1
|
Pueyo Moliner A, Ito K, Zaucke F, Kelly DJ, de Ruijter M, Malda J. Restoring articular cartilage: insights from structure, composition and development. Nat Rev Rheumatol 2025; 21:291-308. [PMID: 40155694 DOI: 10.1038/s41584-025-01236-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/01/2025]
Abstract
Articular cartilage can withstand substantial compressive and shear forces within the joint and also reduces friction during motion. The exceptional mechanical properties of articular cartilage stem from its highly organized extracellular matrix (ECM). The ECM is composed mainly of collagen type II and is pivotal in conferring mechanical durability to the tissue within its proteoglycan-rich matrix. Articular cartilage is prone to injury and degeneration, and current treatments often fail to restore the mechanical function of this tissue. A key challenge is replicating the intricate collagen-proteoglycan network, which is essential for the long-lasting restoration and mechanical durability of the tissue. Understanding articular cartilage development, which arises between late embryonic and early juvenile development, is vital for the creation of durable therapeutic strategies. The development of the articular ECM involves the biosynthesis, fibrillogenesis and self-assembly of the collagen type II network, which, along with proteoglycans and minor ECM components, shapes the architecture of adult articular cartilage. A deeper understanding of these processes could inform biomaterial-based therapies aimed at improving therapeutic outcomes. Emerging biofabrication technologies offer new opportunities to integrate developmental principles into the creation of durable articular cartilage implants. Bridging fundamental biology with innovative engineering offers novel approaches to generating more-durable 3D implants for articular cartilage restoration.
Collapse
Affiliation(s)
- Alba Pueyo Moliner
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Keita Ito
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Frank Zaucke
- Department of Trauma Surgery and Orthopedics, Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mylène de Ruijter
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands.
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands.
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
2
|
Zhang A, Sun B, Nan C, Cong L, Zhao Z, Liu L. Effects of 3D-printed exosome-functionalized brain acellular matrix hydrogel on neuroinflammation in rats following cerebral hemorrhage. Stem Cell Res Ther 2025; 16:196. [PMID: 40254565 PMCID: PMC12010578 DOI: 10.1186/s13287-025-04332-3] [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: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Exosome-based therapeutics have garnered significant attention for intracerebral hemorrhage (ICH) treatment due to their capacity to regulate metabolic dysregulation, restore cellular homeostasis, and modulate the injury microenvironment via bioactive cargoes such as microRNAs and proteins. However, rapid systemic clearance and enzymatic degradation critically limit their therapeutic efficacy. To address this challenge, we engineered a three-dimensional (3D) bioprinted scaffold capable of encapsulating and sustaining the release of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-exos). METHODS Based on previous research [1-3], the scaffold was composed of a decellularized brain matrix (dECM), gelatin-methacryloyl (GelMA), and silk fibroin (SF) crosslinked with a photoinitiator. hUCMSC-exos were precisely incorporated via extrusion-based 3D bioprinting. Release kinetics were assessed via in vitro elution and in vivo imaging. An ICH rat model received stereotaxic implantation of the exosome-laden scaffold (dECM@exo). Neuroinflammatory markers (IL-6, TNF-α, IL-10) and apoptotic activity (JC-1, Annexin V/PI, TUNEL) were quantified. Neurological outcomes were longitudinally evaluated using the modified Longa scale, Bederson scoring, and sensorimotor tests (rotarod, forelimb placement) at 1, 4, 7 and 14 days post-ICH. RESULTS dECM@exo demonstrated sustained exosome release over 14 days, significantly promoting neural tissue regeneration while attenuating perihematomal edema. Mechanistically, the scaffold modulated pathological MMP activity and inflammatory cytokine expression, thereby restoring extracellular matrix homeostasis and reducing neuronal apoptosis. CONCLUSIONS The findings demonstrate that the 3D biological scaffold dECM@exo effectively maintains microenvironmental homeostasis in the early stages of ICH and improves outcomes associated with the condition. dECM@exo is poised to serve as a robust platform for drug delivery and biotherapy in ICH treatment, offering a viable alternative for managing this condition.
Collapse
Affiliation(s)
- Aobo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| | - Boyu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Lulu Cong
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
3
|
Wang L, Hao M, Xu Y, Wang Z, Xie H, Zhang B, Zhang X, Lin J, Sun X, Wang J, Wu Q. Adipose-derived stem cells attenuate rheumatoid arthritis by restoring CX 3CR1 + synovial lining macrophage barrier. Stem Cell Res Ther 2025; 16:111. [PMID: 40038808 PMCID: PMC11881422 DOI: 10.1186/s13287-025-04144-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 01/13/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease and the integrity of CX3CR1+ synovial macrophage barrier significantly impacts its progression. However, the mechanisms driving the dynamic changes of this macrophage barrier remain unclear. Traditional drug therapies for RA have substantial limitations. Mesenchymal stem cells (MSCs)-based cell therapy, especially adipose-derived stem cells (ADSCs), hold therapeutic promise. Nevertheless, the underlying therapeutic mechanism of ADSCs, especially their interactions with CX3CR1+ macrophages, require further investigation. METHODS To explore the interaction between ADSCs and CX3CR1+ synovial macrophages during barrier reconstruction, underlying the therapeutic mechanism of ADSCs and the mechanisms on the dynamic changes of the macrophage barrier, scRNA-seq analysis was conducted 4 days after ADSCs injection in serum transfer-induced arthritis model mice. The roles of mitochondria transfer and ADSCs transplantation were also explored. Bulk RNA-seq analysis was performed after the co-culture of ADSCs and CX3CR1+ synovial macrophages. To study the in vivo fate of ADSCs, bulk RNA-seq was performed on ADSCs retrieved at 0, 2, 4, and 7 days post-injection. RESULTS Intra-articular injection of ADSCs effectively attenuated the pathological progression of mice with serum transfer-induced arthritis. ADSCs gradually adhered to CX3CR1+ macrophages, facilitating the restore of the macrophage barrier, while the absence of this barrier greatly weakened the therapeutic effect of ADSCs. scRNA-seq analysis revealed an Atf3high Ccl3high subset of CX3CR1+ macrophages with impaired oxidative phosphorylation that increased during RA progression. ADSCs-mediated reduction of this subset appeared to be linked to mitochondrial transfer, and transplantation of isolated ADSCs-derived mitochondria also proved effective in treating RA. Both bulk RNA-seq and scRNA-seq analyses revealed multiple interaction mechanisms between ADSCs and CX3CR1+ macrophages, including Cd74/Mif axis and GAS6/MERTK axis, which contribute to barrier restoration and therapeutic effects. Furthermore, bulk RNA-seq analysis showed that ADSCs primarily contribute to tissue repair and immune regulation subsequently. CONCLUSIONS Our results suggest that ADSCs ameliorated the energy metabolism signature of CX3CR1+ lining macrophages and may promote barrier restoration through mitochondria transfer. In addition, we elucidated the fate of ADSCs and the therapeutic potential of mitochondria in RA treatment.
Collapse
Affiliation(s)
- Lei Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ming Hao
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yongyue Xu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhaoyan Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Hanqi Xie
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bo Zhang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xue Zhang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jun Lin
- Department of Orthopaedics, Suzhou Dushu Lake Hospital, The Fourth Affiliated of Soochow University, Medical Center of Soochow University, Suzhou, 215001, Jiangsu, China
| | - Xiaodan Sun
- School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of New Ceramics and Fine Processing, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, Tsinghua University, Beijing, 100084, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qiong Wu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
4
|
Wu Z, Zhang P, Huang W, Zhou Y, Cao Z, Wu C. Qufeng epimedium decoction alleviates rheumatoid arthritis through CYLD-antagonized NF-kB activation by deubiquitinating Sirt1. Immunobiology 2025; 230:152875. [PMID: 39908772 DOI: 10.1016/j.imbio.2025.152875] [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: 10/11/2024] [Revised: 12/20/2024] [Accepted: 01/25/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease that markedly limits the patients´ day-to-day functional abilities and life quality. Currently, there is no known cure for RA. Qufeng epimedium decoction, a traditional Chinese medicine, is widely used in China to treat RA. However, its underlying mechanism remains elusive. METHODS The RA animal model was established to investigate the anti-RA effect and regulatory effect on fibroblast-like synoviocytes (FLS) pyroptosis, qRT-PCR, Western blot, flow cytometry, histology staining, and ELISA were utilized to confirm the gene and protein expressions. The interactions between Sirt1 and CYLD were validated through Co-immunoprecipitation (Co-IP) and RNA-FISH assay. RESULTS Administration with Qufeng epimedium decoction attenuated inflammatory damage, excessive proliferation, and FLSs pyroptosis in an RA rat model. Moreover, treatment of Qufeng epimedium decoction reduced the ubiquitination modification level of Sirt1 in FLSs isolated from an RA rat model. Mechanistically, CYLD, an intermediation for linking Qufeng epimedium decoction and RA, was responsible for Sirt1 deubiquitination to its protein stabilization, thereby deactivating the NF-kB /GSDMD signaling pathway. CONCLUSION Our findings indicate that Qufeng epimedium decoction suppresses FLSs pyroptosis and RA progression via CYLD-mediated Sirt1 deubiquitination and deactivation of the NF-kB /GSDMD signaling pathway. This study sheds light on the underlying mechanism of Qufeng epimedium decoction's effectiveness in RA treatment.
Collapse
Affiliation(s)
- Zhiming Wu
- Chinese Medicine Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, PR China.
| | - Peng Zhang
- Chinese Medicine Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, PR China
| | - Wenyan Huang
- Day Surgery Center, Jiangxi Maternal and Child Health Care Hospital, Nanchang 330000, Jiangxi Province, PR China
| | - Yifen Zhou
- Chinese Medicine Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, PR China
| | - Zhengliu Cao
- Chinese Medicine Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, PR China
| | - Chunhong Wu
- Outpatient Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, PR China
| |
Collapse
|
5
|
Brandt MD, Malone JB, Kean TJ. Advances and Challenges in the Pursuit of Disease-Modifying Osteoarthritis Drugs: A Review of 2010-2024 Clinical Trials. Biomedicines 2025; 13:355. [PMID: 40002768 PMCID: PMC11853018 DOI: 10.3390/biomedicines13020355] [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: 12/09/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Osteoarthritis (OA) is a highly prevalent, degenerative joint disease capable of causing severe pain and impaired mobility. Current treatments mitigate symptoms but do not cure the disease. The development of a disease-modifying osteoarthritis drug (DMOAD) could improve patient outcomes by slowing, halting, or reversing joint damage. Many DMOADs have progressed to clinical trials, but very few have made a significant impact, and none have been approved for clinical use. The purpose of this review is to present an update on the current status of DMOADs with a particular focus on results published since 2010. Methods: A comprehensive search was conducted within PubMed and ClinicalTrials.gov for novel DMOADs enrolled in phase II and III clinical trials between 1 January 2010 and 1 July 2024. Results: Eleven DMOAD candidates are reviewed and critically analyzed for their potential benefit in OA treatment-Lorecivivint (SM04690), TissueGene-C, Cindunistat (SD-6010), Sprifermin, UBX0101, TPX-100, GLPG1972/S201086, Lutikizumab (ABT-981), SAR113945, MIV-711, and LNA043-and relevant challenges to their development are discussed. Conclusions: Six DMOADs have demonstrated statistically significant evidence of a structural or symptomatic benefit without major safety concerns in phase II and III randomized controlled trials post-2010.
