- Department of Orthopaedics and Sports Medicine, Centro Clínico Mãe de Deus, Porto Alegre 90110-270, Brazil;
| | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India;
- Department of Orthopaedics, Apollo Hospitals, Greams Road, Chennai 600006, India;
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, India
| | - Navaladi Shankar A
- Department of Orthopaedics, Apollo Hospitals, Greams Road, Chennai 600006, India;
| | - Gabriel Silva Santos
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, Brazil; (L.F.d.F.); (J.F.L.)
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, Brazil; (L.F.d.F.); (J.F.L.)
- Department of Orthopaedics, The Federal University of São Paulo, São Paulo 04024-002, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, Brazil; (L.F.d.F.); (J.F.L.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Manasi P. Jogalekar
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA;
| | - Alfredo A. Cardoso
- Department of Oncology-Integrative Medicine-Pain Care, IAC—Instituto Ana Cardoso de Práticas Integrativas e Medicina Regenerative, Gramado 95670-000, Brazil;
| | - Alex Eickhoff
- Department of Orthopaedics, Centro Ortopédico Eickhoff, Três de Maio 98910-000, Brazil;
| |
Collapse
|
20
|
Russo V, El Khatib M, Prencipe G, Citeroni MR, Faydaver M, Mauro A, Berardinelli P, Cerveró-Varona A, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Stöckl J, Barboni B. Tendon Immune Regeneration: Insights on the Synergetic Role of Stem and Immune Cells during Tendon Regeneration. Cells 2022; 11:434. [PMID: 35159244 PMCID: PMC8834336 DOI: 10.3390/cells11030434] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Tendon disorders represent a very common pathology in today's population, and tendinopathies that account 30% of tendon-related injuries, affect yearly millions of people which in turn cause huge socioeconomic and health repercussions worldwide. Inflammation plays a prominent role in the development of tendon pathologies, and advances in understanding the underlying mechanisms during the inflammatory state have provided additional insights into its potential role in tendon disorders. Different cell compartments, in combination with secreted immune modulators, have shown to control and modulate the inflammatory response during tendinopathies. Stromal compartment represented by tenocytes has shown to display an important role in orchestrating the inflammatory response during tendon injuries due to the interplay they exhibit with the immune-sensing and infiltrating compartments, which belong to resident and recruited immune cells. The use of stem cells or their derived secretomes within the regenerative medicine field might represent synergic new therapeutical approaches that can be used to tune the reaction of immune cells within the damaged tissues. To this end, promising opportunities are headed to the stimulation of macrophages polarization towards anti-inflammatory phenotype together with the recruitment of stem cells, that possess immunomodulatory properties, able to infiltrate within the damaged tissues and improve tendinopathies resolution. Indeed, the comprehension of the interactions between tenocytes or stem cells with the immune cells might considerably modulate the immune reaction solving hence the inflammatory response and preventing fibrotic tissue formation. The purpose of this review is to compare the roles of distinct cell compartments during tendon homeostasis and injury. Furthermore, the role of immune cells in this field, as well as their interactions with stem cells and tenocytes during tendon regeneration, will be discussed to gain insights into new ways for dealing with tendinopathies.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
21
|
Modulation of the Immune System Promotes Tissue Regeneration. Mol Biotechnol 2022; 64:599-610. [PMID: 35022994 DOI: 10.1007/s12033-021-00430-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/22/2021] [Indexed: 10/19/2022]
Abstract
The immune system plays an essential role in the angiogenesis, repair, and regeneration of damaged tissues. Therefore, the design of scaffolds that manipulate immune cells and factors in such a way that could accelerate the repair of damaged tissues, following implantation, is one of the main goals of regenerative medicine. However, before manipulating the immune system, the function of the various components of the immune system during the repair process should be well understood and the fabrication conditions of the manipulated scaffolds should be brought closer to the physiological state of the body. In this article, we first review the studies aimed at the role of distinct immune cell populations in angiogenesis and support of damaged tissue repair. In the second part, we discuss the use of strategies that promote tissue regeneration by modulating the immune system. Given that various studies have shown an increase in tissue repair rate with the addition of stem cells and growth factors to the scaffolds, and regarding the limited resources of stem cells, we suggest the design of scaffolds that are capable to develop repair of damaged tissue by manipulating the immune system and create an alternative for repair strategies that use stem cells or growth factors.
Collapse
|
22
|
Russo V, El Khatib M, Prencipe G, Cerveró-Varona A, Citeroni MR, Mauro A, Berardinelli P, Faydaver M, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Liverani L, Boccaccini AR, Barboni B. Scaffold-Mediated Immunoengineering as Innovative Strategy for Tendon Regeneration. Cells 2022; 11:cells11020266. [PMID: 35053383 PMCID: PMC8773518 DOI: 10.3390/cells11020266] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Tendon injuries are at the frontier of innovative approaches to public health concerns and sectoral policy objectives. Indeed, these injuries remain difficult to manage due to tendon’s poor healing ability ascribable to a hypo-cellularity and low vascularity, leading to the formation of a fibrotic tissue affecting its functionality. Tissue engineering represents a promising solution for the regeneration of damaged tendons with the aim to stimulate tissue regeneration or to produce functional implantable biomaterials. However, any technological advancement must take into consideration the role of the immune system in tissue regeneration and the potential of biomaterial scaffolds to control the immune signaling, creating a pro-regenerative environment. In this context, immunoengineering has emerged as a new discipline, developing innovative strategies for tendon injuries. It aims at designing scaffolds, in combination with engineered bioactive molecules and/or stem cells, able to modulate the interaction between the transplanted biomaterial-scaffold and the host tissue allowing a pro-regenerative immune response, therefore hindering fibrosis occurrence at the injury site and guiding tendon regeneration. Thus, this review is aimed at giving an overview on the role exerted from different tissue engineering actors in leading immunoregeneration by crosstalking with stem and immune cells to generate new paradigms in designing regenerative medicine approaches for tendon injuries.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
- Correspondence:
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Liliana Liverani
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.L.); (A.R.B.)
| | - Aldo R. Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.L.); (A.R.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
23
|
Hou J, Yang R, Vuong I, Li F, Kong J, Mao HQ. Biomaterials strategies to balance inflammation and tenogenesis for tendon repair. Acta Biomater 2021; 130:1-16. [PMID: 34082095 DOI: 10.1016/j.actbio.2021.05.043] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/15/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
Adult tendon tissue demonstrates a limited regenerative capacity, and the natural repair process leaves fibrotic scar tissue with inferior mechanical properties. Surgical treatment is insufficient to provide the mechanical, structural, and biochemical environment necessary to restore functional tissue. While numerous strategies including biodegradable scaffolds, bioactive factor delivery, and cell-based therapies have been investigated, most studies have focused exclusively on either suppressing inflammation or promoting tenogenesis, which includes tenocyte proliferation, ECM production, and tissue formation. New biomaterials-based approaches represent an opportunity to more effectively balance the two processes and improve regenerative outcomes from tendon injuries. Biomaterials applications that have been explored for tendon regeneration include formation of biodegradable scaffolds presenting topographical, mechanical, and/or immunomodulatory cues conducive to tendon repair; delivery of immunomodulatory or tenogenic biomolecules; and delivery of therapeutic cells such as tenocytes and stem cells. In this review, we provide the biological context for the challenges in tendon repair, discuss biomaterials approaches to modulate the immune and regenerative environment during the healing process, and consider the future development of comprehensive biomaterials-based strategies that can better restore the function of injured tendon. STATEMENT OF SIGNIFICANCE: Current strategies for tendon repair focus on suppressing inflammation or enhancing tenogenesis. Evidence indicates that regulated inflammation is beneficial to tendon healing and that excessive tissue remodeling can cause fibrosis. Thus, it is necessary to adopt an approach that balances the benefits of regulated inflammation and tenogenesis. By reviewing potential treatments involving biodegradable scaffolds, biological cues, and therapeutic cells, we contrast how each strategy promotes or suppresses specific repair steps to improve the healing outcome, and highlight the advantages of a comprehensive approach that facilitates the clearance of necrotic tissue and recruitment of cells during the inflammatory stage, followed by ECM synthesis and organization in the proliferative and remodeling stages with the goal of restoring function to the tendon.
