1
|
Cho S, Lee KS, Lee K, Kim HS, Park S, Yu SE, Ha H, Baek S, Kim J, Kim H, Lee JY, Lee S, Sung HJ. Surface Crystal and Degradability of Shape Memory Scaffold Essentialize Osteochondral Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401989. [PMID: 38855993 DOI: 10.1002/smll.202401989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/27/2024] [Indexed: 06/11/2024]
Abstract
The minimally invasive deployment of scaffolds is a key safety factor for the regeneration of cartilage and bone defects. Osteogenesis relies primarily on cell-matrix interactions, whereas chondrogenesis relies on cell-cell aggregation. Bone matrix expansion requires osteoconductive scaffold degradation. However, chondrogenic cell aggregation is promoted on the repellent scaffold surface, and minimal scaffold degradation supports the avascular nature of cartilage regeneration. Here, a material satisfying these requirements for osteochondral regeneration is developed by integrating osteoconductive hydroxyapatite (HAp) with a chondroconductive shape memory polymer (SMP). The shape memory function-derived fixity and recovery of the scaffold enabled minimally invasive deployment and expansion to fill irregular defects. The crystalline phases on the SMP surface inhibited cell aggregation by suppressing water penetration and subsequent protein adsorption. However, HAp conjugation SMP (H-SMP) enhanced surface roughness and consequent cell-matrix interactions by limiting cell aggregation using crystal peaks. After mouse subcutaneous implantation, hydrolytic H-SMP accelerated scaffold degradation compared to that by the minimal degradation observed for SMP alone for two months. H-SMP and SMP are found to promote osteogenesis and chondrogenesis, respectively, in vitro and in vivo, including the regeneration of rat osteochondral defects using the binary scaffold form, suggesting that this material is promising for osteochondral regeneration.
Collapse
Affiliation(s)
- Sungwoo Cho
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kang Suk Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- TMD LAB Co. Ltd., 6th Floor, 31, Gwangnaru-ro 8-gil, Seongdong-gu, Seoul, 04799, South Korea
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, South Korea
| | - Hye-Seon Kim
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Suji Park
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seung Eun Yu
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyunsu Ha
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sewoom Baek
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jueun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyunjae Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Ji Youn Lee
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sangmin Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- TMD LAB Co. Ltd., 6th Floor, 31, Gwangnaru-ro 8-gil, Seongdong-gu, Seoul, 04799, South Korea
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
2
|
Lippi L, Ferrillo M, Turco A, Folli A, Moalli S, Refati F, Perrero L, Ammendolia A, de Sire A, Invernizzi M. Multidisciplinary Rehabilitation after Hyaluronic Acid Injections for Elderly with Knee, Hip, Shoulder, and Temporomandibular Joint Osteoarthritis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2047. [PMID: 38004096 PMCID: PMC10672933 DOI: 10.3390/medicina59112047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint condition characterized by cartilage deterioration, joint inflammation, and functional limitations, particularly impacting the elderly population. Rehabilitation and hyaluronic acid (HA) injections are common therapeutic approaches routinely used in clinical practice, but their synergistic potential is far from being fully characterized. Thus, the aim of this narrative review was to elucidate the multilevel benefits and synergies of integrating these two approaches in multidisciplinary OA rehabilitation. This narrative review follows the scale for the assessment of narrative review articles (SANRA) criteria and involves a comprehensive literature search from July to August 2023. Two independent reviewers screened studies, including those involving human subjects with OA, rehabilitation strategies, and outcomes following HA injection, published in English. Results: HA injections might improve joint biomechanics, reducing friction, absorbing shocks, and potentially regulating inflammation. Rehabilitation plays a pivotal role in strengthening muscles, increasing the range of motion, and enhancing overall function. Optimizing rehabilitation following HA injection might provide additional benefits in joint health. OA management requires a multidisciplinary approach integrating HA injections, rehabilitation, and personalized care. Challenges in patient adherence and healthcare resources currently exist, but emerging technologies offer opportunities to enhance patient engagement and monitoring optimizing sustainability and outcomes of patients with knee, hip, shoulder, and temporomandibular joint OA.
Collapse
Affiliation(s)
- Lorenzo Lippi
- Department of Health Sciences, University of Eastern Piedmont “A. Avogadro”, 28100 Novara, Italy; (L.L.); (A.T.); (A.F.); (S.M.); (F.R.); (M.I.)
- Translational Medicine, Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Martina Ferrillo
- Department of Health Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| | - Alessio Turco
- Department of Health Sciences, University of Eastern Piedmont “A. Avogadro”, 28100 Novara, Italy; (L.L.); (A.T.); (A.F.); (S.M.); (F.R.); (M.I.)
| | - Arianna Folli
- Department of Health Sciences, University of Eastern Piedmont “A. Avogadro”, 28100 Novara, Italy; (L.L.); (A.T.); (A.F.); (S.M.); (F.R.); (M.I.)
| | - Stefano Moalli
- Department of Health Sciences, University of Eastern Piedmont “A. Avogadro”, 28100 Novara, Italy; (L.L.); (A.T.); (A.F.); (S.M.); (F.R.); (M.I.)
| | - Fjorelo Refati
- Department of Health Sciences, University of Eastern Piedmont “A. Avogadro”, 28100 Novara, Italy; (L.L.); (A.T.); (A.F.); (S.M.); (F.R.); (M.I.)
| | - Luca Perrero
- Neurorehabilitation Unit, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy;
| | - Antonio Ammendolia
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (A.A.); (A.d.S.)
- Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| | - Alessandro de Sire
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (A.A.); (A.d.S.)
- Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| | - Marco Invernizzi
- Department of Health Sciences, University of Eastern Piedmont “A. Avogadro”, 28100 Novara, Italy; (L.L.); (A.T.); (A.F.); (S.M.); (F.R.); (M.I.)