Collapse
Affiliation(s)
- Mckenzie D. Brandt
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Jason B. Malone
- Department of Orthopedic Surgery, Nemours Children’s Health System, Orlando, FL 32827, USA;
| | - Thomas J. Kean
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
6
|
Meenakshi Sundaram RS, Rupert S, Srinivasan P, Sathyanesan J, Govarthanan K, Jeyaraman N, Ramasubramanian S, Jeyaraman M, Chung HY, Gangadaran P, Ahn BC. Decoding Cytokine Dynamics: Wharton's Jelly Stromal Cells and Chondro-Differentiates in PHA-Stimulated Co-Culture. Cells 2025; 14:174. [PMID: 39936966 PMCID: PMC11817647 DOI: 10.3390/cells14030174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION Articular cartilage damage presents a significant clinical challenge, with limited options for effective regeneration. Mesenchymal stromal cells (MSCs) derived from Wharton's jelly (WJ) are a promising cell source for cartilage repair due to their regenerative and immunomodulatory properties. While undifferentiated MSCs have demonstrated potent immunoregulatory effects, the immunomodulatory potential of chondrocytes derived from WJ-MSCs remains underexplored, particularly under inflammatory conditions. This study investigates the differential cytokine expression profiles of WJ-MSC-derived chondrocytes and undifferentiated MSCs under inflammatory stimulation with phytohemagglutinin (PHA) to understand their immunomodulatory capacities. MATERIALS AND METHODS WJ-MSCs were differentiated into chondrocytes using a micromass culture system. Differentiated chondrocytes were then co-cultured with immune cells under PHA-induced inflammatory conditions. Control groups included co-cultured cells without PHA activation and chondrocytes activated with PHA in the absence of immune cell interaction. Cytokine expression profiles were analyzed using the RT2 Customized Gene Array to evaluate pro- and anti-inflammatory markers. Morphological changes were assessed microscopically. The immunomodulatory responses of chondrocytes were compared to those of undifferentiated MSCs under the same experimental conditions. RESULTS Chondrocytes co-cultured with immune cells under PHA activation exhibited downregulation of IDO, HLA-G, PDGF, IL-10, TNF-α, IL-6, and IFN-γ compared to undifferentiated MSCs in similar conditions. In non-PHA co-cultured conditions, chondrocytes showed increased expression of IL-6, IFN-γ, IL-4, VEGF, iNOS, PDGF, PTGS-2 and TGF-β, while TNF-α, IL-10, IDO and HLA-G were decreased. In contrast, chondrocytes activated with PHA without immune cell interaction displayed reduced expression of HLA-G and TNF-α, with no significant changes in IL-6, IFN-γ, IL-4, IL-10, VEGF, PDGF, PTGS-2, TGF-β, IDO, and iNOS compared to PHA-stimulated undifferentiated MSCs. CONCLUSION This study demonstrates that chondrocytes derived from WJ-MSCs exhibit limited immunomodulatory potential compared to undifferentiated MSCs, particularly under PHA-induced inflammatory conditions. Undifferentiated MSCs showed superior regulation of key cytokines associated with immune modulation. These findings suggest that maintaining MSCs in an undifferentiated state may be advantageous for therapeutic applications targeting inflammatory conditions, such as osteoarthritis. Future research should explore strategies to enhance the immunomodulatory efficacy of chondrocytes, potentially through genetic modification or adjunctive therapies.
Collapse
Affiliation(s)
- Raja Sundari Meenakshi Sundaram
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Secunda Rupert
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Prasanna Srinivasan
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Jeswanth Sathyanesan
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Kavitha Govarthanan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India;
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600017, Tamil Nadu, India; (N.J.); (M.J.)
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Swaminathan Ramasubramanian
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600017, Tamil Nadu, India; (N.J.); (M.J.)
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea;
| | - Prakash Gangadaran
- 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, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- 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, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
7
|
Patel PR, Tamas IP, Van Der Bas M, Kegg A, Hilliard BA, Lambi AG, Popoff SN, Barbe MF. Repetitive Overuse Injury Causes Entheseal Damage and Palmar Muscle Fibrosis in Older Rats. Int J Mol Sci 2024; 25:13546. [PMID: 39769311 PMCID: PMC11679654 DOI: 10.3390/ijms252413546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Overuse injury is a frequent diagnosis in occupational medicine and athletics. Using an established model of upper extremity overuse, we sought to characterize changes occurring in the forepaws and forelimbs of mature female rats (14-18 months of age). Thirty-three rats underwent a 4-week shaping period, before performing a high-repetition low-force (HRLF) task for 12 weeks, with the results being compared to 32 mature controls. HRLF animals showed a reduced grip strength versus controls. ELISAs carried out in the HRLF rats, versus controls, showed elevated levels of IL1-α in tendons, IL1-α and TNF-α in distal bones/entheses, and TNF-α, MIP1-α/CCL3, and CINC-2/CXCL-3 in serum, as well as IL-6 in forelimb muscles and tendons, and IL-10 in serum. HRLF rats had elevated collagen deposition in the forepaw intrinsic muscles (i.e., fibrosis), entheseal microdamage, and articular cartilage degradation versus the control rats. CD68/ED1+ osteoclasts and single-nucleated cells were elevated in distal forelimb metaphyses of the HRLF animals, versus controls. Declines in grip strength correlated with muscle fibrosis, entheseal microdamage, articular cartilage damage, distal bone/enthesis IL1-α, and serum IL-6. These data demonstrate inflammatory and persistent degradative changes in the forearm/forepaw tissues of mature female animals exposed to prolonged repetitive tasks, changes with clinical relevance to work-related overuse injuries in mature human females.
Collapse
Affiliation(s)
- Parth R. Patel
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Istvan P. Tamas
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Megan Van Der Bas
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Abby Kegg
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Brendan A. Hilliard
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Alex G. Lambi
- Plastic Surgery Section, New Mexico Veterans Administration Health Care System, Albuquerque, NM 87108, USA;
- Department of Surgery, The University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Steven N. Popoff
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA;
| | - Mary F. Barbe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
8
|
Wu KC, Chang YH, Ding DC, Lin SZ. Mesenchymal Stromal Cells for Aging Cartilage Regeneration: A Review. Int J Mol Sci 2024; 25:12911. [PMID: 39684619 PMCID: PMC11641625 DOI: 10.3390/ijms252312911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cartilage degeneration is a key feature of aging and osteoarthritis, characterized by the progressive deterioration of joint function, pain, and limited mobility. Current treatments focus on symptom relief, not cartilage regeneration. Mesenchymal stromal cells (MSCs) offer a promising therapeutic option due to their capability to differentiate into chondrocytes, modulate inflammation, and promote tissue regeneration. This review explores the potential of MSCs for cartilage regeneration, examining their biological properties, action mechanisms, and applications in preclinical and clinical settings. MSCs derived from bone marrow, adipose tissue, and other sources can self-renew and differentiate into multiple cell types. In aging cartilage, they aid in tissue regeneration by secreting growth factors and cytokines that enhance repair and modulate immune responses. Recent preclinical studies show that MSCs can restore cartilage integrity, reduce inflammation, and improve joint function, although clinical translation remains challenging due to limitations such as cell viability, scalability, and regulatory concerns. Advancements in MSC delivery, including scaffold-based approaches and engineered exosomes, may improve therapeutic effectiveness. Potential risks, such as tumorigenicity and immune rejection, are also discussed, emphasizing the need for optimized treatment protocols and large-scale clinical trials to develop effective, minimally invasive therapies for cartilage regeneration.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
9
|
Zhang A, Cong L, Nan C, Zhao Z, Liu L. 3D biological scaffold delivers Bergenin to reduce neuroinflammation in rats with cerebral hemorrhage. J Transl Med 2024; 22:946. [PMID: 39420402 PMCID: PMC11484212 DOI: 10.1186/s12967-024-05735-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a severe form of stroke characterized by high incidence and mortality rates. Currently, there is a significant lack of effective treatments aimed at improving clinical outcomes. Our research team has developed a three-dimensional (3D) biological scaffold that incorporates Bergenin, allowing for the sustained release of the compound. METHODS This 3D biological scaffold was fabricated using a combination of photoinitiator, GEMA, silk fibroin, and decellularized brain matrix (dECM) to encapsulate Bergenin through advanced 3D bioprinting techniques. The kinetics of drug release were evaluated through both in vivo and in vitro studies. A cerebral hemorrhage model was established, and a 3D biological scaffold containing Bergenin was transplanted in situ. Levels of inflammatory response, oxidative stress, and apoptosis were quantified. The neurological function of rats with cerebral hemorrhage was assessed on days 1, 3, and 5 using the turning test, forelimb placement test, Longa score, and Bederson score. RESULTS The 3D biological scaffold incorporating Bergenin significantly enhances the maintenance of drug concentration in the bloodstream, leading to a marked reduction in inflammatory markers such as IL-6, iNOS, and COX-2 levels in a cerebral hemorrhage model, primarily through the inhibition of the NF-κB pathway. Additionally, the scaffold effectively reduces the expression of hypoxia-inducible factor 1-alpha (HIF-1α) in primary cultured astrocytes, which in turn decreases the production of reactive oxygen species (ROS) and inhibits IL-6 production induced by hemin. Subsequent experiments reveal that the 3D biological scaffold containing Bergenin promotes the activation of the Nrf-2/HO-1 signaling pathway, both in vivo and in vitro, thereby preventing cell death. Moreover, the application of this 3D biological scaffold has been demonstrated to improve drug retention in the bloodstream. CONCLUSION This strategy effectively mitigates inflammation, oxidative stress, and cell death in rats with cerebral hemorrhage by inhibiting the NF-κB pathway while concurrently activating the Nrf-2/HO-1 pathway.
Collapse
Affiliation(s)
- Aobo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Lulu Cong
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
10
|
Kim JR, Hong BK, Pham THN, Kim WU, Kim HA. Interferon-gamma signaling promotes cartilage regeneration after injury. Sci Rep 2024; 14:8046. [PMID: 38580748 PMCID: PMC10997668 DOI: 10.1038/s41598-024-58779-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/03/2024] [Indexed: 04/07/2024] Open
Abstract
Osteoarthritis is a common chronic disease and major cause of disability and chronic pain in ageing populations. In this pathology, the entire joint is involved, and the regeneration of articular cartilage still remains one of the main challenges. Here, we investigated the molecular mechanisms underlying cartilage regeneration in young mice using a full-thickness cartilage injury (FTCI) model. FTCI-induced cartilage defects were created in the femoral trochlea of young and adult C57BL/6 mice. To identify key molecules and pathways involved in the early response to cartilage injury, we performed RNA sequencing (RNA-seq) analysis of cartilage RNA at 3 days after injury. Young mice showed superior cartilage regeneration compared to adult mice after cartilage injury. RNA-seq analysis revealed significant upregulation of genes associated with the immune response, particularly in the IFN-γ signaling pathway and qRT-PCR analysis showed macrophage polarization in the early phase of cartilage regeneration (3 days) in young mice after injury, which might promote the removal of damaged or necrotic cells and initiate cartilage regeneration in response to injury. IFN-γR1- and IFN-γ-deficient mice exhibited impaired cartilage regeneration following cartilage injury. DMM-induced and spontaneous OA phenotypes were exacerbated in IFN-γR1-/- mice than in wild-type mice. Our data support the hypothesis that IFN-γ signaling is necessary for cartilage regeneration, as well as for the amelioration of post-traumatic and age-induced OA.
Collapse
Affiliation(s)
- Ju-Ryoung Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyungchon, Anyang, Kyunggi, 14068, Korea
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Center for Intergrative Rheumatoid Transcriptomics and Dynamics, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Bong-Ki Hong
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Center for Intergrative Rheumatoid Transcriptomics and Dynamics, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Thi Hong Nhung Pham
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyungchon, Anyang, Kyunggi, 14068, Korea
- Institute for Skeletal Aging, Hallym University, Chuncheon, Gangwon-do, 24252, Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Center for Intergrative Rheumatoid Transcriptomics and Dynamics, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Hyun Ah Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyungchon, Anyang, Kyunggi, 14068, Korea.
- Institute for Skeletal Aging, Hallym University, Chuncheon, Gangwon-do, 24252, Korea.
| |
Collapse
|
11
|
Chen X, Yang M, Zhou Z, Sun J, Meng X, Huang Y, Zhu W, Zhu S, He N, Zhu X, Han X, Liu H. An Anti-Oxidative Bioink for Cartilage Tissue Engineering Applications. J Funct Biomater 2024; 15:37. [PMID: 38391890 PMCID: PMC10889144 DOI: 10.3390/jfb15020037] [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: 12/11/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Since chondrocytes are highly vulnerable to oxidative stress, an anti-oxidative bioink combined with 3D bioprinting may facilitate its applications in cartilage tissue engineering. We developed an anti-oxidative bioink with methacrylate-modified rutin (RTMA) as an additional bioactive component and glycidyl methacrylate silk fibroin as a biomaterial component. Bioink containing 0% RTMA was used as the control sample. Compared with hydrogel samples produced with the control bioink, solidified anti-oxidative bioinks displayed a similar porous microstructure, which is suitable for cell adhesion and migration, and the transportation of nutrients and wastes. Among photo-cured samples prepared with anti-oxidative bioinks and the control bioink, the sample containing 1 mg/mL of RTMA (RTMA-1) showed good degradation, promising mechanical properties, and the best cytocompatibility, and it was selected for further investigation. Based on the results of 3D bioprinting tests, the RTMA-1 bioink exhibited good printability and high shape fidelity. The results demonstrated that RTMA-1 reduced intracellular oxidative stress in encapsulated chondrocytes under H2O2 stimulation, which results from upregulation of COLII and AGG and downregulation of MMP13 and MMP1. By using in vitro and in vivo tests, our data suggest that the RTMA-1 bioink significantly enhanced the regeneration and maturation of cartilage tissue compared to the control bioink, indicating that this anti-oxidative bioink can be used for 3D bioprinting and cartilage tissue engineering applications in the future.