Collapse
|
24
|
Theeuwes WF, van den Bosch MHJ, Thurlings RM, Blom AB, van Lent PLEM. The role of inflammation in mesenchymal stromal cell therapy in osteoarthritis, perspectives for post-traumatic osteoarthritis: a review. Rheumatology (Oxford) 2021; 60:1042-1053. [PMID: 33410465 DOI: 10.1093/rheumatology/keaa910] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
OA is a complex and highly prevalent degenerative disease affecting the whole joint, in which factors like genetic predisposition, gender, age, obesity and traumas contribute to joint destruction. ∼50-80% of OA patients develop synovitis. OA-associated risk factors contribute to joint instability and the release of cartilage matrix fragments, activating the synovium to release pro-inflammatory factors and catabolic enzymes in turn damaging the cartilage and creating a vicious circle. Currently, no cure is available for OA. Mesenchymal stromal cells (MSCs) have been tested in OA for their chondrogenic and anti-inflammatory properties. Interestingly, MSCs are most effective when administered during synovitis. This review focusses on the interplay between joint inflammation and the immunomodulation by MSCs in OA. We discuss the potential of MSCs to break the vicious circle of inflammation and describe current perspectives and challenges for clinical application of MSCs in treatment and prevention of OA, focussing on preventing post-traumatic OA.
Collapse
Affiliation(s)
- Wessel F Theeuwes
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Rogier M Thurlings
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
25
|
Muzzio N, Moya S, Romero G. Multifunctional Scaffolds and Synergistic Strategies in Tissue Engineering and Regenerative Medicine. Pharmaceutics 2021; 13:792. [PMID: 34073311 PMCID: PMC8230126 DOI: 10.3390/pharmaceutics13060792] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
The increasing demand for organ replacements in a growing world with an aging population as well as the loss of tissues and organs due to congenital defects, trauma and diseases has resulted in rapidly evolving new approaches for tissue engineering and regenerative medicine (TERM). The extracellular matrix (ECM) is a crucial component in tissues and organs that surrounds and acts as a physical environment for cells. Thus, ECM has become a model guide for the design and fabrication of scaffolds and biomaterials in TERM. However, the fabrication of a tissue/organ replacement or its regeneration is a very complex process and often requires the combination of several strategies such as the development of scaffolds with multiple functionalities and the simultaneous delivery of growth factors, biochemical signals, cells, genes, immunomodulatory agents, and external stimuli. Although the development of multifunctional scaffolds and biomaterials is one of the most studied approaches for TERM, all these strategies can be combined among them to develop novel synergistic approaches for tissue regeneration. In this review we discuss recent advances in which multifunctional scaffolds alone or combined with other strategies have been employed for TERM purposes.
Collapse
Affiliation(s)
- Nicolas Muzzio
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA;
| | - Sergio Moya
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramon 182 C, 20014 Donostia-San Sebastian, Spain;
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznan, Poland
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA;
| |
Collapse
|
26
|
Utsunomiya T, Zhang N, Lin T, Kohno Y, Ueno M, Maruyama M, Rhee C, Huang E, Yao Z, Goodman SB. Different Effects of Intramedullary Injection of Mesenchymal Stem Cells During the Acute vs. Chronic Inflammatory Phase on Bone Healing in the Murine Continuous Polyethylene Particle Infusion Model. Front Cell Dev Biol 2021; 9:631063. [PMID: 33816480 PMCID: PMC8017138 DOI: 10.3389/fcell.2021.631063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic inflammation is a common feature in many diseases of different organ systems, including bone. However, there are few interventions to mitigate chronic inflammation and preserve host tissue. Previous in vitro studies demonstrated that preconditioning of mesenchymal stem cells (pMSCs) using lipopolysaccharide and tumor necrosis factor-α polarized macrophages from a pro-inflammatory to an anti-inflammatory phenotype and increased osteogenesis compared to unaltered MSCs. In the current study, we investigated the local injection of MSCs or pMSCs during the acute versus chronic inflammatory phase in a murine model of inflammation of bone: the continuous femoral intramedullary polyethylene particle infusion model. Chronic inflammation due to contaminated polyethylene particles decreased bone mineral density and increased osteoclast-like cells positively stained with leukocyte tartrate resistant acid phosphatase (TRAP) staining, and resulted in a sustained M1 pro-inflammatory macrophage phenotype and a decreased M2 anti-inflammatory phenotype. Local injection of MSCs or pMSCs during the chronic inflammatory phase reversed these findings. Conversely, immediate local injection of pMSCs during the acute inflammatory phase impaired bone healing, probably by mitigating the mandatory acute inflammatory reaction. These results suggest that the timing of interventions to facilitate bone healing by modulating inflammation is critical to the outcome. Interventions to facilitate bone healing by modulating acute inflammation should be prudently applied, as this phase of bone healing is temporally sensitive. Alternatively, local injection of MSCs or pMSCs during the chronic inflammatory phase may be a potential intervention to mitigate the adverse effects of contaminated particles on bone.
Collapse
Affiliation(s)
- Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
27
|
Xu H, Lee CW, Wang YF, Huang S, Shin LY, Wang YH, Wan Z, Zhu X, Yung PSH, Lee OKS. The Role of Paracrine Regulation of Mesenchymal Stem Cells in the Crosstalk With Macrophages in Musculoskeletal Diseases: A Systematic Review. Front Bioeng Biotechnol 2020; 8:587052. [PMID: 33324622 PMCID: PMC7726268 DOI: 10.3389/fbioe.2020.587052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/28/2020] [Indexed: 12/27/2022] Open
Abstract
The phenotypic change of macrophages (Mφs) plays a crucial role in the musculoskeletal homeostasis and repair process. Although mesenchymal stem cells (MSCs) have been shown as a novel approach in tissue regeneration, the therapeutic potential of MSCs mediated by the interaction between MSC-derived paracrine mediators and Mφs remains elusive. This review focused on the elucidation of paracrine crosstalk between MSCs and Mφs during musculoskeletal diseases and injury. The search method was based on the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) and Cochrane Guidelines. The search strategies included MeSH terms and other related terms of MSC-derived mediators and Mφs. Ten studies formed the basis of this review. The current finding suggested that MSC administration promoted proliferation and activation of CD163+ or CD206+ M2 Mφs in parallel with reduction of proinflammatory cytokines and increase in anti-inflammatory cytokines. During such period, Mφs also induced MSCs into a motile and active phenotype via the influence of proinflammatory cytokines. Such crosstalk between Mφs and MSCs further strengthens the effect of paracrine mediators from MSCs to regulate Mφs phenotypic alteration. In conclusion, MSCs in musculoskeletal system, mediated by the interaction between MSC paracrine and Mφs, have therapeutic potential in musculoskeletal diseases.