- Translational Medicine, Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| |
Collapse
|
3
|
Cui L, Yang Z, Hong J, Zhu Z, Wang Z, Liu Z, Zheng W, Hao Y, He J, Ni P, Cheng G. Injectable and Degradable POSS-Polyphosphate-Polysaccharide Hybrid Hydrogel Scaffold for Cartilage Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20625-20637. [PMID: 37078820 DOI: 10.1021/acsami.2c22947] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The limited self-repair capacity of articular cartilage has motivated the development of stem cell therapy based on artificial scaffolds that mimic the extracellular matrix (ECM) of cartilage tissue. In view of the specificity of articular cartilage, desirable tissue adhesiveness and stable mechanical properties under cyclic mechanical loads are critical for cartilage scaffolds. Herein, we developed an injectable and degradable organic-inorganic hybrid hydrogel as a cartilage scaffold based on polyhedral oligomeric silsesquioxane (POSS)-cored polyphosphate and polysaccharide. Specifically, acrylated 8-arm star-shaped POSS-poly(ethyl ethylene phosphate) (POSS-8PEEP-AC) was synthesized and cross-linked with thiolated hyaluronic acid (HA-SH) to form a degradable POSS-PEEP/HA hydrogel. Incorporation of POSS in the hydrogel increased the mechanical properties. The POSS-PEEP/HA hydrogel showed enzymatic biodegradability and favorable biocompatibility, supporting the growth and differentiation of human mesenchymal stem cells (hMSCs). The chondrogenic differentiation of encapsulated hMSCs was promoted by loading transforming growth factor-β3 (TGF-β3) in the hydrogel. In addition, the injectable POSS-PEEP/HA hydrogel was capable of adhering to rat cartilage tissue and resisting cyclic compression. Furthermore, in vivo results revealed that the transplanted hMSCs encapsulated in the POSS-PEEP/HA hydrogel scaffold significantly improved cartilage regeneration in rats, while the conjugation of TGF-β3 achieved a better therapeutic effect. The present work demonstrated the potential of the injectable, biodegradable, and mechanically enhanced POSS-PEEP/HA hybrid hydrogel as a scaffold biomaterial for cartilage regeneration.
Collapse
Affiliation(s)
- Leisha Cui
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zun Yang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Soochow University, Suzhou 215123, China
| | - Jing Hong
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zhanchi Zhu
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zhaojun Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zhongqing Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Wenlong Zheng
- Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou 215021, Jiangsu, China
| | - Ying Hao
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Jinlin He
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Soochow University, Suzhou 215123, China
| | - Peihong Ni
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Soochow University, Suzhou 215123, China
| | - Guosheng Cheng
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| |
Collapse
|
4
|
Chen Y, Lock J, Liu HH. Nanocomposites for cartilage regeneration. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00018-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
5
|
Miguel F, Barbosa F, Ferreira FC, Silva JC. Electrically Conductive Hydrogels for Articular Cartilage Tissue Engineering. Gels 2022; 8:710. [PMID: 36354618 PMCID: PMC9689960 DOI: 10.3390/gels8110710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 09/10/2023] Open
Abstract
Articular cartilage is a highly specialized tissue found in diarthrodial joints, which is crucial for healthy articular motion. Despite its importance, articular cartilage has limited regenerative capacities, and the degeneration of this tissue is a leading cause of disability worldwide, with hundreds of millions of people affected. As current treatment options for cartilage degeneration remain ineffective, tissue engineering has emerged as an exciting approach to create cartilage substitutes. In particular, hydrogels seem to be suitable candidates for this purpose due to their biocompatibility and high customizability, being able to be tailored to fit the biophysical properties of native cartilage. Furthermore, these hydrogel matrices can be combined with conductive materials in order to simulate the natural electrochemical properties of articular cartilage. In this review, we highlight the most common conductive materials combined with hydrogels and their diverse applications, and then present the current state of research on the development of electrically conductive hydrogels for cartilage tissue engineering. Finally, the main challenges and future perspectives for the application of electrically conductive hydrogels on articular cartilage repair strategies are also discussed.
Collapse
Affiliation(s)
- Filipe Miguel
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Frederico Barbosa
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Frederico Castelo Ferreira
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - João Carlos Silva
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
6
|
Zhu M, Zhong W, Cao W, Zhang Q, Wu G. Chondroinductive/chondroconductive peptides and their-functionalized biomaterials for cartilage tissue engineering. Bioact Mater 2022; 9:221-238. [PMID: 34820567 PMCID: PMC8585793 DOI: 10.1016/j.bioactmat.2021.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/19/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
The repair of articular cartilage defects is still challenging in the fields of orthopedics and maxillofacial surgery due to the avascular structure of articular cartilage and the limited regenerative capacity of mature chondrocytes. To provide viable treatment options, tremendous efforts have been made to develop various chondrogenically-functionalized biomaterials for cartilage tissue engineering. Peptides that are derived from and mimic the functions of chondroconductive cartilage extracellular matrix and chondroinductive growth factors, represent a unique group of bioactive agents for chondrogenic functionalization. Since they can be chemically synthesized, peptides bear better reproducibility, more stable efficacy, higher modifiability and yielding efficiency in comparison with naturally derived biomaterials and recombinant growth factors. In this review, we summarize the current knowledge in the designs of the chondroinductive/chondroconductive peptides, the underlying molecular mechanisms and their-functionalized biomaterials for cartilage tissue engineering. We also systematically compare their in-vitro and in-vivo efficacies in inducing chondrogenesis. Our vision is to stimulate the development of novel peptides and their-functionalized biomaterials for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mingjing Zhu
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
| | - Wenchao Zhong
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Wei Cao
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Mustapich T, Schwartz J, Palacios P, Liang H, Sgaglione N, Grande DA. A Novel Strategy to Enhance Microfracture Treatment With Stromal Cell-Derived Factor-1 in a Rat Model. Front Cell Dev Biol 2021; 8:595932. [PMID: 33634095 PMCID: PMC7902012 DOI: 10.3389/fcell.2020.595932] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