Collapse
Affiliation(s)
- Xin Chen
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Mengni Yang
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha 410082, China
| | - Jingjing Sun
- College of Biology, Hunan University, Changsha 410082, China
| | - Xiaolin Meng
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Yuting Huang
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Wenxiang Zhu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Shuai Zhu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Ning He
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha 410082, China
| | - Xiaolong Zhu
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha 410082, China
| | - Xiaoxiao Han
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha 410082, China
| | - Hairong Liu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
12
|
Zhao T, Li X, Li H, Deng H, Li J, Yang Z, He S, Jiang S, Sui X, Guo Q, Liu S. Advancing drug delivery to articular cartilage: From single to multiple strategies. Acta Pharm Sin B 2023; 13:4127-4148. [PMID: 37799383 PMCID: PMC10547919 DOI: 10.1016/j.apsb.2022.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage (AC) injuries often lead to cartilage degeneration and may ultimately result in osteoarthritis (OA) due to the limited self-repair ability. To date, numerous intra-articular delivery systems carrying various therapeutic agents have been developed to improve therapeutic localization and retention, optimize controlled drug release profiles and target different pathological processes. Due to the complex and multifactorial characteristics of cartilage injury pathology and heterogeneity of the cartilage structure deposited within a dense matrix, delivery systems loaded with a single therapeutic agent are hindered from reaching multiple targets in a spatiotemporal matched manner and thus fail to mimic the natural processes of biosynthesis, compromising the goal of full cartilage regeneration. Emerging evidence highlights the importance of sequential delivery strategies targeting multiple pathological processes. In this review, we first summarize the current status and progress achieved in single-drug delivery strategies for the treatment of AC diseases. Subsequently, we focus mainly on advances in multiple drug delivery applications, including sequential release formulations targeting various pathological processes, synergistic targeting of the same pathological process, the spatial distribution in multiple tissues, and heterogeneous regeneration. We hope that this review will inspire the rational design of intra-articular drug delivery systems (DDSs) in the future.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianwei Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuangpeng Jiang
- Department of Joint Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Ouyang Z, Dong L, Yao F, Wang K, Chen Y, Li S, Zhou R, Zhao Y, Hu W. Cartilage-Related Collagens in Osteoarthritis and Rheumatoid Arthritis: From Pathogenesis to Therapeutics. Int J Mol Sci 2023; 24:9841. [PMID: 37372989 DOI: 10.3390/ijms24129841] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Collagens serve essential mechanical functions throughout the body, particularly in the connective tissues. In articular cartilage, collagens provide most of the biomechanical properties of the extracellular matrix essential for its function. Collagen plays a very important role in maintaining the mechanical properties of articular cartilage and the stability of the ECM. Noteworthily, many pathogenic factors in the course of osteoarthritis and rheumatoid arthritis, such as mechanical injury, inflammation, and senescence, are involved in the irreversible degradation of collagen, leading to the progressive destruction of cartilage. The degradation of collagen can generate new biochemical markers with the ability to monitor disease progression and facilitate drug development. In addition, collagen can also be used as a biomaterial with excellent properties such as low immunogenicity, biodegradability, biocompatibility, and hydrophilicity. This review not only provides a systematic description of collagen and analyzes the structural characteristics of articular cartilage and the mechanisms of cartilage damage in disease states but also provides a detailed characterization of the biomarkers of collagen production and the role of collagen in cartilage repair, providing ideas and techniques for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ziwei Ouyang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Lei Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Feng Yao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| |
Collapse
|
14
|
Bedell ML, Wang Z, Hogan KJ, Torres AL, Pearce HA, Chim LK, Grande-Allen KJ, Mikos AG. The effect of multi-material architecture on the ex vivo osteochondral integration of bioprinted constructs. Acta Biomater 2023; 155:99-112. [PMID: 36384222 PMCID: PMC9805529 DOI: 10.1016/j.actbio.2022.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Extrusion bioprinted constructs for osteochondral tissue engineering were fabricated to study the effect of multi-material architecture on encapsulated human mesenchymal stem cells' tissue-specific matrix deposition and integration into an ex vivo porcine osteochondral explant model. Two extrusion fiber architecture groups with differing transition regions and degrees of bone- and cartilage-like bioink mixing were employed. The gradient fiber (G-Fib) architecture group showed an increase in chondral integration over time, 18.5 ± 0.7 kPa on Day 21 compared to 9.6 ± 1.6 kPa on Day 1 for the required peak push-out force, and the segmented fiber (S-Fib) architecture group did not, which corresponded to the increase in sulfated glycosaminoglycan deposition noted only in the G-Fib group and the staining for cellularity and tissue-specific matrix deposition at the fiber-defect boundary. Conversely, the S-Fib architecture was associated with significant mineralization over time, but the G-Fib architecture was not. Notably, both fiber groups also had similar chondral integration as a re-inserted osteochondral tissue control. While architecture did dictate differences in the cells' responses to their environment, architecture was not shown to distinguish a statistically significant difference in tissue integration via fiber push-out testing within a given time point or explant region. Use of this three-week osteochondral model demonstrates that these bioink formulations support the fabrication of cell-laden constructs that integrate into explanted tissue as capably as natural tissue and encapsulate osteochondral matrix-producing cells, and it also highlights the important role that spatial architecture plays in the engineering of multi-phasic tissue environments. STATEMENT OF SIGNIFICANCE: Here, an ex vivo model was used to interrogate fundamental questions about the effect of multi-material scaffold architectural choices on osteochondral tissue integration. Cell-encapsulating constructs resembling stratified osteochondral tissue were 3D printed with architecture consisting of either gradient transitions or segmented transitions between the bone-like and cartilage-like bioink regions. The printed constructs were assessed alongside re-inserted natural tissue plugs via mechanical tissue integration push-out testing, biochemical assays, and histology. Differences in osteochondral matrix deposition were observed based on architecture, and both printed groups demonstrated cartilage integration similar to the native tissue plug group. As 3D printing becomes commonplace within biomaterials and tissue engineering, this work illustrates critical 3D co-culture interactions and demonstrates the importance of considering architecture when interpreting the results of studies utilizing spatially complex, multi-material scaffolds.
Collapse
Affiliation(s)
| | - Ziwen Wang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Katie J Hogan
- Department of Bioengineering, Rice University, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | | | - Hannah A Pearce
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Letitia K Chim
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, USA; NIBIB/NIH Center for Engineering Complex Tissues, USA.
| |
Collapse
|
15
|
Szponder T, Latalski M, Danielewicz A, Krać K, Kozera A, Drzewiecka B, Nguyen Ngoc D, Dobko D, Wessely-Szponder J. Osteoarthritis: Pathogenesis, Animal Models, and New Regenerative Therapies. J Clin Med 2022; 12:jcm12010005. [PMID: 36614806 PMCID: PMC9821671 DOI: 10.3390/jcm12010005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive, multifactorial disease resulting in a progressive loss of articular cartilage structure and function that is most common in middle-aged and older patients. OA is involved in the loss of extracellular matrix and cartilage as well as cell number decreases within the matrix, especially in the further stages of the disease. The immune system plays a pivotal role in the pathomechanism of this condition. Both humoral and cellular mediators contribute to cartilage destruction, abnormal bone remodeling, synovitis, and joint effusion. The increasing prevalence of this disease has led to a growing interest in using animal models as the primary way to broaden the knowledge of the pathogenesis of OA and possible therapies at each stage of disease development. This review aims to describe the signs, pathogenesis, and classification of OA as well as discuss the advantages and disadvantages of some animal models. The currently used treatment methods include mesenchymal stem cells, exosomes, gene therapies, and blood-derived products. In addition, exogenous growth factors, platelet-rich plasma (PRP), platelet lysate, and autologous conditioned serum (ACS) are discussed with the application of tissue engineering techniques and biomaterials.
Collapse
Affiliation(s)
- Tomasz Szponder
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences, 20-612 Lublin, Poland
| | - Michał Latalski
- Children’s Orthopaedic Department, Medical University of Lublin, 20-093 Lublin, Poland
| | - Anna Danielewicz
- Children’s Orthopaedic Department, Medical University of Lublin, 20-093 Lublin, Poland
| | - Katarzyna Krać
- Students Research Group of Veterinary Analysts, Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-033 Lublin, Poland
| | - Aleksandra Kozera
- Students Research Group of Veterinary Analysts, Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-033 Lublin, Poland
| | - Beata Drzewiecka
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| | - Dominika Nguyen Ngoc
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| | - Dominika Dobko
- Students Research Group of Veterinary Analysts, Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-033 Lublin, Poland
| | - Joanna Wessely-Szponder
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
- Correspondence:
| |
Collapse
|
16
|
Zhang Y, Su D, Wang Y, Wang Z, Ren Y, Liu R, Du B, Duan R, Shi Y, Liu L, Li X, Zhang Q. Locally delivered modified citrus pectin - a galectin-3 inhibitor shows expected anti-inflammatory and unexpected regeneration-promoting effects on repair of articular cartilage defect. Biomaterials 2022; 291:121870. [DOI: 10.1016/j.biomaterials.2022.121870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/22/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
|
17
|
Peshkova M, Kosheleva N, Shpichka A, Radenska-Lopovok S, Telyshev D, Lychagin A, Li F, Timashev P, Liang XJ. Targeting Inflammation and Regeneration: Scaffolds, Extracellular Vesicles, and Nanotechnologies as Cell-Free Dual-Target Therapeutic Strategies. Int J Mol Sci 2022; 23:13796. [PMID: 36430272 PMCID: PMC9694395 DOI: 10.3390/ijms232213796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Osteoarthritis (OA) affects over 250 million people worldwide and despite various existing treatment strategies still has no cure. It is a multifactorial disease characterized by cartilage loss and low-grade synovial inflammation. Focusing on these two targets together could be the key to developing currently missing disease-modifying OA drugs (DMOADs). This review aims to discuss the latest cell-free techniques applied in cartilage tissue regeneration, since they can provide a more controllable approach to inflammation management than the cell-based ones. Scaffolds, extracellular vesicles, and nanocarriers can be used to suppress inflammation, but they can also act as immunomodulatory agents. This is consistent with the latest tissue engineering paradigm, postulating a moderate, controllable inflammatory reaction to be beneficial for tissue remodeling and successful regeneration.
Collapse
Affiliation(s)
- Maria Peshkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- FSBSI Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Anastasia Shpichka
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Stefka Radenska-Lopovok
- Institute for Clinical Morphology and Digital Pathology, Sechenov University, 119991 Moscow, Russia
| | - Dmitry Telyshev
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Moscow, Russia
- Institute of Bionic Technologies and Engineering, Sechenov University, 119991 Moscow, Russia
| | - Alexey Lychagin
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, 119991 Moscow, Russia
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Xing-Jie Liang
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Gan D, Jiang Y, Hu Y, Wang X, Wang Q, Wang K, Xie C, Han L, Lu X. Mussel-inspired extracellular matrix-mimicking hydrogel scaffold with high cell affinity and immunomodulation ability for growth factor-free cartilage regeneration. J Orthop Translat 2022; 33:120-131. [PMID: 35330942 PMCID: PMC8914478 DOI: 10.1016/j.jot.2022.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Background Injury to articular cartilage cause certain degree of disability due to poor self-repair ability of cartilage tissue. To promote cartilage regeneration, it is essential to develop a scaffold that properly mimics the native cartilage extracellular matrix (ECM) in the aspect of compositions and functions. Methods A mussel-inspired strategy was developed to construct an ECM-mimicking hydrogel scaffold by incorporating polydopamine-modified hyaluronic acid (PDA/HA) complex into a dual-crosslinked collagen (Col) matrix for growth factor-free cartilage regeneration. The adhesion, proliferation, and chondrogenic differentiation of cells on the scaffold were examined. A well-established full-thickness cartilage defect model of the knee in rabbits was used to evaluated the efficacy and functionality of the engineered Col/PDA/HA hydrogel scaffold. Results The PDA/HA complex incorporated-hydrogel scaffold with catechol moieties exhibited better cell affinity than bare negatively-charged HA incorporated hydrogel scaffold. In addition, the PDA/HA complex endowed the scaffold with immunomodulation ability, which suppressed the expression of inflammatory cytokines and effectively activated the polarization of macrophages toward M2 phenotypes. The in vivo results revealed that the mussel-inspired Col/PDA/HA hydrogel scaffold showed strong cartilage inducing ability to promote cartilage regeneration. Conclusions The PDA/HA complex-incorporated hydrogel scaffold overcame the cell repellency of negatively-charged polysaccharide-based scaffolds, which facilitated the adhesion and clustering of cells on the scaffold, and therefore enhanced cell-HA interactions for efficient chondrogenic differentiation. Moreover, the hydrogel scaffold modulated immune microenvironment, and created a regenerative microenvironment to enhance cartilage regeneration. The translational potential of this article This study gives insight into the mussel-inspired approach to construct the tissue-inducing hydrogel scaffold in a growth-factor-free manner, which show great advantage in the clinical treatment. The hydrogel scaffold composed of collagen and hyaluronic acid as the major component, providing cartilage ECM-mimicking environment, is promising for cartilage defect repair.