Collapse
Affiliation(s)
- Hongtao Xu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Chien-Wei Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Developmental and Regenerative Biology TRP, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu-Fan Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Lih-Ying Shin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu-Hsuan Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Zihao Wan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaobo Zhu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Patrick Shu Hang Yung
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Oscar Kuang-Sheng Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Department of Orthopadics, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
28
|
Li Y, Zhu T, Wang L, Jiang J, Xie G, Huangfu X, Dong S, Zhao J. Tissue-Engineered Decellularized Allografts for Anterior Cruciate Ligament Reconstruction. ACS Biomater Sci Eng 2020; 6:5700-5710. [PMID: 33320573 DOI: 10.1021/acsbiomaterials.0c00269] [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] [Indexed: 12/19/2022]
Abstract
Anterior cruciate ligament (ACL) reconstruction with allografts is limited by high immunogenicity, poor cellularization, and delayed tendon-bone healing. Decellularized tendons (DAs) have been used as bioscaffolds to reconstruct ligaments with variable success. In the study, four kinds of decellularized allogeneic hamstring tendons were prepared and their microstructure and cytocompatibility were examined in vitro. The results showed that decellularized allografts neutralized by 5% calcium bicarbonate had typical reticular and porous microstructures with optical cytocompatibility. Tissue-engineering decellularized allografts (TEDAs) were prepared with the selected decellularized allografts and tendon stem/progenitor cells and used for ACL reconstruction in a rabbit model. Histological staining showed that the TEDAs promoted cellular infiltration and new vessel formation significantly and improved tendon-bone healing moderately compared to decellularized allografts. Better macroscopic scores and biomechanical results were observed in TEDA groups, but there were no significant differences between DA and TEDA groups at months 1, 2, and 3 postoperatively. Immunohistochemical data showed that the tissue-engineering decellularized allografts enhanced the expression of collagen I at each timepoint and collagen III at months 1 and 2. ELISA analysis showed that the tissue-engineering decellularized allografts reduced the secretion of IgE and IL-1β within 1 month and promoted the secretion of IL-2, IL-4, IL-10, and IL-17 after 1 month. The results showed that tissue-engineering decellularized allografts strengthened intra-articular graft remodeling significantly and provided moderate improvements in tendon-bone healing by creating more suitable immune responses than decellularized allografts. The study revealed that tissue-engineering decellularized allografts as a promising option for ACL reconstruction could achieve more favorable outcomes.
Collapse
Affiliation(s)
- Yamin Li
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Tonghe Zhu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Liren Wang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jia Jiang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Guoming Xie
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xiaoqiao Huangfu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shikui Dong
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
29
|
Chisari E, Rehak L, Khan WS, Maffulli N. The role of the immune system in tendon healing: a systematic review. Br Med Bull 2020; 133:49-64. [PMID: 32163543 DOI: 10.1093/bmb/ldz040] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The role of the immune system in tendon healing relies on polymorphonucleocytes, mast cells, macrophages and lymphocytes, the 'immune cells' and their cytokine production. This systematic review reports how the immune system affects tendon healing. SOURCES OF DATA We registered our protocol (registration number: CRD42019141838). After searching PubMed, Embase and Cochrane Library databases, we included studies of any level of evidence published in peer-reviewed journals reporting clinical or preclinical results. The PRISMA guidelines were applied, and risk of bias and the methodological quality of the included studies were assessed. We excluded all the articles with high risk of bias and/or low quality after the assessment. We included 62 articles assessed as medium or high quality. AREAS OF AGREEMENT Macrophages are major actors in the promotion of proper wound healing as well as the resolution of inflammation in response to pathogenic challenge or tissue damage. The immune cells secrete cytokines involving both pro-inflammatory and anti-inflammatory factors which could affect both healing and macrophage polarization. AREAS OF CONTROVERSY The role of lymphocytes, mast cells and polymorphonucleocytes is still inconclusive. GROWING POINTS The immune system is a major actor in the complex mechanism behind the healing response occurring in tendons after an injury. A dysregulation of the immune response can ultimately lead to a failed healing response. AREAS TIMELY FOR DEVELOPING RESEARCH Further studies are needed to shed light on therapeutic targets to improve tendon healing and in managing new way to balance immune response.
Collapse
Affiliation(s)
- Emanuele Chisari
- University of Catania, Department of General Surgery and Medical Specialities, Via Santa Sofia 78, Catania 95123, Italy
| | - Laura Rehak
- Athena Biomedical innovations, Viale Europa 139, Florence, 50126, Italy
| | - Wasim S Khan
- Division of Trauma & Orthopaedics, Addenbrooke's Hospital, University of Cambridge, Hills Rd, Cambridge CB2 0QQ, UK
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Via Salvator Allende 23, Baronissi, 89100 Salerno, Italy.,Clinica Ortopedica, Ospedale San Giovanni di Dio e Ruggi D'Aragona, Largo Città di Ippocrate, Salerno, 84131 Italy.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, London E1 4DG, UK.,School of Pharmacy and Bioengineering, Keele University of School of Medicine, Guy Hilton Research Centre, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK
| |
Collapse
|
30
|
Shi Y, Kang X, Wang Y, Bian X, He G, Zhou M, Tang K. Exosomes Derived from Bone Marrow Stromal Cells (BMSCs) Enhance Tendon-Bone Healing by Regulating Macrophage Polarization. Med Sci Monit 2020; 26:e923328. [PMID: 32369458 PMCID: PMC7218969 DOI: 10.12659/msm.923328] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Inflammation after tendon-bone junction injury results in the formation of excessive scar tissue and poor biomechanical properties. Recent research has shown that exosomes derived from bone marrow stromal cells (BMSCs) can modulate inflammation during tissue healing. Thus, our study aimed to enhance tendon-bone healing by use of BMSC-derived exosomes (BMSC-Exos). Material/Methods The mouse tendon-bone reconstruction model was established, and the mice were randomly divided into 3 groups: the control group, the hydrogel group, and the hydrogel+exosome group, with 30 mice in each group. At 7 days, 14 days, and 1 month after surgery, tendon-bone junction samples were harvested, and the macrophage polarization and tendon-bone healing were evaluated based on histology, immunofluorescence, and quantitative RT-PCR (qRT-PCR) analysis. Results In the early phase, we observed significantly higher numbers of M2 macrophages and more anti-inflammatory and chondrogenic-related factors in the hydrogel+BMSC-Exos group compared with the control group and the hydrogel group. The M1 macrophages and related proinflammatory factors decreased. Cell apoptosis decreased in the hydrogel+BMSC-Exos group, while cell proliferation increased; in particular, the CD146+ stem cells substantially increased. At 1 month after surgery, there was more fibrocartilage in the hydrogel+BMSC-Exos group than in the other groups. Biomechanical testing showed that the maximum force, strength, and elastic modulus were significantly improved in the hydrogel+BMSC-Exos group. Conclusions Our study provides evidence that the local administration of BMSC-Exos promotes the formation of fibrocartilage by increasing M2 macrophage polarization in tendon-to-bone healing, leading to improved biomechanical properties. These findings provide a basis for the potential clinical use of BMSC-Exos in tendon-bone repair.
Collapse
Affiliation(s)
- Youxing Shi
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| | - Xia Kang
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| | - Yunjiao Wang
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| | - Xuting Bian
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| | - Gang He
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| | - Mei Zhou
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| | - Kanglai Tang
- Sports Medicine Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China (mainland)
| |
Collapse
|
31
|
Seetharaman R, Mahmood A, Kshatriya P, Patel D, Srivastava A. An Overview on Stem Cells in Tissue Regeneration. Curr Pharm Des 2020; 25:2086-2098. [PMID: 31298159 DOI: 10.2174/1381612825666190705211705] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Deteriorations in tissues and decline in organ functions, due to chronic diseases or with advancing age or sometimes due to infections or injuries, can severely compromise the quality of life of an individual. Regenerative medicine, a field of medical research focuses on replacing non-functional or dead cells or repairing or regenerating tissues and organs to restore normal functions of an impaired organ. Approaches used in regenerative therapy for achieving the objective employ a number of means which include soluble biomolecules, stem cell transplants, tissue engineering, gene therapy and reprogramming of cells according to target tissue types. Stem cells transplant and tissue regeneration methods for treating various diseases have rapidly grown in usage over the past decades or so. There are different types of stem cells such as mesenchymal, hematopoietic, embryonic, mammary, intestinal, endothelial, neural, olfactory, neural crest, testicular and induced pluripotent stem cells. METHODS This review covers the recent advances in tissue regeneration and highlights the application of stem cell transplants in treating many life-threatening diseases or in improving quality of life. RESULTS Remarkable progress in stem cell research has established that the cell-based therapy could be an option for treating diseases which could not be cured by conventional medical means till recent. Stem cells play major roles in regenerative medicine with its exceptional characteristics of self-renewal capacity and potential to differentiate into almost all types of cells of a body. CONCLUSION Vast number of reports on preclinical and clinical application of stem cells revealed its vital role in disease management and many pharmacological industries around the globe working to achieve effective stem cell based products.