Background Microfracture is one of the most widely used techniques for the repair of articular cartilage. However, microfracture often results in filling of the chondral defect with fibrocartilage, which exhibits poor durability and sub-optimal mechanical properties. Stromal cell-derived factor-1 (SDF-1) is a potent chemoattractant for mesenchymal stem cells (MSCs) and is expressed at high levels in bone marrow adjacent to developing cartilage during endochondral bone formation. Integrating SDF-1 into an implantable collagen scaffold may provide a chondro-conductive and chondro-inductive milieu via chemotaxis of MSCs and promotion of chondrogenic differentiation, facilitating more robust hyaline cartilage formation following microfracture. Objective This work aimed to confirm the chemoattractive properties of SDF-1 in vitro and develop a one-step method for incorporating SDF-1 in vivo to enhance cartilage repair using a rat osteochondral defect model. Methods Bone marrow-derived MSCs (BMSCs) were harvested from the femurs of Sprague–Dawley rats and cultured in low-glucose Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum, with the medium changed every 3 days. Passage 1 MSCs were analyzed by flow cytometry with an S3 Cell Sorter (Bio-Rad). In vitro cell migration assays were performed on MSCs by labeling cells with carboxyfluorescein diacetate, succinimidyl ester (CFDA-SE; Bio-Rad). For the microfracture model, a 1.6-mm-diameter osteochondral defect was created in the femoral trochleae of 20 Sprague–Dawley rats bilaterally until bone marrow spillage was seen under saline irrigation. One knee was chosen at random to receive implantation of the scaffold, and the contralateral knee was left unfilled as an empty control. Type I collagen scaffolds (Kensey Nash) were coated with either gelatin only or gelatin and SDF-1 using a dip coating process. The rats received implantation of either a gelatin-only scaffold (N = 10) or gelatin-and-SDF-1 scaffold (N = 10) at the site of the microfracture. Femurs were collected for histological analyses at 4- and 8-week time points post-operatively, and sections were stained with Safranin O/Fast Green. The samples were graded blindly by two observers using the Modified O’Driscoll score, a validated scoring system for chondral repair. A minimum of 10 separate grading scores were made per sample and averaged. Quantitative comparisons of cell migration in vitro were performed with one-way ANOVA. Cartilage repair in vivo was also compared among groups with one-way ANOVA, and the results were presented as mean ± standard deviation, with P-values < 0.05 considered as statistically significant. Results MSC migration showed a dose–response relationship with SDF-1, with an optimal dosage for chemotaxis between 10 and 100 ng/ml. After scaffold implantation, the SDF-1-treated group demonstrated complete filling of the cartilage defect with mature cartilage tissue, exhibiting strong proteoglycan content, smooth borders, and good incorporation into marginal cartilage. Modified O’Driscoll scores after 8 weeks showed a significant improvement of cartilage repair in the SDF-1 group relative to the empty control group (P < 0.01), with a trend toward improvement when compared with the gelatin-only-scaffold group (P < 0.1). No significant differences in scores were found between the empty defect group and gelatin-only group. Conclusion In this study, we demonstrated a simple method for improving the quality of cartilage defect repair in a rat model of microfracture. We confirmed the chemotactic properties of SDF-1 on rat MSCs and found an optimized dosage range for chemotaxis between 10 and 100 ng/ml. Furthermore, we demonstrated a strategy to incorporate SDF-1 into gelatin–collagen I scaffolds in vivo at the site of an osteochondral defect. SDF-1-treated defects displayed robust hyaline cartilage resurfacing of the defect with minimal fibrous tissue, in contrast to the empty control group. The results of the in vitro and in vivo studies together suggest that SDF-1-mediated signaling may significantly improve the quality of cartilage regeneration in an osteochondral defect.
Collapse
Affiliation(s)
- Taylor Mustapich
- Orthopaedic Research Laboratory, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - John Schwartz
- Orthopaedic Research Laboratory, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Pablo Palacios
- Orthopaedic Research Laboratory, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Haixiang Liang
- Orthopaedic Research Laboratory, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Nicholas Sgaglione
- Department of Orthopaedic Surgery, Northwell Health, New Hyde Park, NY, United States
| | - Daniel A Grande
- Orthopaedic Research Laboratory, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Department of Orthopaedic Surgery, Northwell Health, New Hyde Park, NY, United States
| |
Collapse
|
8
|
Wei J, Wang K, Hettinghouse A, Liu C. Atsttrin Promotes Cartilage Repair Primarily Through TNFR2-Akt Pathway. Front Cell Dev Biol 2020; 8:577572. [PMID: 33195216 PMCID: PMC7658268 DOI: 10.3389/fcell.2020.577572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/12/2020] [Indexed: 01/16/2023] Open
Abstract
Background Cartilage defects account for substantial economic and humanistic burdens and pose a significant clinical problem. The efficacy of clinical approaches to cartilage repair is often inadequate, in part, owing to the restricted proliferative capacity of chondrocytes. Molecules have the capacity to promote the differentiation of multipotent mesenchymal stem cells into chondrocytes and may also gain the ability to repair the damaged cartilage. Objective This study aimed to investigate the role of Atsttrin (progranulin-derived engineered protein) in cartilage repair as well as the signaling pathway involved. Methods Primary and mesenchymal stem cell lines were used for the micromass culture. A murine cartilage defect model was used to determine the role of Atsttrin in cartilage repair in vivo. Real-time polymerase chain reaction and Western blot analysis were used to monitor the effect of Atsttrin on the transcriptional and protein levels, respectively, of key anabolic and catabolic signaling molecules. Results Atsttrin stimulated chondrogenesis in vitro and accelerated cartilage repair in vivo. In addition, Atsttrin-mediated cartilage repair occurred primarily through tumor necrosis factor receptor 2-initiated Akt signaling and downstream JunB transcription factor. Conclusion Atsttrin might serve as a promising therapeutic modality for cartilage regeneration.
Collapse
Affiliation(s)
- Jianlu Wei
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, China.,Department of Orthopaedic Surgery, New York University Langone Medical Center, New York, NY, United States
| | - Kaidi Wang
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | | | - Chuanju Liu
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, China.,Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
9
|
Klimek K, Ginalska G. Proteins and Peptides as Important Modifiers of the Polymer Scaffolds for Tissue Engineering Applications-A Review. Polymers (Basel) 2020; 12:E844. [PMID: 32268607 PMCID: PMC7240665 DOI: 10.3390/polym12040844] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022] Open
Abstract
Polymer scaffolds constitute a very interesting strategy for tissue engineering. Even though they are generally non-toxic, in some cases, they may not provide suitable support for cell adhesion, proliferation, and differentiation, which decelerates tissue regeneration. To improve biological properties, scaffolds are frequently enriched with bioactive molecules, inter alia extracellular matrix proteins, adhesive peptides, growth factors, hormones, and cytokines. Although there are many papers describing synthesis and properties of polymer scaffolds enriched with proteins or peptides, few reviews comprehensively summarize these bioactive molecules. Thus, this review presents the current knowledge about the most important proteins and peptides used for modification of polymer scaffolds for tissue engineering. This paper also describes the influence of addition of proteins and peptides on physicochemical, mechanical, and biological properties of polymer scaffolds. Moreover, this article sums up the major applications of some biodegradable natural and synthetic polymer scaffolds modified with proteins and peptides, which have been developed within the past five years.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland;
| | | |
Collapse
|
10
|
Le H, Xu W, Zhuang X, Chang F, Wang Y, Ding J. Mesenchymal stem cells for cartilage regeneration. J Tissue Eng 2020; 11:2041731420943839. [PMID: 32922718 PMCID: PMC7457700 DOI: 10.1177/2041731420943839] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022] Open
Abstract
Cartilage injuries are typically caused by trauma, chronic overload, and autoimmune diseases. Owing to the avascular structure and low metabolic activities of chondrocytes, cartilage generally does not self-repair following an injury. Currently, clinical interventions for cartilage injuries include chondrocyte implantation, microfracture, and osteochondral transplantation. However, rather than restoring cartilage integrity, these methods only postpone further cartilage deterioration. Stem cell therapies, especially mesenchymal stem cell (MSCs) therapies, were found to be a feasible strategy in the treatment of cartilage injuries. MSCs can easily be isolated from mesenchymal tissue and be differentiated into chondrocytes with the support of chondrogenic factors or scaffolds to repair damaged cartilage tissue. In this review, we highlighted the full success of cartilage repair using MSCs, or MSCs in combination with chondrogenic factors and scaffolds, and predicted their pros and cons for prospective translation to clinical practice.