Collapse
|
19
|
Zhao J, Wu H, Wang L, Jiang D, Wang W, Yuan G, Pei J, Jia W. The beneficial potential of magnesium-based scaffolds to promote chondrogenesis through controlled Mg2+ release in eliminating the destructive effect of activated macrophages on chondrocytes. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 134:112719. [DOI: 10.1016/j.msec.2022.112719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/28/2022] [Accepted: 02/13/2022] [Indexed: 10/19/2022]
|
20
|
Decellularised extracellular matrix-based biomaterials for repair and regeneration of central nervous system. Expert Rev Mol Med 2022; 23:e25. [PMID: 34994341 PMCID: PMC9884794 DOI: 10.1017/erm.2021.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS), consisting of the brain and spinal cord, regulates the mind and functions of the organs. CNS diseases, leading to changes in neurological functions in corresponding sites and causing long-term disability, represent one of the major public health issues with significant clinical and economic burdens worldwide. In particular, the abnormal changes in the extracellular matrix under various disease conditions have been demonstrated as one of the main factors that can alter normal cell function and reduce the neuroregeneration potential in damaged tissue. Decellularised extracellular matrix (dECM)-based biomaterials have been recently utilised for CNS applications, closely mimicking the native tissue. dECM retains tissue-specific components, including proteoglycan as well as structural and functional proteins. Due to their unique composition, these biomaterials can stimulate sensitive repair mechanisms associated with CNS damages. Herein, we discuss the decellularisation of the brain and spinal cord as well as recellularisation of acellular matrix and the recent progress in the utilisation of brain and spinal cord dECM.
Collapse
|
21
|
Shi W, Fang F, Kong Y, Greer SE, Kuss M, Liu B, Xue W, Jiang X, Lovell P, Mohs AM, Dudley AT, Li T, Duan B. Dynamic hyaluronic acid hydrogel with covalent linked gelatin as an anti-oxidative bioink for cartilage tissue engineering. Biofabrication 2021; 14. [PMID: 34905737 DOI: 10.1088/1758-5090/ac42de] [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: 09/15/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022]
Abstract
In the past decade, cartilage tissue engineering has arisen as a promising therapeutic option for degenerative joint diseases, such as osteoarthritis, in the hope of restoring the structure and physiological functions. Hydrogels are promising biomaterials for developing engineered scaffolds for cartilage regeneration. However, hydrogel-delivered mesenchymal stem cells or chondrocytes could be exposed to elevated levels of reactive oxygen species (ROS) in the inflammatory microenvironment after being implanted into injured joints, which may affect their phenotype and normal functions and thereby hinder the regeneration efficacy. To attenuate ROS induced side effects, a multifunctional hydrogel with an innate anti-oxidative ability was produced in this study. The hydrogel was rapidly formed through a dynamic covalent bond between phenylboronic acid grafted hyaluronic acid (HA-PBA) and poly(vinyl alcohol) and was further stabilized through a secondary crosslinking between the acrylate moiety on HA-PBA and the free thiol group from thiolated gelatin. The hydrogel is cyto-compatible and injectable and can be used as a bioink for 3D bioprinting. The viscoelastic properties of the hydrogels could be modulated through the hydrogel precursor concentration. The presence of dynamic covalent linkages contributed to its shear-thinning property and thus good printability of the hydrogel, resulting in the fabrication of a porous grid construct and a meniscus like scaffold at high structural fidelity. The bioprinted hydrogel promoted cell adhesion and chondrogenic differentiation of encapsulated rabbit adipose derived mesenchymal stem cells. Meanwhile, the hydrogel supported robust deposition of extracellular matrix components, including glycosaminoglycans and type II collagen, by embedded mouse chondrocytesin vitro. Most importantly, the hydrogel could protect encapsulated chondrocytes from ROS induced downregulation of cartilage-specific anabolic genes (ACAN and COL2) and upregulation of a catabolic gene (MMP13) after incubation with H2O2. Furthermore, intra-articular injection of the hydrogel in mice revealed adequate stability and good biocompatibilityin vivo. These results demonstrate that this hydrogel can be used as a novel bioink for the generation of 3D bioprinted constructs with anti-ROS ability to potentially enhance cartilage tissue regeneration in a chronic inflammatory and elevated ROS microenvironment.
Collapse
Affiliation(s)
- Wen Shi
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Fang Fang
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | - Yunfan Kong
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Sydney E Greer
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Mitchell Kuss
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Bo Liu
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Wen Xue
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Xiping Jiang
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Paul Lovell
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States of America.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Andrew T Dudley
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Tieshi Li
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States of America.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America.,Department of Mechanical Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America.,Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
22
|
Wang Y, Pei YA, Sun Y, Zhou S, Zhang XB, Pei M. Stem cells immortalized by hTERT perform differently from those immortalized by SV40LT in proliferation, differentiation, and reconstruction of matrix microenvironment. Acta Biomater 2021; 136:184-198. [PMID: 34551328 PMCID: PMC8627502 DOI: 10.1016/j.actbio.2021.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
Although matrix microenvironment has the potential to improve expanded stem cell proliferation and differentiation capacity, decellularized extracellular matrix (dECM) deposited by senescent cells does not contribute to the rejuvenation of adult stem cells, which has become a barrier to personalized stem cell therapy. Genetic modification is an effective strategy to protect cells from senescence but it carries the increased risk of malignant transformation and genetic instability. In this study, lentivirus carrying either human telomerase reverse transcriptase (hTERT) or simian virus 40 large T antigen (SV40LT) was used to transduce human infrapatellar fat pad-derived stem cells (IPFSCs). We found that virus transduction modified the proliferative, chondrogenic, and adipogenic abilities of IPFSCs. Interestingly, dECM deposited by immortalized cells significantly influenced replicative senescent IPFSCs in proliferation and differentiation preference, the effect of which is hinged on the approach of immortalization using either SV40LT or hTERT. Our findings indicate both dECM expansion and immortalization strategies can be used for replicative senescent adult stem cells' proliferation and lineage-specific differentiation, which benefits future stem cell-based tissue regeneration. This approach may also work for adult stem cells with premature senescence in elderly/aged patients, which needs further investigation. STATEMENT OF SIGNIFICANCE: Adult stem cells are a promising solution for autologous cell-based therapy. Unfortunately, cell senescence due to donor age and/or ex vivo expansion prevents clinical application. Recent progress with decellularized extracellular matrix provides a potential for the rejuvenation of senescent stem cells by improving their proliferation and differentiation capacities. Given the fact that the young matrix can provide a healthy and energetic microenvironment, in this study, two approaches using lentivirus transduction of hTERT and SV40LT were compared. The goal was to immortalize donor cells for deposition of decellularized extracellular matrix. The matrix was demonstrated to contribute diverging effects on the chondrogenic and adipogenic differentiation of expanded stem cells and exhibited proliferation benefits as well. These findings provide an invaluable asset for stem cell-based tissue regeneration.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Department of Joint Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Medicine, Loma Linda University, Loma Linda, CA, USA.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
23
|
Filho MCB, Dos Santos Haupenthal DP, Zaccaron RP, de Bem Silveira G, de Roch Casagrande L, Lupselo FS, Alves N, de Sousa Mariano S, do Bomfim FRC, de Andrade TAM, Machado-de-Ávila RA, Silveira PCL. Intra-articular treatment with hyaluronic acid associated with gold nanoparticles in a mechanical osteoarthritis model in Wistar rats. J Orthop Res 2021; 39:2546-2555. [PMID: 33580538 DOI: 10.1002/jor.25008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 11/26/2020] [Accepted: 02/10/2021] [Indexed: 02/04/2023]
Abstract
This study aimed to evaluate the effects of intra-articular treatment with hyaluronic acid (HA) associated with GNPs in a mechanical model of osteoarthritis induced by median meniscectomy (MM). Fifty Wistar rats (2 months weighing between 250 and 300 g) were used, divided into five groups of 10 animals each: Sham, osteoarthritis (OA), OA + HA, OA + gold nanoparticles (GNPs), and OA + HA + GNPs. Intra-articular treatment was started 30 days after the model was induced, with a frequency of 2 weeks for 60 days. Fifteen days after the last application, the animals were euthanized with the removal of the joint tissue for biochemical and histological analysis. The model used was able to mimic osteoarthritis, characterized by the presence of high levels of proinflammatory cytokines, oxidative stress, and degeneration of joint surfaces (Grade III, according to SCORE OARSI). The isolated use of HA or GNPs provided beneficial results to the joint; however, only the group subjected to the association between HA and GNPs showed the attenuation of oxidative stress and reduced proinflammatory markers, with a simultaneous increase in levels of anti-inflammatory cytokines and growth factors. Upon histological analysis, only the OA + HA + GNPs group achieved the restoration of the thickness of the joint cartilage with reduced damage and return to the intact joint surface. The results found demonstrated that the association of GNPs with HA was able to reverse the deleterious effects caused by the model by inhibiting the progressive degeneration of joint surfaces, representing a promising treatment for osteoarthritis.
Collapse
Affiliation(s)
- Mario Cesar Búrigo Filho
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Daniela Pacheco Dos Santos Haupenthal
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Rubya Pereira Zaccaron
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Gustavo de Bem Silveira
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Laura de Roch Casagrande
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Fernando Silva Lupselo
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Naiara Alves
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation, Araras, São Paulo, Brazil
| | - Samara de Sousa Mariano
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation, Araras, São Paulo, Brazil
| | | | | | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| |
Collapse
|
24
|
Foo JB, Looi QH, How CW, Lee SH, Al-Masawa ME, Chong PP, Law JX. Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals (Basel) 2021; 14:1093. [PMID: 34832875 PMCID: PMC8618513 DOI: 10.3390/ph14111093] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are the small extracellular vesicles secreted by cells for intercellular communication. Exosomes are rich in therapeutic cargos such as microRNA (miRNA), long non-coding RNA (lncRNA), small interfering RNA (siRNA), DNA, protein, and lipids. Recently, many studies have focused on miRNAs as a promising therapeutic factor to support cartilage regeneration. Exosomes are known to contain a substantial amount of a variety of miRNAs. miRNAs regulate the post-transcriptional gene expression by base-pairing with the target messenger RNA (mRNA), leading to gene silencing. Several exosomal miRNAs have been found to play a role in cartilage regeneration by promoting chondrocyte proliferation and matrix secretion, reducing scar tissue formation, and subsiding inflammation. The exosomal miRNA cargo can be modulated using techniques such as cell transfection and priming as well as post-secretion modifications to upregulate specific miRNAs to enhance the therapeutic effect. Exosomes are delivered to the joints through direct injection or via encapsulation within a scaffold for sustained release. To date, exosome therapy for cartilage injuries has yet to be optimized as the ideal cell source for exosomes, and the dose and method of delivery have yet to be identified. More importantly, a deeper understanding of the role of exosomal miRNAs in cartilage repair is paramount for the development of more effective exosome therapy.