Collapse
Affiliation(s)
| | | | | | | | - Anand Srivastava
- Global Institute of Stem Cell Therapy and Research, 4660 La Jolla Village Drive, San Diego, CA 92122, United States
| |
Collapse
|
32
|
Copper Does Not Induce Tenogenic Differentiation but Promotes Migration and Increases Lysyl Oxidase Activity in Adipose-Derived Mesenchymal Stromal Cells. Stem Cells Int 2020; 2020:9123281. [PMID: 32148523 PMCID: PMC7053469 DOI: 10.1155/2020/9123281] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 01/08/2023] Open
Abstract
Background Copper belongs to the essential trace metals that play a key role in the course of cellular processes maintaining the whole body's homeostasis. As there is a growing interest in transplanting mesenchymal stromal cells (MSCs) into the site of injury to improve the regeneration of damaged tendons, the purpose of the study was to verify whether copper supplementation may have a positive effect on the properties of human adipose tissue-derived MSCs (hASCs) which potentially can contribute to improvement of tendon healing. Results Cellular respiration of hASCs decreased with increasing cupric sulfate concentrations after 5 days of incubation. The treatment with CuSO4 did not positively affect the expression of genes associated with tenogenesis (COL1α1, COL3α1, MKX, and SCX). However, the level of COL1α1 protein, whose transcript was decreased in comparison to a control, was elevated after a 5-day exposition to 25 μM CuSO4. The content of the MKX and SCX protein in hASCs exposed to cupric sulfate was reduced compared to that of untreated control cells, and the level of the COL3α1 protein, whose transcript was decreased in comparison to a control, was elevated after a 5-day exposition to 25 μM CuSO4. The content of the MKX and SCX protein in hASCs exposed to cupric sulfate was reduced compared to that of untreated control cells, and the level of the COL3. Conclusion Copper sulfate supplementation can have a beneficial effect on tendon regeneration not by inducing tenogenic differentiation, but by improving the recruitment of MSCs to the site of injury, where they can secrete growth factors, cytokines and chemokines, and prevent the effects of oxidative stress at the site of inflammation, as well as improve the stabilization of collagen fibers, thereby accelerating the process of tendon healing.
Collapse
|
33
|
Influence of inflammatory conditions provided by macrophages on osteogenic ability of mesenchymal stem cells. Stem Cell Res Ther 2020; 11:57. [PMID: 32054534 PMCID: PMC7020593 DOI: 10.1186/s13287-020-1578-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/04/2019] [Accepted: 02/03/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The mechanisms by which macrophage phenotype contributes to mesenchymal stem cells (MSC)-mediated bone repair remain unclear. In this work, we investigated the influence of factors released by human macrophages polarized to a pro-inflammatory or an anti-inflammatory phenotype on the ability of human MSC to attach, migrate, and differentiate toward the osteoblastic lineage. We focused on the role of TNF-α and IL-10, key pro-inflammatory and anti-inflammatory cytokines, respectively, in regulating MSC functions. METHODS MSC were treated with media conditioned by pro-inflammatory or anti-inflammatory macrophages to study their influence in cell attachment, migration, and osteogenic differentiation. The involvement of TNF-α and IL-10 in the regulation of MSC functions was investigated using neutralizing antibodies and recombinant cytokines. RESULTS Treatment of MSC with media conditioned by pro-inflammatory or anti-inflammatory macrophages promoted cell elongation and enhanced MSC ability to attach and migrate. These effects were more noticeable when MSC were treated with media from pro-inflammatory macrophages. Interestingly, MSC osteogenic activity was enhanced by factors released by anti-inflammatory macrophages, but not by pro-inflammatory macrophages. Significant IL-10 levels originated from anti-inflammatory macrophages enhanced MSC osteogenesis by increasing ALP activity and mineralization in MSC layers cultured under osteogenic conditions. Moreover, macrophage-derived IL-10 regulated the expression of the osteogenic markers RUNX2, COL1A1, and ALPL. Notably, low TNF-α levels secreted by anti-inflammatory macrophages increased ALP activity in differentiating MSC whereas high TNF-α levels produced by pro-inflammatory macrophages had no effects on osteogenesis. Experiments in which MSC were treated with cytokines revealed that IL-10 was more effective in promoting matrix maturation and mineralization than TNF-α. CONCLUSIONS Factors secreted by pro-inflammatory macrophages substantially increased MSC attachment and migration whereas those released by anti-inflammatory macrophages enhanced MSC osteogenic activity as well as cell migration. IL-10 was identified as an important cytokine secreted by anti-inflammatory macrophages that potentiates MSC osteogenesis. Our findings provide novel insights into how environments provided by macrophages regulate MSC osteogenesis, which may be helpful to develop strategies to enhance bone regeneration.
Collapse
|
34
|
Leong NL, Kator JL, Clemens TL, James A, Enamoto-Iwamoto M, Jiang J. Tendon and Ligament Healing and Current Approaches to Tendon and Ligament Regeneration. J Orthop Res 2020; 38:7-12. [PMID: 31529731 PMCID: PMC7307866 DOI: 10.1002/jor.24475] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 09/10/2019] [Indexed: 02/04/2023]
Abstract
Ligament and tendon injuries are common problems in orthopedics. There is a need for treatments that can expedite nonoperative healing or improve the efficacy of surgical repair or reconstruction of ligaments and tendons. Successful biologically-based attempts at repair and reconstruction would require a thorough understanding of normal tendon and ligament healing. The inflammatory, proliferative, and remodeling phases, and the cells involved in tendon and ligament healing will be reviewed. Then, current research efforts focusing on biologically-based treatments of ligament and tendon injuries will be summarized, with a focus on stem cells endogenous to tendons and ligaments. Statement of clinical significance: This paper details mechanisms of ligament and tendon healing, as well as attempts to apply stem cells to ligament and tendon healing. Understanding of these topics could lead to more efficacious therapies to treat ligament and tendon injuries. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:7-12, 2020.
Collapse
Affiliation(s)
- Natalie L Leong
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
- Department of Surgery, Baltimore VA Medical Center, Baltimore, Maryland
| | - Jamie L Kator
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Aaron James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Motomi Enamoto-Iwamoto
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
| | - Jie Jiang
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
| |
Collapse
|
35
|
Shen H, Yoneda S, Abu-Amer Y, Guilak F, Gelberman RH. Stem cell-derived extracellular vesicles attenuate the early inflammatory response after tendon injury and repair. J Orthop Res 2020; 38:117-127. [PMID: 31286564 PMCID: PMC6917960 DOI: 10.1002/jor.24406] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/02/2019] [Indexed: 02/04/2023]
Abstract
Adipose-derived stem cells (ASCs) have the potential to enhance tendon repair via paracrine regulation of the inflammatory response to injury. Extracellular vesicles (EVs), which are secreted by ASCs, have shown promise in mediating this process. This study was designed to evaluate the effect of ASC EVs on early tendon healing using a mouse Achilles tendon injury and repair model. EVs were isolated from the conditioned medium of naïve and interferonγ-primed ASCs and applied to the repair site via a collagen sheet. Tendon healing was assessed in nuclear factor-κB (NF-κB)-luciferase reporter mice up to 7 days after suture repair. As anticipated, repair site NF-κB activity increased greater than twofold following tendon repair. Treatment with EVs from primed but not naïve ASCs effectively suppressed the response. Accordingly, the pro-inflammatory genes Il1b and Ifng were both dramatically increased in repaired tendons, while primed, but not naïve ASC EVs attenuated the response. Compared with control repairs, primed ASC EVs further reduced the rate of post-repair tendon gap formation and rupture and facilitated collagen formation at the injury site. Additional experiments demonstrated that EVs target macrophages and that primed ASC EVs were most effective in blocking macrophage NF-κB activity. Collectively, the findings of this study demonstrate that primed ASC EVs, similar to ASCs, attenuate the early tendon inflammatory response after injury via modulation of the macrophage inflammatory response. Statement of clinical significance: These findings introduce a new cell-free therapy, derived from stem cells, for tendon repair with the potential for improved therapeutic efficacy and safety. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:117-127, 2020.