Collapse
Affiliation(s)
- Hanxiang Le
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, P.R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Xiuli Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Yinan Wang
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, P.R. China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, P.R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| |
Collapse
|
11
|
Browe DC, Mahon OR, Díaz‐Payno PJ, Cassidy N, Dudurych I, Dunne A, Buckley CT, Kelly DJ. Glyoxal cross‐linking of solubilized extracellular matrix to produce highly porous, elastic, and chondro‐permissive scaffolds for orthopedic tissue engineering. J Biomed Mater Res A 2019; 107:2222-2234. [DOI: 10.1002/jbm.a.36731] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- David C. Browe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Olwyn R. Mahon
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- School of Biochemistry and Immunology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland
| | - Pedro J. Díaz‐Payno
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Nina Cassidy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
| | - Ivan Dudurych
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland
| | - Conor T. Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| |
Collapse
|
12
|
Tam WL, Luyten FP, Roberts SJ. From skeletal development to the creation of pluripotent stem cell-derived bone-forming progenitors. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0218. [PMID: 29786553 DOI: 10.1098/rstb.2017.0218] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Bone has many functions. It is responsible for protecting the underlying soft organs, it allows locomotion, houses the bone marrow and stores minerals such as calcium and phosphate. Upon damage, bone tissue can efficiently repair itself. However, healing is hampered if the defect exceeds a critical size and/or is in compromised conditions. The isolation or generation of bone-forming progenitors has applicability to skeletal repair and may be used in tissue engineering approaches. Traditionally, bone engineering uses osteochondrogenic stem cells, which are combined with scaffold materials and growth factors. Despite promising preclinical data, limited translation towards the clinic has been observed to date. There may be several reasons for this including the lack of robust cell populations with favourable proliferative and differentiation capacities. However, perhaps the most pertinent reason is the failure to produce an implant that can replicate the developmental programme that is observed during skeletal repair. Pluripotent stem cells (PSCs) can potentially offer a solution for bone tissue engineering by providing unlimited cell sources at various stages of differentiation. In this review, we summarize key embryonic signalling pathways in bone formation coupled with PSC differentiation strategies for the derivation of bone-forming progenitors.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Wai Long Tam
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Frank P Luyten
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Scott J Roberts
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium .,Bone Therapeutic Area, UCB Pharma, 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| |
Collapse
|
13
|
McAllister MJ, Chemaly M, Eakin AJ, Gibson DS, McGilligan VE. NLRP3 as a potentially novel biomarker for the management of osteoarthritis. Osteoarthritis Cartilage 2018; 26:612-619. [PMID: 29499288 DOI: 10.1016/j.joca.2018.02.901] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 02/09/2018] [Accepted: 02/20/2018] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) was previously thought of as 'wear and tear' as humans age, however there is increasing evidence to support an inflammatory theory. The nucleotide-binding and oligomerization domain-like receptor containing protein 3 (NLRP3) inflammasome has been implicated in the pathogenesis of a number of arthritic disorders, producing proinflammatory cytokines and degradative enzymes such as Interleukin-1 beta (IL-1β), Tumour necrosis factor alpha (TNF-α) and Matrix metalloproteinase-3 (MMP-3) which drive cartilage degeneration and synovial inflammation. This review aims to summarise the evidence of NLRP3 involvement in OA. Currently, treatment options focus on management of the disease and to date there is no cure. The development of novel biomarkers for OA could improve diagnosis, treatment and management. Importantly, this review provides detail on the involvement of the NLRP3 inflammasome in OA pathology and how its members could act as potential biomarkers to assist clinical decisions.
Collapse
Affiliation(s)
- M J McAllister
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom.
| | - M Chemaly
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom
| | - A J Eakin
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom
| | - D S Gibson
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom.
| | - V E McGilligan
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom
| |
Collapse
|
14
|
Yang W, Cao Y, Zhang Z, Du F, Shi Y, Li X, Zhang Q. Targeted delivery of FGF2 to subchondral bone enhanced the repair of articular cartilage defect. Acta Biomater 2018; 69:170-182. [PMID: 29408545 DOI: 10.1016/j.actbio.2018.01.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/28/2017] [Accepted: 01/25/2018] [Indexed: 01/25/2023]
Abstract
It is reported that growth factor (GF) is able to enhance the repair of articular cartilage (AC) defect, however underlying mechanisms of which are not fully elucidated yet. Moreover, the strategy for delivering GF needs to be optimized. The crosstalk between AC and subchondral bone (SB) play important role in the homeostasis and integrity of AC, therefore SB targeted delivery of GF represents one promising way to facilitate the repair of AC defect. In this study, we firstly investigated the effects and mechanism of FGF2 on surrounding SB and cartilage of detect defects in rabbits by using a homogenous collagen-based membranes. It was found that FGF2 had a modulating effect on the defect-surrounding SB via upregulation of bone morphogenetic protein (BMP)-2, BMP4 and SOX9 at the early stage. Low dose FGF2 improved the repair upon directly injected to SB. Inhibition of BMP signaling pathway compromised the beneficial effects of FGF2, which indicated the pivotal roles of BMP in the process. To facilitate SB targeted FGF2 delivery, a double-layered inhomogeneous collagen membrane was prepared and it induced increase of BMP2 and BMP4 in the synovial fluid, and subsequent successful repair of AC defect. Taken together, this targeted delivery of FGF2 to SB provides a promising strategy for AC repair owing to the relatively clear mechanism, less amount of it, and short duration of delivery. STATEMENT OF SIGNIFICANCE Articular cartilage (AC) and subchondral bone (SB) form an integral functional unit. The homeostasis and integrity of AC depend on its crosstalk with the SB. However, the function of the SB in AC defect repair is not completely understood. The application of growth factors to promote the repair articular cartilage defect is a promising strategy, but still under the optimization. Our study demonstrate that SB plays important roles in the repair of AC defect. Particularly, SB is the effective target of fibroblast growth factor 2 (FGF2), and targeted delivery of FGF2 can modulate SB and thus significantly enhances the repair of AC defect. Therefore, targeted delivery of growth factor to SB is a novel promising strategy to improve the repair of AC defect.