Collapse
Affiliation(s)
- Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Qi Hao Looi
- My Cytohealth Sdn. Bhd., D353a, Menara Suezcap 1, KL Gateway, no. 2, Jalan Kerinchi, Gerbang Kerinchi Lestari, Kuala Lumpur 59200, Malaysia;
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
| | - Sau Har Lee
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| | - Pei Pei Chong
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
25
|
Jiang Y, Zhang C, Long L, Ge L, Guo J, Fan Z, Yu G. A Comprehensive Analysis of SE-lncRNA/mRNA Differential Expression Profiles During Chondrogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:721205. [PMID: 34589487 PMCID: PMC8475951 DOI: 10.3389/fcell.2021.721205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/12/2021] [Indexed: 01/22/2023] Open
Abstract
Objective: Articular cartilage injury is common and difficult to treat clinically because of the characteristics of the cartilage. Bone marrow-derived mesenchymal stem cell (BMSC)-mediated cartilage regeneration is a promising therapy for treating articular cartilage injury. BMSC differentiation is controlled by numerous molecules and signaling pathways in the microenvironment at both the transcriptional and post-transcriptional levels. However, the possible function of super enhancer long non-coding RNAs (SE-lncRNAs) in the chondrogenic differentiation of BMSCs is still unclear. Our intention was to explore the expression profile of SE-lncRNAs and potential target genes regulated by SE-lncRNAs during chondrogenic differentiation in BMSCs. Materials and Methods: In this study, we conducted a human Super-Enhancer LncRNA Microarray to investigate the differential expression profile of SE-lncRNAs and mRNAs during chondrogenic differentiation of BMSCs. Subsequent bioinformatic analysis was performed to clarify the important signaling pathways, SE-lncRNAs, and mRNAs associated with SE-lncRNAs regulating the chondrogenic differentiation of BMSCs. Results: A total of 77 SE-lncRNAs were identified, of which 47 were upregulated and 30 were downregulated during chondrogenic differentiation. A total of 308 mRNAs were identified, of which 245 were upregulated and 63 were downregulated. Some pathways, such as focal adhesion, extracellular matrix (ECM)–receptor interaction, transforming growth factor-β (TGF-β) signaling pathway, and PI3K–Akt signaling pathway, were identified as the key pathways that may be implicated in the chondrogenic differentiation of BMSCs. Moreover, five potentially core regulatory mRNAs (PMEPA1, ENC1, TES, CDK6, and ADIRF) and 37 SE-lncRNAs in chondrogenic differentiation were identified by bioinformatic analysis. Conclusion: We assessed the differential expression levels of SE-lncRNAs and mRNAs, along with the chondrogenic differentiation of BMSCs. By analyzing the interactions and co-expression, we identified the core SE-lncRNAs and mRNAs acting as regulators of the chondrogenic differentiation potential of BMSCs. Our study also provided novel insights into the mechanism of BMSC chondrogenic and cartilage regeneration.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Stomatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chen Zhang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Lujue Long
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Hunan, China
| | - Lihua Ge
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Jing Guo
- The Key Laboratory of Oral Biomedicine, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Guoxia Yu
- Department of Stomatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
26
|
Kulchar RJ, Denzer BR, Chavre BM, Takegami M, Patterson J. A Review of the Use of Microparticles for Cartilage Tissue Engineering. Int J Mol Sci 2021; 22:10292. [PMID: 34638629 PMCID: PMC8508725 DOI: 10.3390/ijms221910292] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue and organ failure has induced immense economic and healthcare concerns across the world. Tissue engineering is an interdisciplinary biomedical approach which aims to address the issues intrinsic to organ donation by providing an alternative strategy to tissue and organ transplantation. This review is specifically focused on cartilage tissue. Cartilage defects cannot readily regenerate, and thus research into tissue engineering approaches is relevant as a potential treatment option. Cells, scaffolds, and growth factors are three components that can be utilized to regenerate new tissue, and in particular recent advances in microparticle technology have excellent potential to revolutionize cartilage tissue regeneration. First, microspheres can be used for drug delivery by injecting them into the cartilage tissue or joint space to reduce pain and stimulate regeneration. They can also be used as controlled release systems within tissue engineering constructs. Additionally, microcarriers can act as a surface for stem cells or chondrocytes to adhere to and expand, generating large amounts of cells, which are necessary for clinically relevant cell therapies. Finally, a newer application of microparticles is to form them together into granular hydrogels to act as scaffolds for tissue engineering or to use in bioprinting. Tissue engineering has the potential to revolutionize the space of cartilage regeneration, but additional research is needed to allow for clinical translation. Microparticles are a key enabling technology in this regard.
Collapse
Affiliation(s)
- Rachel J. Kulchar
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; (R.J.K.); (B.M.C.)
| | - Bridget R. Denzer
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA;
| | - Bharvi M. Chavre
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; (R.J.K.); (B.M.C.)
| | - Mina Takegami
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA;
| | - Jennifer Patterson
- Independent Consultant, 3000 Leuven, Belgium
- Biomaterials and Regenerative Medicine Group, IMDEA Materials Institute, 28906 Madrid, Spain
| |
Collapse
|
27
|
The potential utility of hybrid photo-crosslinked hydrogels with non-immunogenic component for cartilage repair. NPJ Regen Med 2021; 6:54. [PMID: 34508081 PMCID: PMC8433347 DOI: 10.1038/s41536-021-00166-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Finding a suitable biomaterial for scaffolding in cartilage tissue engineering has proved to be far from trivial. Nonetheless, it is clear that biomimetic approaches based on gelatin (Gel) and hyaluronic acid (HA) have particular promise. Herein, a set of formulations consisting of photo-polymerizable Gel; photo-polymerizable HA, and allogenic decellularized cartilage matrix (DCM), is synthesized and characterized. The novelty of this study lies particularly in the choice of DCM, which was harvested from an abnormal porcine with α-1,3-galactose gene knockout. The hybrid hydrogels were prepared and studied extensively, by spectroscopic methods, for their capacity to imbibe water, for their behavior under compression, and to characterize microstructure. Subsequently, the effects of the hydrogels on contacting cells (in vitro) were studied, i.e., cytotoxicity, morphology, and differentiation through monitoring the specific markers ACAN, Sox9, Coll2, and Col2α1, hypertrophy through monitoring the specific markers alkaline phosphatase (ALP) and Col 10A1. In vivo performance of the hydrogels was assessed in a rat knee cartilage defect model. The new data expand our understanding of hydrogels built of Gel and HA, since they reveal that a significant augmenting role can be played by DCM. The data strongly suggest that further experimentation in larger cartilage-defect animal models is worthwhile and has potential utility for tissue engineering and regenerative medicine.
Collapse
|
28
|
Liu Z, Wang H, Wang S, Gao J, Niu L. PARP-1 inhibition attenuates the inflammatory response in the cartilage of a rat model of osteoarthritis. Bone Joint Res 2021; 10:401-410. [PMID: 34254815 PMCID: PMC8333032 DOI: 10.1302/2046-3758.107.bjr-2020-0200.r2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aims Poly (ADP-ribose) polymerase (PARP) inhibitor has been reported to attenuate inflammatory response in rat models of inflammation. This study was designed to investigate the effect of PARP signalling in osteoarthritis (OA) cartilage inflammatory response in an OA rat model. Methods The OA model was established by anterior cruciate ligament transection with medial meniscectomy in Wistar rats. The poly (ADP-ribose) polymerase 1 (PARP-1) shRNA (short hairpin (sh)-PARP-1) and negative control shRNA (sh-NC) were delivered using a lentiviral vector and were intra-articularly injected into rats after surgery. The weight-bearing distribution of the hind limbs and the knee joint width were measured every two weeks. The expression levels of PARP-1, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in cartilage were determined using real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot. The serum concentrations of inflammatory cytokines were detected using enzyme-linked immunosorbent assay (ELISA). Results PARP-1 expression level significantly increased in the cartilage of the established OA rat model. sh-PARP-1 treatment suppressed PARP-1 levels, decreased the Δ Force (the difference between the weight on ipsilateral limb and contralateral limb) and the knee joint width, inhibited cartilage matrix catabolic enzymes, and ameliorated OA cartilage degradation and attenuated inflammatory response. Conclusion PARP-1 inhibition attenuates OA cartilage inflammatory response in the OA rat model. Cite this article: Bone Joint Res 2021;10(7):401–410.
Collapse
Affiliation(s)
- Zili Liu
- Department of Microscopic Orthopedics, The Hefei Second People's Hospital & Hefei Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Honglin Wang
- Department of Microscopic Orthopedics, The Hefei Second People's Hospital & Hefei Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shaoqian Wang
- Department of Microscopic Orthopedics, The Hefei Second People's Hospital & Hefei Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jie Gao
- Department of Microscopic Orthopedics, The Hefei Second People's Hospital & Hefei Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Niu
- Department of Microscopic Orthopedics, The Hefei Second People's Hospital & Hefei Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
An artificial membrane binding protein-polymer surfactant nanocomplex facilitates stem cell adhesion to the cartilage extracellular matrix. Biomaterials 2021; 276:120996. [PMID: 34280823 DOI: 10.1016/j.biomaterials.2021.120996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/12/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
One of the major challenges within the emerging field of injectable stem cell therapies for articular cartilage (AC) repair is the retention of sufficient viable cell numbers at the site of injury. Even when delivered via intra-articular injection, the number of stem cells retained at the target is often low and declines rapidly over time. To address this challenge, an artificial plasma membrane binding nanocomplex was rationally designed to provide human mesenchymal stem cells (hMSCs) with increased adhesion to articular cartilage tissue. The nanocomplex comprises the extracellular matrix (ECM) binding peptide of a placenta growth factor-2 (PlGF-2) fused to a supercharged green fluorescent protein (scGFP), which was electrostatically conjugated to anionic polymer surfactant chains to yield [S-]scGFP_PlGF2. The [S-]scGFP_PlGF2 nanocomplex spontaneously inserts into the plasma membrane of hMSCs, is not cytotoxic, and does not inhibit differentiation. The nanocomplex-modified hMSCs showed a significant increase in affinity for immobilised collagen II, a key ECM protein of cartilage, in both static and dynamic cell adhesion assays. Moreover, the cells adhered strongly to bovine ex vivo articular cartilage explants resulting in high cell numbers. These findings suggest that the re-engineering of hMSC membranes with [S-]scGFP_PlGF2 could improve the efficacy of injectable stem cell-based therapies for the treatment of damaged articular cartilage.
Collapse
|
30
|
Tampieri A, Sandri M, Iafisco M, Panseri S, Montesi M, Adamiano A, Dapporto M, Campodoni E, Dozio SM, Degli Esposti L, Sprio S. Nanotechnological approach and bio-inspired materials to face degenerative diseases in aging. Aging Clin Exp Res 2021; 33:805-821. [PMID: 31595428 DOI: 10.1007/s40520-019-01365-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 09/21/2019] [Indexed: 12/22/2022]
Abstract
The aging of the world population is increasingly claimed as an alarming situation, since an ever-raising number of persons in advanced age but still physically active is expected to suffer from invalidating and degenerative diseases. The impairment of the endogenous healing potential provoked by the aging requires the development of more effective and personalized therapies, based on new biomaterials and devices able to direct the cell fate to stimulate and sustain the regrowth of damaged or diseased tissues. To obtain satisfactory results, also in cases where the cell senescence, typical of the elderly, makes the regeneration process harder and longer, the new solutions have to possess excellent ability to mimic the physiological extracellular environment and thus exert biomimetic stimuli on stem cells. To this purpose, the "biomimetic concept" is today recognized as elective to fabricate bioactive and bioresorbable devices such as hybrid osteochondral scaffolds and bioactive bone cements closely resembling the natural hard tissues and with enhanced regenerative ability. The review will illustrate some recent results related to these new biomimetic materials developed for application in different districts of the musculoskeletal system, namely bony, osteochondral and periodontal regions, and the spine. Further, it will be shown how new bioactive and superparamagnetic calcium phosphate nanoparticles can give enhanced results in cardiac regeneration and cancer therapy. Since tissue regeneration will be a major demand in the incoming decades, the high potential of biomimetic materials and devices is promising to significantly increase the healing rate and improve the clinical outcomes even in aged patients.
Collapse
Affiliation(s)
- Anna Tampieri
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Monica Sandri
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Monica Montesi
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Alessio Adamiano
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Massimiliano Dapporto
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Elisabetta Campodoni
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Samuele M Dozio
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Lorenzo Degli Esposti
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Simone Sprio
- Institute of Science and Technology for Ceramics, National Research Council, Via Granarolo 64, 48018, Faenza, RA, Italy.
| |
Collapse
|
31
|
Sumayya AS, Muraleedhara Kurup G. In vitro anti-inflammatory potential of marine macromolecules cross-linked bio-composite scaffold on LPS stimulated RAW 264.7 macrophage cells for cartilage tissue engineering applications. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1040-1056. [PMID: 33682617 DOI: 10.1080/09205063.2021.1899590] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Biomaterials serve as an integral component of tissue engineering. They are designed to provide architectural framework of native extracellular matrix so as to encourage cell growth and eventual tissue regeneration. Naturally occurring biopolymers as scaffolds offer options for cartilage tissue engineering due to anti-inflammatory, biocompatibility, biodegradability, low toxicity of degradation by-products and plasticity in processing into a variety of material formats. Here we studied in vitro anti-inflammatory potential of marine macromolecules cross-linked bio-composite scaffold composed of hydroxyapatite, alginate, chitosan and fucoidan named as HACF on LPS stimulated RAW 264.7 macrophage cells. The effects of HACF on the viability of RAW264.7 cells, nitrite level, intracellular ROS as well as the mRNA levels of NF-κB, iNOS, COX-2, TNF-α, IL-1β and IL-6 were examined in LPS induced RAW264.7 macrophage cells. The results revealed that HACF hydrogel scaffold exerts anti-inflammatory effect by inhibiting the production of ROS, suppress NF-kB translocation to the nucleus and thereby inhibiting the production of inflammatory mediators. Hence, our results confirm that HACF has a strong anti-oxidant capacity to inhibit inflammation associated gene expression by suppressing NF-kB signaling pathway. It clearly reveals the anti-oxidant and anti-inflammatory effect of HACF hydrogel scaffold on LPS induced RAW 264.7 cells.