Collapse
Affiliation(s)
- Hua Shen
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children – St. Louis, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children – St. Louis, St. Louis, MO, USA
| | - Richard H. Gelberman
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
36
|
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]
|
37
|
Ruan D, Zhu T, Huang J, Le H, Hu Y, Zheng Z, Tang C, Chen Y, Ran J, Chen X, Yin Z, Qian S, Pioletti D, Heng BC, Chen W, Shen W, Ouyang HW. Knitted Silk-Collagen Scaffold Incorporated with Ligament Stem/Progenitor Cells Sheet for Anterior Cruciate Ligament Reconstruction and Osteoarthritis Prevention. ACS Biomater Sci Eng 2019; 5:5412-5421. [PMID: 33464061 DOI: 10.1021/acsbiomaterials.9b01041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Current surgical management of anterior cruciate ligament (ACL) rupture still remains an intractable challenge in ACL regeneration due to the weak self-healing capability of ACL. Inadequate cell numbers and vascularization within the articular cavity contribute mainly to the poor prognosis. This time, we fabricated a new tissue engineering scaffold by adding ligament stem/progenitor cell (LSPC) sheets to our previous knitted silk-collagen sponge scaffold, which overcame these limitations by providing sufficient numbers of seed cells and a natural extracellular matrix to facilitate regeneration. LSPCs display excellent proliferation and multilineage differentiation capacity. Upon ectopic implantation, the knitted silk-collagen sponge scaffold incorporated with an LSPC sheet exhibited less immune cells but more fibroblast-like cells, deposited ECM and neovascularization, and better tissue ingrowth. In a rabbit model, we excised the ACL and performed a reconstructive surgery with our scaffold. Increased expression of ligament-specific genes and better collagen fibril formation could be observed after orthotopic transplantation. After 6 months, the LSPC sheet group showed better results on ligament regeneration and ligament-bone healing. Furthermore, no obvious cartilage and meniscus degeneration were observed at 6 months postoperation. In conclusion, these results indicated that the new tissue engineering scaffold can promote ACL regeneration and slow down the progression of osteoarthritis, thus suggesting its high clinical potential as an ideal graft in ACL reconstruction.
Collapse
Affiliation(s)
- Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China
| | - Ting Zhu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Department of Cardiothoracic Surgery, Shaoxing People's Hospital, Shaoxin Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| | - Jiayun Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China
| | - Huihui Le
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China
| | - Yejun Hu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China
| | - Zefeng Zheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Department of Orthopedic Surgery, Children's Hospital, Zhejiang University School of Medicine, Zhejiang, 310052, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China
| | - Yangwu Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China
| | - Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China
| | - Xiao Chen
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Zi Yin
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China
| | - Shengjun Qian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China
| | | | | | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Department of Orthopedics, Research Institute of Zhejiang University, Zhejiang, 310027, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China.,Laboratory of Biomechanical Orthopedics, EPFL, Lausanne, Switzerland.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Hong-Wei Ouyang
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang, 310009, China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| |
Collapse
|
38
|
Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal Stem Cells for Regenerative Medicine. Cells 2019; 8:E886. [PMID: 31412678 PMCID: PMC6721852 DOI: 10.3390/cells8080886] [Citation(s) in RCA: 715] [Impact Index Per Article: 119.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
In recent decades, the biomedical applications of mesenchymal stem cells (MSCs) have attracted increasing attention. MSCs are easily extracted from the bone marrow, fat, and synovium, and differentiate into various cell lineages according to the requirements of specific biomedical applications. As MSCs do not express significant histocompatibility complexes and immune stimulating molecules, they are not detected by immune surveillance and do not lead to graft rejection after transplantation. These properties make them competent biomedical candidates, especially in tissue engineering. We present a brief overview of MSC extraction methods and subsequent potential for differentiation, and a comprehensive overview of their preclinical and clinical applications in regenerative medicine, and discuss future challenges.
Collapse
Affiliation(s)
- Yu Han
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Xuezhou Li
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Yanbo Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China.
| | - Yuping Han
- Department of Urology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China.
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
39
|
Nguyen TL, Choi Y, Kim J. Mesoporous Silica as a Versatile Platform for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803953. [PMID: 30417454 DOI: 10.1002/adma.201803953] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/07/2018] [Indexed: 04/14/2023]
Abstract
Immunotherapy has been recognized for decades as a promising therapeutic method for cancer treatment. To enhance host immune responses against cancer, antigen-presenting cells (APCs; e.g., dendritic cells) or T cells are educated using immunomodulatory agents including tumor-associated antigens and adjuvants, and manipulated to induce a cascading adaptive immune response targeting tumor cells. Mesoporous silica materials are promising candidates to improve cancer immunotherapy based on their attractive properties that include high porosity, high biocompatibility, facile surface modification, and self-adjuvanticity. Here, the recent progress on mesoporous-silica-based immunotherapies based on two material forms is summarized: 1) mesoporous silica nanoparticles (MSNs), which can be internalized into APCs, and 2) micrometer-sized mesoporous silica rods (MSRs) that can form a 3D space to recruit APCs. Subcutaneously injected MSN-based cancer vaccines can be taken up by peripheral APCs or by APCs in lymphoid organs to educate the immune system against cancer cells. MSR cancer vaccines can recruit immune cells into the MSR scaffold to induce cancer-specific immunity. Both vaccine systems successfully stimulate the adaptive immune response to eradicate cancer in vivo. Thus, mesoporous silica has potential value as a material platform for the treatment of cancer or infectious diseases.
Collapse
Affiliation(s)
- Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Youngjin Choi
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| |
Collapse
|
40
|
Wu Y, Mu Y, Yin L, Wang Z, Liu W, Wan H. Complications in the Management of Acute Achilles Tendon Rupture: A Systematic Review and Network Meta-analysis of 2060 Patients. Am J Sports Med 2019; 47:2251-2260. [PMID: 30781966 DOI: 10.1177/0363546518824601] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Acute Achilles tendon rupture (ATR) has increased in the past decade, and many new treatments and rehabilitation regimens have been introduced. But major complications in ATR management remain an unsolved problem. PURPOSE To compare the risk of major complications of acute ATR after different combinations of treatments and rehabilitation regimens. STUDY DESIGN Systematic review and network meta-analysis. METHOD The authors searched 4 databases (PubMed, Medline, Embase, and the Cochrane Library) from the date of inception until February 2018 for articles in English. The authors considered randomized controlled trials comparing interventions and rehabilitation protocols for acute ATR and restricted (1) interventions to nonoperative treatment, minimally invasive surgery, and open surgery and (2) rehabilitation protocols to accelerated rehabilitation and early immobilization. Major complications were assessed-namely, rerupture, deep infection, and deep vein thrombosis (DVT). Only patients with primary acute ATR were considered. Quality assessment was performed with the Cochrane "risk of bias" tool. A series of additional tests were conducted to ensure the validity of the results. RESULTS Twenty-nine randomized controlled trials with 2060 patients were included in this Bayesian network meta-analysis. The mean incidence of overall major complications from all managements was 9.13% (median, 6.67%). The mean incidence rates of rerupture, deep infection, and DVT from all managements were 5%, 1.50%, and 2.67%, respectively. According to relative risk, nonoperative treatment combined with early immobilization was significantly associated with a higher risk of major complications. According to the surface under the cumulative ranking curve, minimally invasive surgery with accelerated rehabilitation had the highest possibility (79.7%) of being the best management with regard to minimizing major complications. CONCLUSION For treating acute ATR, management combining minimally invasive surgery with accelerated rehabilitation had the highest possibility of being superior in terms of major complication risks, according to the surface under the cumulative ranking curve. Management combining nonoperative treatment with early immobilization was statistically associated with a higher risk of complications as compared with the other methods of management.