Collapse
|
15
|
Ambra LF, de Girolamo L, Mosier B, Gomoll AH. Review: Interventions for Cartilage Disease: Current State-of-the-Art and Emerging Technologies. Arthritis Rheumatol 2017; 69:1363-1373. [PMID: 28294573 DOI: 10.1002/art.40094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Luiz Felipe Ambra
- Universidade Federal de São Paulo, Sao Paulo, Brazil, and Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Brian Mosier
- Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Andreas H Gomoll
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
16
|
Forget J, Awaja F, Gugutkov D, Gustavsson J, Gallego Ferrer G, Coelho-Sampaio T, Hochman-Mendez C, Salmeron-Sánchez M, Altankov G. Differentiation of Human Mesenchymal Stem Cells Toward Quality Cartilage Using Fibrinogen-Based Nanofibers. Macromol Biosci 2016; 16:1348-59. [DOI: 10.1002/mabi.201600080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/02/2016] [Indexed: 12/27/2022]
Affiliation(s)
| | - Firas Awaja
- Molecular Dynamics at Cell Biomaterial Interface; Institute for Bioengineering of Catalonia (IBEC); Barcelona Spain
| | - Dencho Gugutkov
- Molecular Dynamics at Cell Biomaterial Interface; Institute for Bioengineering of Catalonia (IBEC); Barcelona Spain
| | - Juhan Gustavsson
- Molecular Dynamics at Cell Biomaterial Interface; Institute for Bioengineering of Catalonia (IBEC); Barcelona Spain
| | - Gloria Gallego Ferrer
- Center for Biomaterials and Tissue Engineering (CBIT); Universitat Politècnica de València; Valencia Spain
- Biomedical Research Networking Center in Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
| | - Tatiana Coelho-Sampaio
- Molecular Dynamics at Cell Biomaterial Interface; Institute for Bioengineering of Catalonia (IBEC); Barcelona Spain
- Institute of Biomedical Sciences; Federal University of Rio de Janeiro; Rio de Janeiro Brazil
- Institute of Biophysics Carlos Chagas Filho; Federal University of Rio de Janeiro; Rio de Janeiro Brazil
| | - Camila Hochman-Mendez
- Institute of Biomedical Sciences; Federal University of Rio de Janeiro; Rio de Janeiro Brazil
- Institute of Biophysics Carlos Chagas Filho; Federal University of Rio de Janeiro; Rio de Janeiro Brazil
| | - Manuel Salmeron-Sánchez
- Division of Biomedical Engineering; School of Engineering; University of Glasgow; Glasgow G12 8LT UK
| | - George Altankov
- Molecular Dynamics at Cell Biomaterial Interface; Institute for Bioengineering of Catalonia (IBEC); Barcelona Spain
- Biomedical Research Networking Center in Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA); Barcelona Spain
| |
Collapse
|
17
|
Jeuken RM, Roth AK, Peters RJRW, Van Donkelaar CC, Thies JC, Van Rhijn LW, Emans PJ. Polymers in Cartilage Defect Repair of the Knee: Current Status and Future Prospects. Polymers (Basel) 2016; 8:E219. [PMID: 30979313 PMCID: PMC6432241 DOI: 10.3390/polym8060219] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 02/06/2023] Open
Abstract
Cartilage defects in the knee are often seen in young and active patients. There is a need for effective joint preserving treatments in patients suffering from cartilage defects, as untreated defects often lead to osteoarthritis. Within the last two decades, tissue engineering based techniques using a wide variety of polymers, cell sources, and signaling molecules have been evaluated. We start this review with basic background information on cartilage structure, its intrinsic repair, and an overview of the cartilage repair treatments from a historical perspective. Next, we thoroughly discuss polymer construct components and their current use in commercially available constructs. Finally, we provide an in-depth discussion about construct considerations such as degradation rates, cell sources, mechanical properties, joint homeostasis, and non-degradable/hybrid resurfacing techniques. As future prospects in cartilage repair, we foresee developments in three areas: first, further optimization of degradable scaffolds towards more biomimetic grafts and improved joint environment. Second, we predict that patient-specific non-degradable resurfacing implants will become increasingly applied and will provide a feasible treatment for older patients or failed regenerative treatments. Third, we foresee an increase of interest in hybrid construct, which combines degradable with non-degradable materials.
Collapse
Affiliation(s)
- Ralph M Jeuken
- Department of Orthopaedic Surgery, Maastricht University Medical Center, P. Debyelaan 25, Maastricht 6229 HX, The Netherlands.
| | - Alex K Roth
- Department of Orthopaedic Surgery, Maastricht University Medical Center, P. Debyelaan 25, Maastricht 6229 HX, The Netherlands.
| | | | - Corrinus C Van Donkelaar
- Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands.
| | - Jens C Thies
- DSM Biomedical, Koestraat 1, Geleen 6167 RA, The Netherlands.
| | - Lodewijk W Van Rhijn
- Department of Orthopaedic Surgery, Maastricht University Medical Center, P. Debyelaan 25, Maastricht 6229 HX, The Netherlands.
| | - Pieter J Emans
- Department of Orthopaedic Surgery, Maastricht University Medical Center, P. Debyelaan 25, Maastricht 6229 HX, The Netherlands.
| |
Collapse
|
18
|
Zhang W, Ouyang H, Dass CR, Xu J. Current research on pharmacologic and regenerative therapies for osteoarthritis. Bone Res 2016; 4:15040. [PMID: 26962464 PMCID: PMC4772471 DOI: 10.1038/boneres.2015.40] [Citation(s) in RCA: 325] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 12/05/2015] [Accepted: 12/06/2015] [Indexed: 01/05/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disorder commonly encountered in clinical practice, and is the leading cause of disability in elderly people. Due to the poor self-healing capacity of articular cartilage and lack of specific diagnostic biomarkers, OA is a challenging disease with limited treatment options. Traditional pharmacologic therapies such as acetaminophen, non-steroidal anti-inflammatory drugs, and opioids are effective in relieving pain but are incapable of reversing cartilage damage and are frequently associated with adverse events. Current research focuses on the development of new OA drugs (such as sprifermin/recombinant human fibroblast growth factor-18, tanezumab/monoclonal antibody against β-nerve growth factor), which aims for more effectiveness and less incidence of adverse effects than the traditional ones. Furthermore, regenerative therapies (such as autologous chondrocyte implantation (ACI), new generation of matrix-induced ACI, cell-free scaffolds, induced pluripotent stem cells (iPS cells or iPSCs), and endogenous cell homing) are also emerging as promising alternatives as they have potential to enhance cartilage repair, and ultimately restore healthy tissue. However, despite currently available therapies and research advances, there remain unmet medical needs in the treatment of OA. This review highlights current research progress on pharmacologic and regenerative therapies for OA including key advances and potential limitations.