Collapse
Affiliation(s)
- A S Sumayya
- Faculty, Department of Biochemistry, T.K.M. College of Arts and Science, Kollam, India
| | | |
Collapse
|
32
|
Meng X, Grad S, Wen C, Lai Y, Alini M, Qin L, Wang X. An impaired healing model of osteochondral defect in papain-induced arthritis. J Orthop Translat 2020; 26:101-110. [PMID: 33437629 PMCID: PMC7773975 DOI: 10.1016/j.jot.2020.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022] Open
Abstract
Background Osteochondral defects (OCD) are common in osteoarthritis (OA) and difficult to heal. Numerous tissue engineering approaches and novel biomaterials are developed to solve this challenging condition. Although most of the novel methods can successfully treat osteochondral defects in preclinical trials, their clinical application in OA patients is not satisfactory, due to a high spontaneous recovery rate of many preclinical animal models by ignoring the inflammatory environment. In this study, we developed a sustained osteochondral defect model in osteoarthritic rabbits and compared the cartilage and subchondral bone regeneration in normal and arthritic environments. Methods Rabbits were injected with papain (1.25%) in the right knee joints (OA group), and saline in the left knee joints (Non-OA group) at day 1 and day 3. One week later a cylindrical osteochondral defect of 3.2 mm in diameter and 3 mm depth was made in the femoral patellar groove. After 16 weeks, newly regenerated cartilage and bone inside the defect were evaluated by micro-CT, histomorphology and immunohistochemistry. Results One week after papain injection, extracellular matrix in the OA group demonstrated dramatically less safranin O staining intensity than in the non-OA group. Until 13 weeks of post-surgery, knee width remained significantly higher in the OA group than the non-OA control group. Sixteen weeks after surgery, the OA group had 11.3% lower International Cartilage Regeneration and Joint Preservation Society score and 32.5% lower O’Driscoll score than the non-OA group. There were less sulfated glycosaminoglycan and type II collagen but 74.1% more MMP-3 protein in the regenerated cartilage of the OA group compared with the non-OA group. As to the regenerated bone, bone volume fraction, trabecular thickness and trabecular number were all about 28% lower, while the bone mineral density was 26.7% higher in the OA group compared to the non-OA group. Dynamic histomorphometry parameters including percent labeled perimeter, mineral apposition rate and bone formation rate were lower in the OA group than in the non-OA group. Immunohistochemistry data showed that the OA group had 15.9% less type I collagen than the non-OA group. Conclusion The present study successfully established a non-self-healing osteochondral defect rabbit model in papain-induced OA, which was well simulating the clinical feature and pathology. In addition, we confirmed that both cartilage and subchondral bone regeneration were further impaired in arthritic environment. The translational potential of this article The present study provides an osteochondral defect in a small osteoarthritic model. This non-self-healing model and the evaluation protocol could be used to evaluate the efficacy and study the mechanism of newly developed biomaterials or tissue engineering methods preclinically; as methods tested in reliable preclinical models are expected to achieve improved success rate when tested clinically for treatment of OCD in OA patients.
Collapse
Affiliation(s)
- Xiangbo Meng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,CAS-HK Joint Lab of Biomaterials, Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, Translational Medicine Research and Development Center, Shenzhen Institutes of Advanced Technology of Chinese Academy of Sciences and the Chinese University of Hong Kong, China.,Shenzhen Engineering Research Center for Medical Bioactive Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sibylle Grad
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - Chunyi Wen
- Interdisciplinary Division of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Yuxiao Lai
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,CAS-HK Joint Lab of Biomaterials, Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, Translational Medicine Research and Development Center, Shenzhen Institutes of Advanced Technology of Chinese Academy of Sciences and the Chinese University of Hong Kong, China.,Shenzhen Engineering Research Center for Medical Bioactive Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.,CAS-HK Joint Lab of Biomaterials, Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, Translational Medicine Research and Development Center, Shenzhen Institutes of Advanced Technology of Chinese Academy of Sciences and the Chinese University of Hong Kong, China
| | - Xinluan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.,CAS-HK Joint Lab of Biomaterials, Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, Translational Medicine Research and Development Center, Shenzhen Institutes of Advanced Technology of Chinese Academy of Sciences and the Chinese University of Hong Kong, China.,Shenzhen Engineering Research Center for Medical Bioactive Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
33
|
Mendes C, Dos Santos Haupenthal DP, Zaccaron RP, de Bem Silveira G, Corrêa MEAB, de Roch Casagrande L, de Sousa Mariano S, de Souza Silva JI, de Andrade TAM, Feuser PE, Machado-de-Ávila RA, Silveira PCL. Effects of the Association between Photobiomodulation and Hyaluronic Acid Linked Gold Nanoparticles in Wound Healing. ACS Biomater Sci Eng 2020; 6:5132-5144. [PMID: 33455264 DOI: 10.1021/acsbiomaterials.0c00294] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Healing is the process responsible for restoring the integrity of the body's internal or external structures when they rupture. Photobiomodulation (PBM) stands out as one of the most efficient resources in the treatment of epithelial lesions, as well as hyaluronic acid (HA), which has been emerging as a new molecule for the treatment of dermal and epidermal lesions. The biological application of gold nanoparticles (GNPs) shows promising results. This study aimed to investigate the possible anti-inflammatory and antioxidant effects of the association between PBM and GNPs-linked HA in an epithelial lesion model. Fifty Wistar rats were randomly distributed in the Control Group (CG); (PBM); (PBM + HA); (PBM + GNPs); (PBM + GNPs-HA). The animals were anesthetized, trichotomized, and induced to a surgical incision in the dorsal region. Topical treatment with HA (0.9%) and/or GNPs (30 mg/kg) occurred daily associated with 904 nm laser irradiation, dose of 5 J/cm2, which started 24 h after the lesion and was performed daily until the seventh day. The levels of proinflammatory (IL1 and TNFα), anti-inflammatory (IL10 and IL4) and growth factors (FGF and TGFβ) cytokines and oxidative stress parameters were evaluated, besides histological analysis through inflammatory infiltrate, fibroblasts, new vessels, and collagen production area. Finally, for the analysis of wound size reduction, digital images were performed and subsequently analyzed by the IMAGEJ software. The treated groups showed a decrease in proinflammatory cytokine levels and an increase in anti-inflammatory cytokines. TGFβ and FGF levels also increased in the treated groups, especially in the combination therapy group (PBM + GNPs-HA). Regarding the oxidative stress parameters, MPO, DCF, and Nitrite levels decreased in the treated groups, as well as the oxidative damage (Carbonyl and Thiol groups). In contrast, antioxidant defense increased in the groups with the appropriate therapies proposed compared to the control group. Histological sections were analyzed where the inflammatory infiltrate was lower in the PBM + GNPs-HA group. The number of fibroblasts was higher in the PBM and PBM + HA treated groups, whereas collagen production was higher in all treated groups. Finally, in the analysis of the wound area contraction, the injury group presented a larger area in cm2 compared to the other groups. Taken together, these results allow us to observe that the combination of PBM + GNPs-HA optimized the secretion of anti-inflammatory cytokines, proliferation and cell differentiation growth factors, and made an earlier transition to the chronic phase, contributing to the repair process.
Collapse
Affiliation(s)
- Carolini Mendes
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Daniela Pacheco Dos Santos Haupenthal
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Rubya Pereira Zaccaron
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Gustavo de Bem Silveira
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Maria Eduarda Anastácio Borges Corrêa
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Laura de Roch Casagrande
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Samara de Sousa Mariano
- Graduate Program of Biomedical Science, University Center of Herminio Ometto Foundation, 13607-339 Araras São Paolo Brazil
| | - Jennyffer Ione de Souza Silva
- Graduate Program of Biomedical Science, University Center of Herminio Ometto Foundation, 13607-339 Araras São Paolo Brazil
| | | | - Paulo Emilio Feuser
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, Santa Catarina Brazil
| |
Collapse
|
34
|
Tran HD, Park KD, Ching YC, Huynh C, Nguyen DH. A comprehensive review on polymeric hydrogel and its composite: Matrices of choice for bone and cartilage tissue engineering. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Zheng L, Wang Y, Qiu P, Xia C, Fang Y, Mei S, Fang C, Shi Y, Wu K, Chen Z, Fan S, He D, Lin X, Chen P. Primary chondrocyte exosomes mediate osteoarthritis progression by regulating mitochondrion and immune reactivity. Nanomedicine (Lond) 2020; 14:3193-3212. [PMID: 31855117 DOI: 10.2217/nnm-2018-0498] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We aimed to investigate the proteomics of primary chondrocyte exosomes and the effect of exosomes in osteoarthritis (OA) treatment. Materials & methods: We isolated exosomes from primary chondrocytes cultured in normal (D0) and inflammatory environments induced by IL-1β and determined the proteomics of these exosomes. Next, we investigated what effect and mechanism D0 chondrocytes exosomes have in OA treatment. Results: There were more proteins that belonged to mitochondrion and were involved in immune system processes in D0 exosomes. Notably, intra-articular administration of D0 exosomes successfully prevented the development of OA. D0 chondrocyte exosomes could restore mitochondrial dysfunction and polarize macrophage response toward an M2 phenotype. Conclusion: Our findings demonstrated that primary chondrocyte exosomes are efficient in OA treatment.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China.,Department of Orthopedics, 5th Affiliated Hospital, Lishui Municipal Central Hospital, Wenzhou Medical University, Lishui, PR China
| | - Yiyun Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Pengcheng Qiu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Chen Xia
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Yifan Fang
- Hangzhou Foreign Languages School, Hangzhou, PR China
| | - Sheng Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Chen Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Yiling Shi
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Kaiwei Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Zhijun Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Dengwei He
- Department of Orthopedics, 5th Affiliated Hospital, Lishui Municipal Central Hospital, Wenzhou Medical University, Lishui, PR China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| |
Collapse
|
36
|
Câmara DAD, Shibli JA, Müller EA, De-Sá-Junior PL, Porcacchia AS, Blay A, Lizier NF. Adipose Tissue-Derived Stem Cells: The Biologic Basis and Future Directions for Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3210. [PMID: 32708508 PMCID: PMC7420246 DOI: 10.3390/ma13143210] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 01/12/2023]
Abstract
Mesenchymal stem cells (MSCs) have been isolated from a variety of tissues using different methods. Active research have confirmed that the most accessible site to collect them is the adipose tissue; which has a significantly higher concentration of MSCs. Moreover; harvesting from adipose tissue is less invasive; there are no ethical limitations and a lower risk of severe complications. These adipose-derived stem cells (ASCs) are also able to increase at higher rates and showing telomerase activity, which acts by maintaining the DNA stability during cell divisions. Adipose-derived stem cells secret molecules that show important function in other cells vitality and mechanisms associated with the immune system, central nervous system, the heart and several muscles. They release cytokines involved in pro/anti-inflammatory, angiogenic and hematopoietic processes. Adipose-derived stem cells also have immunosuppressive properties and have been reported to be "immune privileged" since they show negative or low expression of human leukocyte antigens. Translational medicine and basic research projects can take advantage of bioprinting. This technology allows precise control for both scaffolds and cells. The properties of cell adhesion, migration, maturation, proliferation, mimicry of cell microenvironment, and differentiation should be promoted by the printed biomaterial used in tissue engineering. Self-renewal and potency are presented by MSCs, which implies in an open-source for 3D bioprinting and regenerative medicine. Considering these features and necessities, ASCs can be applied in the designing of tissue engineering products. Understanding the heterogeneity of ASCs and optimizing their properties can contribute to making the best therapeutic use of these cells and opening new paths to make tissue engineering even more useful.