Collapse
Affiliation(s)
- Yahong Wu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yuan Mu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Liangjun Yin
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Zhuoqun Wang
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Wenke Liu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Haimin Wan
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
41
|
Regmi S, Pathak S, Kim JO, Yong CS, Jeong JH. Mesenchymal stem cell therapy for the treatment of inflammatory diseases: Challenges, opportunities, and future perspectives. Eur J Cell Biol 2019; 98:151041. [PMID: 31023504 DOI: 10.1016/j.ejcb.2019.04.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising alternative agents for the treatment of inflammatory disorders due to their immunomodulatory functions, and several clinical trials on MSC-based products are currently being conducted. In this review, we discuss recent progress made on the use of MSCs as immunomodulatory agents, developmental challenges posed by MSC-based therapy, and the strategies being used to overcome these challenges. In this context, current understanding of the mechanisms responsible for MSC interactions with the immune system and the molecular responses of MSCs to inflammatory signals are discussed. The immunosuppressive activities of MSCs are initiated by cell-to-cell contact and the release of immuno-regulatory molecules. By doing so, MSCs can inhibit the proliferation and function of T cells, natural killer cells, B cells, and dendritic cells, and can also increase the proliferation of regulatory T cells. However, various problems, such as low transplanted cell viability, poor homing and engraftment into injured tissues, MSC heterogeneity, and lack of adequate information on optimum MSC doses impede clinical applications. On the other hand, it has been shown that the immunomodulatory activities and viabilities of MSCs might be enhanced by 3D-cultured systems, genetic modifications, preconditioning, and targeted-delivery.
Collapse
Affiliation(s)
- Shobha Regmi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Shiva Pathak
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
42
|
Aynardi M, Zahoor T, Mitchell R, Loube J, Feltham T, Manandhar L, Paudel S, Schon L, Zhang Z. Orthotopic Transplantation of Achilles Tendon Allograft in Rats: With or without Incorporation of Autologous Mesenchymal Stem Cells. Cell Transplant 2019; 27:245-255. [PMID: 29637821 PMCID: PMC5898692 DOI: 10.1177/0963689717745891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The biology and function of orthotopic transplantation of Achilles tendon allograft are unknown. Particularly, the revitalization of Achilles allograft is a clinical concern. Achilles allografts were harvested from donor rats and stored at -80 °C. Subcutaneous adipose tissue was harvested from the would-be allograft recipient rats for isolation of mesenchymal stem cells (MSCs). MSCs were cultured with growth differentiation factor-5 (GDF-5) and applied onto Achilles allografts on the day of transplantation. After the native Achilles tendon was resected from the left hind limb of the rats, Achilles allograft, with or without autologous MSCs, was implanted and sutured with calf muscles proximally and calcaneus distally. Animal gait was recorded presurgery and postsurgery weekly. The animals were sacrificed at week 4, and the transplanted Achilles allografts were collected for biomechanical testing and histology. The operated limbs had altered gait. By week 4, the paw print intensity, stance time, and duty cycle (percentage of the stance phase in a step cycle) of the reconstructed limbs were mostly recovered to the baselines recorded before surgery. Maximum load of failure was not different between Achilles allografts, with or without MSCs, and the native tendons. The Achilles allograft supplemented with MSCs had higher cellularity than the Achilles allograft without MSCs. Deposition of fine collagen (type III) fibers was active in Achilles allograft, with or without MSCs, but it was more evenly distributed in the allografts that were incubated with MSCs. In conclusion, orthotopically transplanted Achilles allograft healed with host tissues, regained strength, and largely restored Achilles function in 4 wk in rats. It is therefore a viable option for the reconstruction of a large Achilles tendon defect. Supplementation of MSCs improved repopulation of Achilles allograft, but large animal models, with long-term follow up and cell tracking, may be required to fully appreciate the functional benefits of MSCs.
Collapse
Affiliation(s)
| | | | - Reed Mitchell
- 3 MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Jeffrey Loube
- 3 MedStar Union Memorial Hospital, Baltimore, MD, USA
| | | | | | | | - Lew Schon
- 3 MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Zijun Zhang
- 3 MedStar Union Memorial Hospital, Baltimore, MD, USA
| |
Collapse
|
43
|
Chamberlain CS, Clements AEB, Kink JA, Choi U, Baer GS, Halanski MA, Hematti P, Vanderby R. Extracellular Vesicle-Educated Macrophages Promote Early Achilles Tendon Healing. Stem Cells 2019; 37:652-662. [PMID: 30720911 PMCID: PMC6850358 DOI: 10.1002/stem.2988] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 12/14/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022]
Abstract
Tendon healing follows a complex series of coordinated events, which ultimately produces a mechanically inferior tissue more scar‐like than native tendon. More regenerative healing occurs when anti‐inflammatory M2 macrophages play a more dominant role. Mesenchymal stromal/stem cells (MSCs) are able to polarize macrophages to an M2 immunophenotype via paracrine mechanisms. We previously reported that coculture of CD14+ macrophages (MQs) with MSCs resulted in a unique M2‐like macrophage. More recently, we generated M2‐like macrophages using only extracellular vesicles (EVs) isolated from MSCs creating “EV‐educated macrophages” (also called exosome‐educated macrophages [EEMs]), thereby foregoing direct use of MSCs. For the current study, we hypothesized that cell therapy with EEMs would improve in vivo tendon healing by modulating tissue inflammation and endogenous macrophage immunophenotypes. We evaluated effects of EEMs using a mouse Achilles tendon rupture model and compared results to normal tendon healing (without any biologic intervention), MSCs, MQs, or EVs. We found that exogenous administration of EEMs directly into the wound promoted a healing response that was significantly more functional and more regenerative. Injured tendons treated with exogenous EEMs exhibited (a) improved mechanical properties, (b) reduced inflammation, and (c) earlier angiogenesis. Treatment with MSC‐derived EVs alone were less effective functionally but stimulated a biological response as evidenced by an increased number of endothelial cells and decreased M1/M2 ratio. Because of their regenerative and immunomodulatory effects, EEM treament could provide a novel strategy to promote wound healing in this and various other musculoskeletal injuries or pathologies where inflammation and inadequate healing is problematic. Stem Cells2019;37:652–662
Collapse
Affiliation(s)
- Connie S Chamberlain
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - Anna E B Clements
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - John A Kink
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Ugeun Choi
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| | - Geoffrey S Baer
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - Matthew A Halanski
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Ray Vanderby
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
44
|
Saldaña L, Bensiamar F, Vallés G, Mancebo FJ, García-Rey E, Vilaboa N. Immunoregulatory potential of mesenchymal stem cells following activation by macrophage-derived soluble factors. Stem Cell Res Ther 2019; 10:58. [PMID: 30760316 PMCID: PMC6375172 DOI: 10.1186/s13287-019-1156-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/09/2019] [Accepted: 01/27/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Immunoregulatory capacity of mesenchymal stem cells (MSC) is triggered by the inflammatory environment, which changes during tissue repair. Macrophages are essential in mediating the inflammatory response after injury and can adopt a range of functional phenotypes, exhibiting pro-inflammatory and anti-inflammatory activities. An accurate characterization of MSC activation by the inflammatory milieu is needed for improving the efficacy of regenerative therapies. In this work, we investigated the immunomodulatory functions of MSC primed with factors secreted from macrophages polarized toward a pro-inflammatory or an anti-inflammatory phenotype. We focused on the role of TNF-α and IL-10, prototypic pro-inflammatory and anti-inflammatory cytokines, respectively, as priming factors for MSC. METHODS Secretion of immunoregulatory mediators from human MSC primed with media conditioned by human macrophages polarized toward a pro-inflammatory or an anti-inflammatory phenotype was determined. Immunomodulatory potential of primed MSC on polarized macrophages was studied using indirect co-cultures. Involvement of TNF-α and IL-10 in priming MSC and of PGE2 in MSC-mediated immunomodulation was investigated employing neutralizing antibodies. Collagen hydrogels were used to study MSC and macrophages interactions in a more physiological environment. RESULTS Priming MSC with media conditioned by pro-inflammatory or anti-inflammatory macrophages enhanced their immunomodulatory potential through increased PGE2 secretion. We identified the pro-inflammatory cytokine TNF-α as a priming factor for MSC. Notably, the anti-inflammatory IL-10, mainly produced by pro-resolving macrophages, potentiated the priming effect of TNF-α. Collagen hydrogels acted as instructive microenvironments for MSC and macrophages functions and their crosstalk. Culturing macrophages on hydrogels stimulated anti-inflammatory versus pro-inflammatory cytokine secretion. Encapsulation of MSC within hydrogels increased PGE2 secretion and potentiated immunomodulation on macrophages, attenuating macrophage pro-inflammatory state and sustaining anti-inflammatory activation. Priming with inflammatory factors conferred to MSC loaded in hydrogels greater immunomodulatory potential, promoting anti-inflammatory activity of macrophages. CONCLUSIONS Factors secreted by pro-inflammatory and anti-inflammatory macrophages activated the immunomodulatory potential of MSC. This was partially attributed to the priming effect of TNF-α and IL-10. Immunoregulatory functions of primed MSC were enhanced after encapsulation in hydrogels. These findings may provide insight into novel strategies to enhance MSC immunoregulatory potency.