Collapse
Affiliation(s)
- Wei Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China
- School of Pathology and Laboratory Medicine, The University of Western Australia, Perth WA 6009, Australia
| | - Hongwei Ouyang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Crispin R Dass
- School of Pharmacy, Building 306, Curtin University, Bentley, Perth WA 6102, Australia
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, The University of Western Australia, Perth WA 6009, Australia
| |
Collapse
|
19
|
Wang CC, Lee CH, Peng YJ, Salter DM, Lee HS. Platelet-Rich Plasma Attenuates 30-kDa Fibronectin Fragment-Induced Chemokine and Matrix Metalloproteinase Expression by Meniscocytes and Articular Chondrocytes. Am J Sports Med 2015; 43:2481-9. [PMID: 26306780 DOI: 10.1177/0363546515597489] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Proteolytic fragments of fibronectin have catabolic effects on cartilage and menisci. Platelet-rich plasma (PRP) is increasingly being used to treat a range of joint conditions, but it is unknown whether PRP influences fibronectin fragment (FN-f) procatabolic activity. HYPOTHESES The procatabolic activity of FN-f on meniscocytes and articular chondrocytes is attenuated by cotreatment with PRP. STUDY DESIGN Controlled laboratory study. METHODS Human meniscocytes were treated with FN-f (30 kDa) with or without PRP coincubation, and gene expression was analyzed by complementary DNA microarray analysis. Validation of altered expression of known and novel chemokine and protease genes was undertaken by real-time polymerase chain reaction (RT-PCR) in articular chondrocytes and meniscocytes. Chemokine release was assayed by enzyme-linked immunosorbent assay, and intracellular pathway signaling was evaluated by Western immunoblotting. RESULTS Microarray analysis and RT-PCR showed increased expression of matrix metalloproteinase (MMP)1, MMP2, MMP3, MMP9, MMP13, interleukin (IL)-6, IL-8 (CXCL8), CCL5, CCL20, and CXCL10 chemokines in meniscocytes after treatment with FN-f. Upregulation of these genes was significantly attenuated by PRP. Similar results were seen with articular chondrocytes, although no changes in MMP2 or MMP9 levels were identified. PRP-induced suppression of gene expression was associated with activation of Akt and p44/p42. CONCLUSION PRP treatment attenuates the 30-kDa FN-f-induced expression of a range of proinflammatory chemokines and MMPs, including IL-8, IL-6, CCL20, CCL5, CXCL10, MMP1, MMP3, and MMP13, by both meniscocytes and articular chondrocytes. CLINICAL RELEVANCE These observations provide support for the use and further trials of PRP in management of cartilage and meniscal injuries.
Collapse
Affiliation(s)
- Chih-Chien Wang
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan Department of Orthopedics, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Chian-Her Lee
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Donald M Salter
- Osteoarticular Research Group, Centre for Genomics and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Herng-Sheng Lee
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Ji X, Guo S, Zeng C, Wang C, Zhang L. Continuous generation of alginate microfibers with spindle-knots by using a simple microfluidic device. RSC Adv 2015. [DOI: 10.1039/c4ra10389k] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Calcium alginate microfibers with spindle-knots are fabricated by combining microfluidic technique with wet-spinning method. The structures of the knots can be conveniently regulated by changing the two-phase flow rate ratio and the micropipette diameter.
Collapse
Affiliation(s)
- Xiaobo Ji
- State Key Laboratory of Materials-Oriented Chemical Engineering
- College of Chemistry and Chemical Engineering
- Nanjing Tech University
- Nanjing 210009
- P. R. China
| | - Song Guo
- State Key Laboratory of Materials-Oriented Chemical Engineering
- College of Chemistry and Chemical Engineering
- Nanjing Tech University
- Nanjing 210009
- P. R. China
| | - Changfeng Zeng
- College of Mechanical and Power Engineering
- Nanjing Tech University
- Nanjing 210009
- P. R. China
| | - Chongqing Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering
- College of Chemistry and Chemical Engineering
- Nanjing Tech University
- Nanjing 210009
- P. R. China
| | - Lixiong Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering
- College of Chemistry and Chemical Engineering
- Nanjing Tech University
- Nanjing 210009
- P. R. China
| |
Collapse
|
21
|
Radially oriented collagen scaffold with SDF-1 promotes osteochondral repair by facilitating cell homing. Biomaterials 2015; 39:114-23. [DOI: 10.1016/j.biomaterials.2014.10.049] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/19/2014] [Indexed: 12/13/2022]
|
22
|
Chahine NO, Collette NM, Thomas CB, Genetos DC, Loots GG. Nanocomposite scaffold for chondrocyte growth and cartilage tissue engineering: effects of carbon nanotube surface functionalization. Tissue Eng Part A 2014; 20:2305-15. [PMID: 24593020 DOI: 10.1089/ten.tea.2013.0328] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The goal of this study was to assess the long-term biocompatibility of single-wall carbon nanotubes (SWNTs) for tissue engineering of articular cartilage. We hypothesized that SWNT nanocomposite scaffolds in cartilage tissue engineering can provide an improved molecular-sized substrate for stimulation of chondrocyte growth, as well as structural reinforcement of the scaffold's mechanical properties. The effect of SWNT surface functionalization (-COOH or -PEG) on chondrocyte viability and biochemical matrix deposition was examined in two-dimensional cultures, in three-dimensional (3D) pellet cultures, and in a 3D nanocomposite scaffold consisting of hydrogels+SWNTs. Outcome measures included cell viability, histological and SEM evaluation, GAG biochemical content, compressive and tensile biomechanical properties, and gene expression quantification, including extracellular matrix (ECM) markers aggrecan (Agc), collagen-1 (Col1a1), collagen-2 (Col2a1), collagen-10 (Col10a1), surface adhesion proteins fibronectin (Fn), CD44 antigen (CD44), and tumor marker (Tp53). Our findings indicate that chondrocytes tolerate functionalized SWNTs well, with minimal toxicity of cells in 3D culture systems (pellet and nanocomposite constructs). Both SWNT-PEG and SWNT-COOH groups increased the GAG content in nanocomposites relative to control. The compressive biomechanical properties of cell-laden SWNT-COOH nanocomposites were significantly elevated relative to control. Increases in the tensile modulus and ultimate stress were observed, indicative of a tensile reinforcement of the nanocomposite scaffolds. Surface coating of SWNTs with -COOH also resulted in increased Col2a1 and Fn gene expression throughout the culture in nanocomposite constructs, indicative of increased chondrocyte metabolic activity. In contrast, surface coating of SWNTs with a neutral -PEG moiety had no significant effect on Col2a1 or Fn gene expression, suggesting that the charged nature of the -COOH surface functionalization may promote ECM expression in this culture system. The results of this study indicate that SWNTs exhibit a unique potential for cartilage tissue engineering, where functionalization with bioactive molecules may provide an improved substrate for stimulation of cellular growth and repair.