Collapse
Affiliation(s)
| | - Jamil Awad Shibli
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., Jundiaí 13212-213, Brazil;
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07040-170, Brazil;
| | - Eduardo Alexandre Müller
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07040-170, Brazil;
| | | | - Allan Saj Porcacchia
- Department of Psychobiology, Federal University of São Paulo, São Paulo 04021-001, Brazil;
| | - Alberto Blay
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., Jundiaí 13212-213, Brazil;
| | - Nelson Foresto Lizier
- Nicell-Pesquisa e Desenvolvimento Científico LTDA, São Paulo 04006-000, Brazil;
- Department of Psychobiology, Federal University of São Paulo, São Paulo 04021-001, Brazil;
| |
Collapse
|
37
|
Helgeland E, Pedersen TO, Rashad A, Johannessen AC, Mustafa K, Rosén A. Angiostatin-functionalized collagen scaffolds suppress angiogenesis but do not induce chondrogenesis by mesenchymal stromal cells in vivo. J Oral Sci 2020; 62:371-376. [PMID: 32684573 DOI: 10.2334/josnusd.19-0327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Tissue engineering for fibrocartilage regeneration using mesenchymal stromal cells (MSC) and biomaterial scaffolds is emerging as a promising strategy, but inhibiting vascularization to prevent endochondral ossification is important to develop stable implants. The objective of this study was to investigate the effect of angiostatin on inhibition of angiogenesis and promotion of chondrogenesis by collagen scaffolds with or without MSC implanted subcutaneously in rats. One scaffold from the following groups was implanted in each animal: Collagen scaffolds only, scaffolds functionalized with angiostatin, scaffolds loaded with MSC and scaffolds functionalized with angiostatin and loaded with MSC. The various scaffolds were harvested after 2 and 8 weeks for histological analysis, Real-time quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence quantification. Results demonstrated significantly decreased expression of inflammatory (interleukin 1 alpha and beta) and angiogenic genes (platelet and endothelial cell adhesion molecule 1) in scaffolds functionalized with angiostatin after 2 weeks in vivo. Histologically, after 8 weeks, the scaffolds with angiostatin had less inflammatory cells and more collagen matrix formation, but no fibrocartilage formation was detected. Thus, although angiostatin suppressed angiogenesis, it did not stimulate ectopic chondrogenesis in tissue engineered constructs in vivo.
Collapse
Affiliation(s)
| | - Torbjørn O Pedersen
- Department of Clinical Dentistry, University of Bergen.,Department of Oral and Maxillofacial Surgery, University of Bergen and Haukeland University Hospital
| | - Ahmad Rashad
- Department of Clinical Dentistry, University of Bergen
| | - Anne C Johannessen
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen.,Department of Pathology, Haukeland University Hospital
| | - Kamal Mustafa
- Department of Clinical Dentistry, University of Bergen
| | - Annika Rosén
- Department of Clinical Dentistry, University of Bergen.,Department of Oral and Maxillofacial Surgery, University of Bergen and Haukeland University Hospital
| |
Collapse
|
38
|
Asgari N, Bagheri F, Eslaminejad MB, Ghanian MH, Sayahpour FA, Ghafari AM. Dual functional construct containing kartogenin releasing microtissues and curcumin for cartilage regeneration. Stem Cell Res Ther 2020; 11:289. [PMID: 32678019 PMCID: PMC7367357 DOI: 10.1186/s13287-020-01797-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/15/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Regeneration of articular cartilage poses a tremendous challenge due to its limited self-repair capability and inflammation at the damaged site. To generate the desired structures that mimic the structure of native tissue, microtissues with repeated functional units such as cell aggregates have been developed. Multicellular aggregates of mesenchymal stem cells (MSCs) can be used as microscale building blocks of cartilage due to their potential for cell-cell contact, cell proliferation, and differentiation. METHODS Chondrogenic microtissues were developed through incorporation of kartogenin-releasing poly (lactic-co-glycolic acid) (PLGA) microparticles (KGN-MP) within the MSC aggregates. The chondrogenic potential of KGN-MP treated MSC aggregates was proven in vitro by studying the chondrogenic markers at the RNA level and histological analysis. In order to address the inflammatory responses at the defect site, the microtissues were delivered in vivo via an injectable, anti-inflammatory hydrogel that contained gelatin methacryloyl (GelMA) loaded with curcumin (Cur). RESULTS The KGN-MPs were fabricated to support MSCs during cartilage differentiation. According to real-time RT-PCR analysis, the presence of KGN in the aggregates led to the expression of cartilage markers by the MSCs. Both toluidine blue (TB) and safranin O (SO) staining demonstrated homogeneous glycosaminoglycan production throughout the KGN-MP incorporated MSC aggregates. The curcumin treatment efficiently reduced the expressions of hypertrophy markers by MSCs in vitro. The in vivo results showed that implantation of chondrogenic microtissues (KGN-MP incorporated MSC aggregates) using the curcumin loaded GelMA hydrogel resulted in cartilage tissue regeneration that had characteristic features close to the natural hyaline cartilage according to observational and histological results. CONCLUSIONS The use of this novel construct that contained chondrogenic cell blocks and curcumin is highly desired for cartilage regeneration.
Collapse
Affiliation(s)
- Negin Asgari
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Jalal Ale Ahmad Street, P.O.Box: 14115-111, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat Highway, P.O. Box 16635-148, Tehran, Iran.
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forogh Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat Highway, P.O. Box 16635-148, Tehran, Iran
| | - Amir Mohammad Ghafari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Center for Functional Materials, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| |
Collapse
|
39
|
Park E, Hart ML, Rolauffs B, Stegemann JP, T Annamalai R. Bioresponsive microspheres for on-demand delivery of anti-inflammatory cytokines for articular cartilage repair. J Biomed Mater Res A 2019; 108:722-733. [PMID: 31788947 DOI: 10.1002/jbm.a.36852] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022]
Abstract
Despite innovations in surgical interventions, treatment of cartilage injury in osteoarthritic joints remains a challenge due to concomitant inflammation. Obstructing a single dominant inflammatory cytokine has shown remarkable clinical benefits in rheumatoid arthritis, and similar strategies are being suggested to target inflammatory pathways in osteoarthritis (OA). Here, we describe the utility of gelatin microspheres that are responsive to proteolytic enzymes typically expressed in arthritic flares, resulting in on-demand and spatiotemporally controlled release of anti-inflammatory cytokines for cartilage preservation and repair. These microspheres were designed with a net negative charge to sequester cationic anti-inflammatory cytokines, and the magnitude of the negative charge potential increased with an increase in crosslinking density. Collagenase-mediated degradation of the microspheres was dependent on the concentration of the enzyme. Release of anti-inflammatory cytokines from the loaded microspheres directly correlated with the degradation of the gelatin matrix. Exposure of the IL-4 and IL-13 loaded microspheres reduced the inflammation of chondrocytes up to 80%. Hence, the delivery of these microspheres in an OA joint can attenuate the stimulation of chondrocytes and the resulting secretion of catabolic factors such as proteinases and nitric oxide. The microsphere format also allows for minimally invasive delivery and is less susceptible to mechanically induced drug release. Consequently, bioresponsive microspheres can be an effective tool for cartilage preservation and arthritis treatment.
Collapse
Affiliation(s)
- Eunjae Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Melanie L Hart
- Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Bernd Rolauffs
- Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Ramkumar T Annamalai
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
40
|
Li X, Guo W, Zha K, Jing X, Wang M, Zhang Y, Hao C, Gao S, Chen M, Yuan Z, Wang Z, Zhang X, Shen S, Li H, Zhang B, Xian H, Zhang Y, Sui X, Qin L, Peng J, Liu S, Lu S, Guo Q. Enrichment of CD146 + Adipose-Derived Stem Cells in Combination with Articular Cartilage Extracellular Matrix Scaffold Promotes Cartilage Regeneration. Theranostics 2019; 9:5105-5121. [PMID: 31410204 PMCID: PMC6691381 DOI: 10.7150/thno.33904] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Heterogeneity of mesenchymal stem cells (MSCs) influences the cell therapy outcome and the application in tissue engineering. Also, the application of subpopulations of MSCs in cartilage regeneration remains poorly characterized. CD146+ MSCs are identified as the natural ancestors of MSCs and the expression of CD146 are indicative of greater pluripotency and self-renewal potential. Here, we sorted a CD146+ subpopulation from adipose-derived mesenchymal stem cells (ADSCs) for cartilage regeneration. Methods: CD146+ ADSCs were sorted using magnetic activated cell sorting (MACS). Cell surface markers, viability, apoptosis and proliferation were evaluated in vitro. The molecular signatures were analyzed by mRNA and protein expression profiling. By intra-articular injections of cells in a rat osteochondral defect model, we assessed the role of the specific subpopulation in cartilage microenvironment. Finally, CD146+ ADSCs were combined with articular cartilage extracellular matrix (ACECM) scaffold for long term (3, 6 months) cartilage repair. Results: The enriched CD146+ ADSCs showed a high expression of stem cell and pericyte markers, good viability, and immune characteristics to avoid allogeneic rejection. Gene and protein expression profiles revealed that the CD146+ ADSCs had different cellular functions especially in regulation inflammation. In a rat model, CD146+ ADSCs showed a better inflammation-modulating property in the early stage of intra-articular injections. Importantly, CD146+ ADSCs exhibited good biocompatibility with the ACECM scaffold and the CD146+ cell-scaffold composites produced less subcutaneous inflammation. The combination of CD146+ ADSCs with ACECM scaffold can promote better cartilage regeneration in the long term. Conclusion: Our data elucidated the function of the CD146+ ADSC subpopulation, established their role in promoting cartilage repair, and highlighted the significance of cell subpopulations as a novel therapeutic for cartilage regeneration.
Collapse
Affiliation(s)
- Xu Li
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Weimin Guo
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kangkang Zha
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaoguang Jing
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Mingjie Wang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yu Zhang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Chunxiang Hao
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuang Gao
- Center for Biomedical Material and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Mingxue Chen
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Zhiguo Yuan
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Zhenyong Wang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xueliang Zhang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shi Shen
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Haojiang Li
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Bin Zhang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Hai Xian
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yuan Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiang Sui
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shibi Lu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopaedics; Key Laboratory of Musculoskeletal Trauma & War Injuries,PLA; 28 Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
41
|
Ma F, Ge Y, Liu N, Pang X, Shen X, Tang B. In situ fabrication of a composite hydrogel with tunable mechanical properties for cartilage tissue engineering. J Mater Chem B 2019; 7:2463-2473. [DOI: 10.1039/c8tb01331d] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A composite hydrogel with tunable mechanical properties has been fabricated and characterized in this study.
Collapse
Affiliation(s)
- Fenbo Ma
- Department of Biomedical Engineering
- Southern University of Science and Technology
- Shenzhen
- P. R. China
| | - Yongmei Ge
- Department of Biomedical Engineering
- Southern University of Science and Technology
- Shenzhen
- P. R. China
| | - Nian Liu
- Department of Biomedical Engineering
- Southern University of Science and Technology
- Shenzhen
- P. R. China
| | - Xiangchao Pang
- Department of Biomedical Engineering
- Southern University of Science and Technology
- Shenzhen
- P. R. China
- College of Materials Science and Engineering
| | - Xingyu Shen
- Department of Biomedical Engineering
- Southern University of Science and Technology
- Shenzhen
- P. R. China
| | - Bin Tang
- Department of Biomedical Engineering
- Southern University of Science and Technology
- Shenzhen
- P. R. China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research
| |
Collapse
|
42
|
Lolli A, Colella F, De Bari C, van Osch GJVM. Targeting anti-chondrogenic factors for the stimulation of chondrogenesis: A new paradigm in cartilage repair. J Orthop Res 2019; 37:12-22. [PMID: 30175861 DOI: 10.1002/jor.24136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/09/2018] [Indexed: 02/04/2023]
Abstract
Trauma and age-related cartilage disorders represent a major global cause of morbidity, resulting in chronic pain and disability in patients. A lack of effective therapies, together with a rapidly aging population, creates an impressive clinical and economic burden on healthcare systems. In this scenario, experimental therapies based on transplantation or in situ stimulation of skeletal Mesenchymal Stem/progenitor Cells (MSCs) have raised great interest for cartilage repair. Nevertheless, the challenge of guiding MSC differentiation and preventing cartilage hypertrophy and calcification still needs to be overcome. While research has mostly focused on the stimulation of cartilage anabolism using growth factors, several issues remain unresolved prompting the field to search for novel solutions. Recently, inhibition of anti-chondrogenic regulators has emerged as an intriguing opportunity. Anti-chondrogenic regulators include extracellular proteins as well as intracellular transcription factors and microRNAs that act as potent inhibitors of pro-chondrogenic signals. Suppression of these inhibitors can enhance MSC chondrogenesis and production of cartilage matrix. We here review the current knowledge concerning different types of anti-chondrogenic regulators. We aim to highlight novel therapeutic targets for cartilage repair and discuss suitable tools for suppressing their anti-chondrogenic functions. Further effort is needed to unveil the therapeutic perspectives of this approach and pave the way for effective treatment of cartilage injuries in patients. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands
| | - Fabio Colella
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands.,Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
43
|
Dai M, Liu X, Wang N, Sun J. Squid type II collagen as a novel biomaterial: Isolation, characterization, immunogenicity and relieving effect on degenerative osteoarthritis via inhibiting STAT1 signaling in pro-inflammatory macrophages. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 89:283-294. [PMID: 29752100 DOI: 10.1016/j.msec.2018.04.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/21/2017] [Accepted: 04/10/2018] [Indexed: 12/22/2022]
Abstract
Collagen from marine organisms has a broad prospect in biomedical field, yet the knowledge on marine-derived type II collagen is rare. Herein, a novel type II collagen was successfully isolated from squid cartilage for the first time. After being characterized, the immunogenicity of squid type II collagen (SCII) was evaluated and compared with that of bovine type II collagen (BCII). Then investigations were further conducted for the impacts of SCII on pro-inflammatory macrophages and macrophage chemotaxis. The degenerative osteoarthritis (OA) -relieving effects of SCII were explored using OA rat model in vivo. Our results demonstrated that the isolated SCII maintained triple-superhelical structure of native collagen with high purity. Different from BCII, SCII presented no immunogenicity since it neither induced abnormal proliferation of lymphocytes in vitro nor changed the basic levels of IgM, IgG, anti-type II collagen IgG and CD4+/CD8+ lymphocytes ratio in vivo. Additionally, SCII also exerted prominent anti-inflammatory effects. SCII significantly reduced the production of pro-inflammatory cytokines by enhancing the activity of TCPTP and subsequently prompting the dephosphorylation of p-STAT1 in pro-inflammatory macrophages. Besides, it indirectly prevented hypertrophic changes of chondrocytes, and markedly impeded chemotaxis of macrophages. Moreover, inflammation condition in OA rats was significantly alleviated under treatment with SCII. These data suggested that the newly developed SCII could not only avoid the immunogenic risks of collagen derived from terrestrial animals, but more importantly, provide new choice for the control and treatment of OA.