Collapse
Affiliation(s)
- Laura Saldaña
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Fátima Bensiamar
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Gema Vallés
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Francisco J. Mancebo
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Eduardo García-Rey
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
| | - Nuria Vilaboa
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| |
Collapse
|
45
|
Kim W, Lee SK, Kwon YW, Chung SG, Kim S. Pioglitazone-Primed Mesenchymal Stem Cells Stimulate Cell Proliferation, Collagen Synthesis and Matrix Gene Expression in Tenocytes. Int J Mol Sci 2019; 20:ijms20030472. [PMID: 30678291 PMCID: PMC6387004 DOI: 10.3390/ijms20030472] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 11/16/2022] Open
Abstract
Various therapeutic effects of mesenchymal stem cells (MSCs) have been reported. However, the rapid clearance of these cells in vivo, difficulties in identifying their therapeutic mechanism of action, and insufficient production levels remain to be resolved. We investigated whether a pioglitazone pre-treatment of MSCs (Pio-MSCs) would stimulate the proliferation of co-cultured tenocytes. Pioglitazone increased the proliferation of MSCs and enhanced the secretion of VEGF (vascular endothelial growth factor) and collagen in these cells. We then examined the effects of Pio-MSCs on tenocytes using an indirect transwell culture system. A significant increase in tenocyte proliferation and cell cycle progression was observed in these co-cultures. Significant increases were observed in wound scratch closure by tenocytes from a Pio-MSC co-culture. Pio-MSCs also enhanced the secretion of collagen from tenocytes. A higher mRNA level of collagen type 1 (Col 1) and type 3 (Col 3), scleraxis (Scx), and tenascin C (TnC) was found in the tenocytes in Pio-MSC co-cultures compared with monocultured cells or tenocytes cultured with non-treated MSCs. Our results indicate that pioglitazone enhances the therapeutic effects of MSCs on tendon repair.
Collapse
Affiliation(s)
- Won Kim
- Department of Rehabilitation Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
- Department of Rehabilitation Medicine, College of Medicine, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea.
| | - Seul Ki Lee
- Stem Cell Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea.
| | - Young-Won Kwon
- Department of Rehabilitation Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
| | - Sun G Chung
- Department of Rehabilitation Medicine, College of Medicine, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea.
- Institute of Aging, Seoul National University, Seoul 03080, Korea.
- Rheumatism Research Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea.
| | - Soo Kim
- Stem Cell Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea.
| |
Collapse
|
46
|
Kouroupis D, Sanjurjo-Rodriguez C, Jones E, Correa D. Mesenchymal Stem Cell Functionalization for Enhanced Therapeutic Applications. TISSUE ENGINEERING PART B-REVIEWS 2018; 25:55-77. [PMID: 30165783 DOI: 10.1089/ten.teb.2018.0118] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IMPACT STATEMENT Culture expansion of MSCs has detrimental effects on various cell characteristics and attributes (e.g., phenotypic changes and senescence), which, in addition to inherent interdonor variability, negatively impact the standardization and reproducibility of their therapeutic potential. The identification of innate distinct functional MSC subpopulations, as well as the description of ex vivo protocols aimed at maintaining phenotypes and enhancing specific functions have the potential to overcome these limitations. The incorporation of those approaches into cell-based therapy would significantly impact the field, as more reproducible clinical outcomes may be achieved.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- 1 Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, Florida.,2 Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Clara Sanjurjo-Rodriguez
- 3 Leeds Institute of Rheumatic and Musculoskeletal Disease, Saint James University Hospital, University of Leeds, Leeds, United Kingdom.,4 Department of Biomedical Sciences, Medicine and Physiotherapy, University of A Coruña, CIBER-BBN-Institute of Biomedical Research of A Coruña (INIBIC), A Coruña, Spain
| | - Elena Jones
- 3 Leeds Institute of Rheumatic and Musculoskeletal Disease, Saint James University Hospital, University of Leeds, Leeds, United Kingdom
| | - Diego Correa
- 1 Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, Florida.,2 Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
47
|
Song H, Yin Z, Wu T, Li Y, Luo X, Xu M, Duan L, Li J. Enhanced Effect of Tendon Stem/Progenitor Cells Combined With Tendon-Derived Decellularized Extracellular Matrix on Tendon Regeneration. Cell Transplant 2018; 27:1634-1643. [PMID: 30298746 PMCID: PMC6299202 DOI: 10.1177/0963689718805383] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Decellularized extracellular matrices have been clinically used for tendon regeneration. However, only a few systematic studies have compared tendon stem/progenitor cells to mesenchymal stromal cells on the tendon-derived decellularized matrix. In the present study, we prepared extracellular matrix derived from porcine tendons and seeded with tendon stem/progenitor cells, embryonic stem cell-derived mesenchymal stromal cells or without stem cells. Then we implanted the mixture (composed of stem cells and scaffold) into the defect of a rat Achilles tendon. Next, 4 weeks post-surgery the regenerated tendon tissue was collected. Histological staining, immunohistochemistry, determination of collagen content, transmission electron microscopy, and biomechanical testing were performed to evaluate the tendon structure and biomechanical properties. Our study collectively demonstrated that decellularized extracellular matrix derived from porcine tendons significantly promoted the regeneration of injured tendons when combined with tendon stem/progenitor cells or embryonic stem cell-mesenchymal stromal cells. Compared to embryonic stem cell-mesenchymal stromal cells, tendon stem/progenitor cells combined with decellularized matrix showed more improvement in the structural and biomechanical properties of regenerated tendons in vivo. These findings suggest a promising strategy for functional tendon tissue regeneration and further studies are warranted to develop a functional tendon tissue regeneration utilizing tendon stem/progenitor cells integrated with a tendon-derived decellularized matrix.