Collapse
Affiliation(s)
- Nadeen O Chahine
- 1 Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research , Manhasset, New York
| | | | | | | | | |
Collapse
|
23
|
Mathews S, Bhonde R, Gupta PK, Totey S. Novel biomimetic tripolymer scaffolds consisting of chitosan, collagen type 1, and hyaluronic acid for bone marrow-derived human mesenchymal stem cells-based bone tissue engineering. J Biomed Mater Res B Appl Biomater 2014; 102:1825-34. [PMID: 24723571 DOI: 10.1002/jbm.b.33152] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 03/04/2014] [Accepted: 03/13/2014] [Indexed: 12/17/2022]
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) are an ideal osteogenic cell source for bone tissue engineering (BTE). A scaffold, in the context of BTE, is the extracellular matrix (ECM) that provides the unique microenvironment and play significant role in regulating cell behavior, differentiation, and development in an in vitro culture system. In this study, we have developed novel biomimetic tripolymer scaffolds for BTE using an ECM protein, collagen type 1; an ECM glycosaminoglycan, hyaluronic acid; and a natural osteoconductive polymer, chitosan. The scaffolds were characterized by scanning electron microscopy (SEM) and swelling ratio. The scaffolds were seeded with hMSCs and tested for cytocompatibility and osteogenic potential. The scaffolds supported cell adhesion, enhanced cell proliferation, promoted cell migration, showed good cell viability, and osteogenic potential. The cells were able to migrate out from the scaffolds in favorable conditions. SEM, alkaline phosphatase assay, and immunofluorescent staining confirmed the differentiation of hMSCs to osteogenic lineage in the scaffolds. In conclusion, we have successfully developed biomimetic scaffolds that supported the proliferation and differentiation of hMSCs. These scaffolds hold great promise as a cell-delivery vehicle for regenerative therapies and as a support system for enhancing bone regeneration.
Collapse
Affiliation(s)
- Smitha Mathews
- Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad-500007, India
| | | | | | | |
Collapse
|
24
|
Lanni S, Martini A, Malattia C. Heading Toward a Modern Imaging Approach in Juvenile Idiopathic Arthritis. Curr Rheumatol Rep 2014; 16:416. [DOI: 10.1007/s11926-014-0416-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
25
|
Orth P, Rey-Rico A, Venkatesan JK, Madry H, Cucchiarini M. Current perspectives in stem cell research for knee cartilage repair. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2014; 7:1-17. [PMID: 24520197 PMCID: PMC3897321 DOI: 10.2147/sccaa.s42880] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protocols based on the delivery of stem cells are currently applied in patients, showing encouraging results for the treatment of articular cartilage lesions (focal defects, osteoarthritis). Yet, restoration of a fully functional cartilage surface (native structural organization and mechanical functions) especially in the knee joint has not been reported to date, showing the need for improved designs of clinical trials. Various sources of progenitor cells are now available, originating from adult tissues but also from embryonic or reprogrammed tissues, most of which have already been evaluated for their chondrogenic potential in culture and for their reparative properties in vivo upon implantation in relevant animal models of cartilage lesions. Nevertheless, particular attention will be needed regarding their safe clinical use and their potential to form a cartilaginous repair tissue of proper quality and functionality in the patient. Possible improvements may reside in the use of biological supplements in accordance with regulations, while some challenges remain in establishing standardized, effective procedures in the clinics.
Collapse
Affiliation(s)
- Patrick Orth
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Henning Madry
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany ; Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
26
|
Grande DA, Schwartz JA, Brandel E, Chahine NO, Sgaglione N. Articular Cartilage Repair: Where We Have Been, Where We Are Now, and Where We Are Headed. Cartilage 2013; 4:281-5. [PMID: 26069673 PMCID: PMC4297160 DOI: 10.1177/1947603513494402] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This review traces the genealogy of the field of articular cartilage repair from its earliest attempts to its present day vast proliferation of research advances. Prior to the 1980s there was only sporadic efforts to regenerate articular cartilage as it was considered to be incapable of regeneration based on historical dogma. The first flurry of reports documented the use of various cell types ultimately leading to the first successful demonstration of autologous chondrocyte transplantation which was later translated to clinical use and has resulted in the revised axiom that cartilage regeneration is possible. The current field of cartilage repair is multifaceted and some of the 1980s' vintage concepts have been revisited with state of the art technology now available. The future of the field is now poised to undertake the repair of whole cartilage surfaces beyond focal defects and an appreciation for integrated whole joint health to restore cartilage homeostasis.
Collapse
Affiliation(s)
- Daniel A. Grande
- Department of Orthopaedic Surgery, Feinstein Institute for Medical Research, North Shore–LIJ Health Systems, Manhasset, NY, USA
| | - John A. Schwartz
- Department of Orthopaedic Surgery, Feinstein Institute for Medical Research, North Shore–LIJ Health Systems, Manhasset, NY, USA
| | - Eric Brandel
- Department of Orthopaedic Surgery, Feinstein Institute for Medical Research, North Shore–LIJ Health Systems, Manhasset, NY, USA
| | - Nadeen O. Chahine
- Department of Orthopaedic Surgery, Feinstein Institute for Medical Research, North Shore–LIJ Health Systems, Manhasset, NY, USA
| | - Nicholas Sgaglione
- Department of Orthopaedic Surgery, Feinstein Institute for Medical Research, North Shore–LIJ Health Systems, Manhasset, NY, USA
| |
Collapse
|
27
|
McCullen SD, Autefage H, Callanan A, Gentleman E, Stevens MM. Anisotropic fibrous scaffolds for articular cartilage regeneration. Tissue Eng Part A 2012; 18:2073-83. [PMID: 22655795 DOI: 10.1089/ten.tea.2011.0606] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Articular cartilage lesions, which can progress to osteoarthritis, are a particular challenge for regenerative medicine strategies, as cartilage function stems from its complex depth-dependent microstructural organization, mechanical properties, and biochemical composition. Fibrous scaffolds offer a template for cartilage extracellular matrix production; however, the success of homogeneous scaffolds is limited by their inability to mimic the cartilage's zone-specific organization and properties. We fabricated trilaminar scaffolds by sequential electrospinning and varying fiber size and orientation in a continuous construct, to create scaffolds that mimicked the structural organization and mechanical properties of cartilage's collagen fibrillar network. Trilaminar composite scaffolds were then compared to homogeneous aligned or randomly oriented fiber scaffolds to assess in vitro cartilage formation. Bovine chondrocytes proliferated and produced a type II collagen and a sulfated glycosaminoglycan-rich extracellular matrix on all scaffolds. Furthermore, all scaffolds promoted significant upregulation of aggrecan and type II collagen gene expression while downregulating that of type I collagen. Compressive testing at physiological strain levels further demonstrated that the mechanical properties of trilaminar composite scaffolds approached those of native cartilage. Our results demonstrate that trilaminar composite scaffolds mimic key organizational characteristics of native cartilage, support in vitro cartilage formation, and have superior mechanical properties to homogenous scaffolds. We propose that these scaffolds offer promise in regenerative medicine strategies to repair articular cartilage lesions.