Collapse
Affiliation(s)
- Meilu Dai
- Shanghai Biomaterials Research & Testing Center, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200023, China
| | - Xin Liu
- Shanghai Biomaterials Research & Testing Center, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200023, China
| | - Nanping Wang
- Shanghai Fisheries Research Institute, Shanghai 200433, China
| | - Jiao Sun
- Shanghai Biomaterials Research & Testing Center, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200023, China.
| |
Collapse
|
44
|
Hassan G, Bahjat M, Kasem I, Soukkarieh C, Aljamali M. Platelet lysate induces chondrogenic differentiation of umbilical cord-derived mesenchymal stem cells. Cell Mol Biol Lett 2018; 23:11. [PMID: 29568314 PMCID: PMC5859745 DOI: 10.1186/s11658-018-0080-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Purpose Articular cartilage has a poor capacity for self-repair, and thus still presents a major challenge in orthopedics. Mesenchymal stem cells (MSCs) are multipotent stem cells with the potential to differentiate into chondrocytes in the presence of transforming growth factor beta (TGF-β). Platelet lysate (PL) contains a relatively large number of growth factors, including TGF-β, and has been shown to ameliorate cartilage repair. Here, we investigated the ability of PL to direct chondrogenic differentiation of MSCs along with other standard differentiation components in a pellet culture system. Methods We isolated and expanded MSCs from human umbilical cords using a PL-supplemented medium and characterized the cells based on immunophenotype and potential for differentiation to adipocytes and osteocytes. We further cultured MSCs as pellets in a chondrogenic-differentiation medium supplemented with PL. After 21 days, the pellets were processed for histological analysis and stained with alician blue and acridine orange. The expression of SOX9 was investigated using RT-PCR. Results MSCs maintained their stemness characteristics in the PL-supplemented medium. However, the distribution of cells in the pellets cultured in the PL-supplemented chondrogenic differentiation medium had a greater similarity to cartilage tissue-derived chondrocytes than to the negative control. The intense alician blue staining indicated an increased production of mucopolysaccharides in the differentiated pellets, which also showed elevated expression of SOX9. Conclusions Our data suggest that MSCs could be differentiated to chondrocytes in the presence of PL and absence of exogenous TGF-β. Further research needs to be conducted to understand the exact role and potential of PL in chondrogenic differentiation and chondrocyte regeneration.
Collapse
Affiliation(s)
- Ghmkin Hassan
- 1Faculty of Pharmacy, Damascus University, Damascus, Syria
| | | | - Issam Kasem
- 2Faculty of Sciences, Damascus University, Damascus, Syria.,National Commission for Biotechnology (NCBT), Damascus, Syria
| | - Chadi Soukkarieh
- 2Faculty of Sciences, Damascus University, Damascus, Syria.,National Commission for Biotechnology (NCBT), Damascus, Syria
| | - Majd Aljamali
- 1Faculty of Pharmacy, Damascus University, Damascus, Syria.,2Faculty of Sciences, Damascus University, Damascus, Syria.,National Commission for Biotechnology (NCBT), Damascus, Syria
| |
Collapse
|
45
|
Zhou L, Chen X, Liu T, Zhu C, Si M, Jargstorf J, Li M, Pan G, Gong Y, Luo ZP, Yang H, Pei M, He F. SIRT1-dependent anti-senescence effects of cell-deposited matrix on human umbilical cord mesenchymal stem cells. J Tissue Eng Regen Med 2018; 12:e1008-e1021. [PMID: 28107614 PMCID: PMC9805355 DOI: 10.1002/term.2422] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/07/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023]
Abstract
Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) are considered an attractive cell source for tissue regeneration. However, environmental oxidative stress can trigger premature senescence in MSCs and thus compromises their regenerative potential. Extracellular matrix (ECM) derived from MSCs has been shown to facilitate cell proliferation and multi-lineage differentiation. This investigation evaluated the effect of cell-deposited decellularized ECM (DECM) on oxidative stress-induced premature senescence in UC-MSCs. Sublethal dosages of H2 O2 , ranging from 50 μm to 200 μm, were used to induce senescence in MSCs. We found that DECM protected UC-MSCs from oxidative stress-induced premature senescence. When treated with H2 O2 at the same concentration, cell proliferation of DECM-cultured UC-MSCs was twofold higher than those on standard tissue culture polystyrene (TCPS). After exposure to 100 μm H2 O2 , fewer senescence-associated β-galactosidase-positive cells were observed on DECM than those on TCPS (17.6 ± 4.0% vs. 60.4 ± 6.2%). UC-MSCs cultured on DECM also showed significantly lower levels of senescence-related regulators, such as p16INK4α and p21. Most importantly, DECM preserved the osteogenic differentiation potential of UC-MSCs with premature senescence. The underlying molecular mechanisms involved the silent information regulator type 1 (SIRT1)-dependent signalling pathway, confirmed by the fact that the SIRT1 inhibitor nicotinamide counteracted the DECM-mediated anti-senescent effect. Collagen type I, rather than fibronectin, partially contributed to the protective effect of decellularized matrix. These findings provide a new strategy of using stem cell-deposited matrix to overcome the challenge of cellular senescence and to facilitate the clinical application of MSCs in regenerative medicine. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Long Zhou
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi Chen
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caihong Zhu
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Michelle Si
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Departments of Biology and Chemistry, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Joseph Jargstorf
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Biology, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Mao Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guoqing Pan
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Yihong Gong
- School of Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zong-Ping Luo
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
46
|
Zhang S, Chuah SJ, Lai RC, Hui JHP, Lim SK, Toh WS. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials 2018; 156:16-27. [DOI: 10.1016/j.biomaterials.2017.11.028] [Citation(s) in RCA: 373] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/17/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022]
|
47
|
Zeineddine HA, Frush TJ, Saleh ZM, El-Othmani MM, Saleh KJ. Applications of Tissue Engineering in Joint Arthroplasty: Current Concepts Update. Orthop Clin North Am 2017; 48:275-288. [PMID: 28577777 DOI: 10.1016/j.ocl.2017.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Research in tissue engineering has undoubtedly achieved significant milestones in recent years. Although it is being applied in several disciplines, tissue engineering's application is particularly advanced in orthopedic surgery and in degenerative joint diseases. The literature is full of remarkable findings and trials using tissue engineering in articular cartilage disease. With the vast and expanding knowledge, and with the variety of techniques available at hand, the authors aimed to review the current concepts and advances in the use of cell sources in articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Hussein A Zeineddine
- Department of Surgery, University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | - Todd J Frush
- Department of Orthopaedics and Sports Medicine, Detroit Medical Center, University Health Center (UHC) 9B, 4201 Saint Antoine Street, Detroit, MI 48201-2153, USA
| | - Zeina M Saleh
- Department of Surgery, American University of Beirut Medical Center, Bliss Street, Riad El-Solh, Beirut 11072020, Lebanon
| | - Mouhanad M El-Othmani
- Department of Orthopaedics and Sports Medicine, Musculoskeletal Institute of Excellence, Detroit Medical Center, University Health Center (UHC) 9B, 4201 Saint Antoine Street, Detroit, MI 48201-2153, USA
| | - Khaled J Saleh
- Department of Orthopaedics and Sports Medicine, Detroit Medical Center, University Health Center (UHC) 9B, 4201 Saint Antoine Street, Detroit, MI 48201-2153, USA.
| |
Collapse
|
48
|
Liu Y, Li D, Yin Z, Luo X, Liu W, Zhang W, Zhang Z, Cao Y, Liu Y, Zhou G. Prolonged in vitro precultivation alleviates post-implantation inflammation and promotes stable subcutaneous cartilage formation in a goat model. ACTA ACUST UNITED AC 2016; 12:015006. [PMID: 27910822 DOI: 10.1088/1748-605x/12/1/015006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Synthetic biodegradable scaffolds such as polylactic acid coated polyglycolic acid (PLA-PGA) are especially suitable for engineering shaped cartilage such as auricle, but they induce a serious inflammatory reaction particularly in the immunologically aggressive subcutaneous site, leading to resorption of the engineered autologous cartilage. Our previous study in a rabbit model has demonstrated 2 weeks of in vitro precultivation could significantly alleviate the post-implantation inflammation induced by PLA-PGA engineered cartilaginous grafts, but reproduction of this result failed in a preclinical goat model. The aims of the current study were to investigate whether prolonged in vitro precultivation could form a mature cartilaginous graft to resist the acute host response and promote stable subcutaneous cartilage formation in a preclinical goat model. Goat chondrocytes were seeded onto PLA-PGA scaffolds, in vitro precultivated for 2, 4, 8, and 12 weeks, and then implanted subcutaneously in autologous goats for 1 and 8 weeks. The in vitro engineered cartilage (vitro-EC) was examined histologically (hematoxylin and eosin, safranin-O, collagen II). The 1 week explants were examined histologically and stained for CD3, CD68, collagen I, and apoptosis. The 8 week explants were evaluated by histology, wet weight, volume, glycosaminoglycan (GAG) quantification and Young's modulus. With prolonged in vitro time, the quality of vitro-EC improved and the amount of scaffold residue decreased; more pronounced cartilage formation with fewer immune cells (CD3 and CD68 positive), apoptotic cells, and less collagen I expression were observed in explants that had been in vitro precultivated for a longer period. The subcutaneously regenerated neocartilage became more mature after prolonged implantation. These results suggested that prolonged in vitro precultivation allowed formation of a mature cartilaginous graft to resist the acute host response and promoted stable subcutaneous cartilage formation in autologous goats. These findings may provide useful reference for engineering auricle, trachea, nose, and eyelid shaped cartilage, for example.
Collapse
Affiliation(s)
- Yi Liu
- Shanghai 9th People's Hospital, School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai, People's Republic of China. Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, People's of Republic of China. These authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Pei M. Environmental preconditioning rejuvenates adult stem cells' proliferation and chondrogenic potential. Biomaterials 2016; 117:10-23. [PMID: 27923196 DOI: 10.1016/j.biomaterials.2016.11.049] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 11/24/2016] [Indexed: 12/13/2022]
Abstract
Adult stem cells are a promising cell source for cartilage regeneration. Unfortunately, due to donor age and ex vivo expansion, stem cell senescence becomes a huge hurdle for these cells to be used clinically. Increasing evidence indicates that environmental preconditioning is a powerful approach in promoting stem cells' ability to resist a harsh environment post-engraftment, such as hypoxia and inflammation. However, few reports organize and evaluate the literature regarding the rejuvenation effect of environmental preconditioning on stem cell proliferation and chondrogenic differentiation capacity, which are important variables for stem cell based tissue regeneration. This report aims to identify several critical environmental factors such as oxygen concentration, growth factors, and extracellular matrix and to discuss their preconditioning influence on stem cells' rejuvenation including proliferation and chondrogenic potential as well as underlying molecular mechanisms. We believe that environmental preconditioning based rejuvenation is a simpler and safer strategy to program pre-engraftment stem cells for better survival and enhanced proliferation and differentiation capacity without the undesired effects of some treatments, such as genetic manipulation.
Collapse
Affiliation(s)
- Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Exercise Physiology, West Virginia University, Morgantown, WV, USA; Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
50
|
Dhollander A, Verdonk P, Tirico LEP, Gomoll AH. Treatment of failed cartilage repair: State of the Art. J ISAKOS 2016. [DOI: 10.1136/jisakos-2016-000057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|