Collapse
Affiliation(s)
- Haixin Song
- Department of Rehabilitation, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Tao Wu
- Department of Rehabilitation, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yangzheng Li
- Department of Rehabilitation, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong, China.,Shenzhen Sanming Group, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Mingzhu Xu
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA.,Department of Rehabilitation, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Lihong Duan
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA.,Shenzhen Institute of Geriatrics, Shenzhen, China
| | - Jianhua Li
- Department of Rehabilitation, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
48
|
Devana SK, Kelley BV, McBride OJ, Kabir N, Jensen AR, Park SJ, Eliasberg CD, Dar A, Mosich GM, Kowalski TJ, Péault B, Petrigliano FA, SooHoo NF. Adipose-derived Human Perivascular Stem Cells May Improve Achilles Tendon Healing in Rats. Clin Orthop Relat Res 2018; 476:2091-2100. [PMID: 30179944 PMCID: PMC6259872 DOI: 10.1097/corr.0000000000000461] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Achilles tendon rupture is a common injury and the best treatment option remains uncertain between surgical and nonoperative methods. Biologic approaches using multipotent stem cells such as perivascular stem cells pose a possible treatment option, although there is currently a paucity of evidence regarding their clinical therapeutic use. QUESTIONS/PURPOSES The purpose of this study was to determine whether injected perivascular stem cells (PSCs) would (1) improve histologic signs of tendon healing (such as percent area of collagen); and (2) improve biomechanical properties (peak load or stiffness) in a rat model of Achilles tendon transection. METHODS Two subtypes of PSCs were derived from human adipose tissue: pericytes (CD146CD34CD45CD31) and adventitial cells (CD146CD34CD45CD31). Thirty-two athymic rats underwent right Achilles transection and were randomized to receive injection with saline (eight tendons), hydrogel (four tendons), pericytes in hydrogel (four tendons), or adventitial cells in hydrogel (eight tendons) 3 days postoperatively with the left serving as an uninjured control. Additionally, a subset of pericytes was labeled with CM-diI to track cell viability and localization. At 3 weeks, the rats were euthanized, and investigators blinded to treatment group allocation evaluated tendon healing by peak load and stiffness using biomechanical testing and percent area of collagen using histologic analysis with picrosirius red staining. RESULTS Histologic analysis showed a higher mean percent area collagen for pericytes (30%) and adventitial cells (28%) than hydrogel (21%) or saline (26%). However, a nonparametric statistical analysis yielded no statistical difference. Mechanical testing demonstrated that the pericyte group had a higher peak load than the saline group (41 ± 7 N versus 26 ± 9 N; mean difference 15 N; 95% confidence interval [CI], 4-27 N; p = 0.003) and a higher peak load than the hydrogel group (41 ± 7 N versus 25 ± 3 N; mean difference 16; 95% CI, 8-24 N; p = 0.001). The pericyte group demonstrated higher stiffness than the hydrogel group (36 ± 12 N/mm versus 17 ± 6 N/mm; mean difference 19 N/mm; 95% CI, 5-34 N/mm; p = 0.005). CONCLUSIONS Our results suggest that injection of PSCs improves mechanical but not the histologic properties of early Achilles tendon healing. CLINICAL RELEVANCE This is a preliminary study that provides more insight into the use of adipose-derived PSCs as a percutaneous therapy in the setting of Achilles tendon rupture. Further experiments to characterize the function of these cells may serve as a pathway to development of minimally invasive intervention aimed at improving nonoperative management while avoiding the complications associated with surgical treatment down the line.
Collapse
|
49
|
Lipar M, Zdilar B, Kreszinger M, Ćorić M, Radišić B, Samardžija M, Žic R, Pećin M. Extracellular matrix supports healing of transected rabbit Achilles tendon. Heliyon 2018; 4:e00781. [PMID: 30225380 PMCID: PMC6138787 DOI: 10.1016/j.heliyon.2018.e00781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/20/2018] [Accepted: 09/06/2018] [Indexed: 01/17/2023] Open
Abstract
Extracellular matrix (ECM) has been intensively used in cardio surgery. The main goal of this research was to determine if Achilles tendon healing could be promoted by applying extracellular matrix scaffold (CorMatrix®, USA). Sixteen (n = 16) New Zealand white mature rabbits (Oryctolagus cuniculus) were randomly allocated into two groups. Following complete surgical transection, rabbits in group A (ECM applied) (n = 8) had their Achilles tendons reconstructed using both, nylon suture and extracellular matrix scaffold, whereas in group B (without ECM) the tendons were reconstructed using nylon suture only. After four weeks, the rabbits were euthanized and tendon samples harvested and stained with hematoxylin eosin, Mallory, and Gomory and subsequently histologically analyzed according to modified Bonnar scale. Group B had significantly stronger inflammatory response, including abundant cell infiltration and neovascularization. In group A collagen fibers were predominantly found, whereas in group B reticular fibers were more abundant. Extracellular matrix scaffold has been found to have the real potential for promoting tendon healing through accelerating collagen formation, which is crucial for restoring biomechanical properties of a tendon, decreasing peritendineous adhesion formation, and reducing inflammatory edema and subsequently pain.
Collapse
Affiliation(s)
- Marija Lipar
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
| | - Boris Zdilar
- Hospital Sveti Duh, Ulica Sveti Duh 1, 10000 Zagreb, Croatia
| | - Mario Kreszinger
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
| | - Marijana Ćorić
- School of Medicine, University of Zagreb, Ulica Šalata 2, 10000 Zagreb, Croatia
| | - Berislav Radišić
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
| | - Marko Samardžija
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
| | - Rado Žic
- School of Medicine, University of Zagreb, Ulica Šalata 2, 10000 Zagreb, Croatia
| | - Marko Pećin
- Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
| |
Collapse
|
50
|
Barrachina L, Remacha AR, Romero A, Vitoria A, Albareda J, Prades M, Roca M, Zaragoza P, Vázquez FJ, Rodellar C. Assessment of effectiveness and safety of repeat administration of proinflammatory primed allogeneic mesenchymal stem cells in an equine model of chemically induced osteoarthritis. BMC Vet Res 2018; 14:241. [PMID: 30119668 PMCID: PMC6098603 DOI: 10.1186/s12917-018-1556-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/26/2018] [Indexed: 01/15/2023] Open
Abstract
Background This study aimed at assessing the effectiveness and safety of repeated administrations of allogeneic bone marrow-derived mesenchymal stem cells (BM-MSCs) primed with tumor necrosis factor (TNF)-α and interferon-γ in an equine model of chemically-induced osteoarthritis. Arthritis was induced in both radio-carpal (RC)-joints by amphotericin-B in 18 ponies, divided into three groups depending on the treatment injected: MSC-naïve (n = 7), MSC-primed (n = 7) and control (n = 4). The study consisted of two phases and used one RC-joint of each animal in each phase, with four months time-lapse, in order to assess two end-points. Clinical, synovial, radiological and ultrasonographic follow-up was performed. At six months, animals were euthanized and both carpi were assessed by magnetic resonance imaging (MRI), gross anatomy, histopathology, histochemistry and gene expression. Results Clinical and synovial inflammatory signs were quicker reduced in MSC-treated groups and repeated allogeneic administration did not produce adverse reactions, but MSC-primed group showed slight and transient local inflammation after second injection. Radiology and MRI did not show significant differences between treated and control groups, whereas ultrasonography suggested reduced synovial effusion in MSC-treated groups. Both MSC-treated groups showed enhanced cartilage gross appearance at two compared to six months (MSC-naïve, p < 0.05). Cartilage histopathology did not reveal differences but histochemistry suggested delayed progression of proteoglycan loss in MSC-treated groups. Synovium histopathology indicated decreased inflammation (p < 0.01) in MSC-primed and MSC-naïve at two and six months, respectively. At two months, cartilage from MSC-primed group significantly (p < 0.05) upregulated collagen type II (COL2A1) and transforming growth factor (TGF)-β1 and downregulated cyclooxygenase-2 and interleukin (IL)-1β. At six months, MSC-treatments significantly downregulated TNFα (p < 0.05), plus MSC-primed upregulated (p < 0.05) COL2A1, aggrecan, cartilage oligomeric protein, tissue inhibitor of metalloproteinases-2 and TGF-β1. In synovium, both MSC-treatments decreased (p < 0.01) matrix metalloproteinase-13 expression at two months and MSC-primed also downregulated TNFα (p < 0.05) and IL-1β (p < 0.01). Conclusions Both MSC-treatments provided beneficial effects, mostly observed at short-term. Despite no huge differences between MSC-treatments, the findings suggested enhanced anti-inflammatory and regulatory potential of MSC-primed. While further research is needed to better understand these effects and clarify immunogenicity implications, these findings contribute to enlarge the knowledge about MSC therapeutics and how they could be influenced. Electronic supplementary material The online version of this article (10.1186/s12917-018-1556-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Barrachina
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Ana Rosa Remacha
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Antonio Romero
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Arantza Vitoria
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Jorge Albareda
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía Ortopédica y Traumatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza. Avda. San Juan Bosco, 15, 50009, Zaragoza, Spain
| | - Marta Prades
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Departament de Medicina i Cirugia Animal, Universidad Autónoma de Barcelona, Edifici H, UAB, 08193 Bellaterra, Barcelona, Spain
| | - Mercedes Roca
- Clínica Doctora Roca Diagnóstico Médico, Carrera del Sábado 4, local (Edificio Europa), 50006, Zaragoza, Spain
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Francisco José Vázquez
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Clementina Rodellar
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto Agroalimentario de Aragón IA2 - Instituto de Investigación Sanitaria de Aragón IIS, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.
| |
Collapse
|