Collapse
Affiliation(s)
- Seth D McCullen
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
28
|
Abstract
Joint destruction occurs in both osteoarthritis and rheumatoid arthritis. Even in the era of biologic agents, this destruction can be delayed but not averted. As cartilage has limited ability to self-regenerate, joint arthroplasty is required. Here, we outline current tissue engineering procedures (including autologous chondrocyte implantation and in situ mesenchymal stem cell recruitment) that are routinely applied for the regenerative treatment of injured or early osteoarthritic cartilage. Potential future regenerative therapies, including administration of multipotent or pluripotent stem cells, are also discussed. In the future, cell-free, material-based (for cartilage lesions) or cell-free, factor-based (for osteoarthritic cartilage) therapies to facilitate the recruitment of repair cells and improve cartilage metabolism are likely to become more important. Moreover, delivery of anti-inflammatory factors or immunomodulatory cells could be a regenerative treatment option for rheumatoid arthritis. Tissue engineering faces a crucial phase to translate products into clinical routine and the regulatory framework for cell-based products in particular is an important issue.
Collapse
|
29
|
Moran CJ, Barry FP, Maher SA, Shannon FJ, Rodeo SA. Advancing regenerative surgery in orthopaedic sports medicine: the critical role of the surgeon. Am J Sports Med 2012; 40:934-44. [PMID: 22085730 DOI: 10.1177/0363546511426677] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The constant desire to improve outcomes in orthopaedic sports medicine requires us to continuously consider the challenges faced in the surgical repair or reconstruction of soft tissue and cartilaginous injury. In many cases, surgical efforts targeted at restoring normal anatomy and functional status are ultimately impaired by the biological aspect of the natural history of these injuries, which acts as an obstacle to a satisfactory repair process after surgery. The clinical management of sports injuries and the delivery of appropriate surgical intervention are continuously evolving, and it is likely that the principles of regenerative medicine will have an increasing effect in this specialized field of orthopaedic practice going forward. Ongoing advances in arthroscopy and related surgical techniques should facilitate this process. In contrast to the concept of engineered replacement of entire tissues, it is probable that the earliest effect of regenerative strategies seen in clinical practice will involve biological augmentation of current operative techniques via a synergistic process that might be best considered "regenerative surgery." This article provides an overview of the principles of regenerative surgery in cartilage repair and related areas of orthopaedic surgery sports medicine. The possibilities and challenges of a gradual yet potential paradigm shift in treatment through the increased use of biological augmentation are considered. The translational process and critical role to be played by the specialist surgeon are also addressed. We conclude that increased understanding of the potential and challenges of regenerative surgery should allow those specializing in orthopaedic surgery sports medicine to lead the way in advancing the frontiers of biological strategies to enhance modern clinical care in an evidence-based manner.
Collapse
Affiliation(s)
- Cathal J Moran
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.
| | | | | | | | | |
Collapse
|
30
|
LaPorta TF, Richter A, Sgaglione NA, Grande DA. Clinical relevance of scaffolds for cartilage engineering. Orthop Clin North Am 2012; 43:245-54, vi. [PMID: 22480473 DOI: 10.1016/j.ocl.2012.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The repair of articular cartilage defects in patients' knees presents a particular challenge to the orthopedic surgeon because cartilage lacks the ability to repair or regenerate itself. Various cartilage repair techniques have not produced a superior or uniform outcome, which has led to a new generation of cartilage repair based on tissue-engineering strategies and the use of biological scaffolds. Clinical advances have been made regarding the regeneration of articular cartilage, and continue to be made toward the achievement of a suitable treatment method for resurfacing osteochondral defects, through cartilage tissue engineering and the use of pluripotent cells seeded on bio-scaffolds.
Collapse
Affiliation(s)
- Thomas F LaPorta
- Department of Orthopaedics, Long Island Jewish Medical Center, Street 270-05 76th Avenue, New Hyde Park, NY 11040, USA.
| | | | | | | |
Collapse
|
31
|
Gomoll AH, Filardo G, de Girolamo L, Espregueira-Mendes J, Marcacci M, Rodkey WG, Steadman JR, Zaffagnini S, Kon E. Surgical treatment for early osteoarthritis. Part I: cartilage repair procedures. Knee Surg Sports Traumatol Arthrosc 2012; 20:450-66. [PMID: 22113219 DOI: 10.1007/s00167-011-1780-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/08/2011] [Indexed: 01/30/2023]
Abstract
Young patients with early osteoarthritis (OA) represent a challenging population due to a combination of high functional demands and limited treatment options. Conservative measures such as injection and physical therapy can provide short-term pain relief but are only palliative in nature. Joint replacement, a successful procedure in the older population, is controversial in younger patients, who are less satisfied and experience higher failure rates. Therefore, while traditionally not indicated for the treatment of OA, cartilage repair has become a focus of increased interest due to its potential to provide pain relief and alter the progression of degenerative disease, with the hope of delaying or obviating the need for joint replacement. This review of cartilage repair techniques will discuss currently available procedures, specifically pertaining to experiences in the setting of early OA. Level of evidence IV.
Collapse
Affiliation(s)
- A H Gomoll
- Cartilage Repair Center, Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kang E, Jeong GS, Choi YY, Lee KH, Khademhosseini A, Lee SH. Digitally tunable physicochemical coding of material composition and topography in continuous microfibres. NATURE MATERIALS 2011; 10:877-83. [PMID: 21892177 DOI: 10.1038/nmat3108] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 08/01/2011] [Indexed: 05/20/2023]
Abstract
Heterotypic functional materials with compositional and topographical properties that vary spatiotemporally on the micro- or nanoscale are common in nature. However, fabricating such complex materials in the laboratory remains challenging. Here we describe a method to continuously create microfibres with tunable morphological, structural and chemical features using a microfluidic system consisting of a digital, programmable flow control that mimics the silk-spinning process of spiders. With this method we fabricated hydrogel microfibres coded with varying chemical composition and topography along the fibre, including gas micro-bubbles as well as nanoporous spindle-knots and joints that enabled directional water collection. We also explored the potential use of the coded microfibres for tissue engineering applications by creating multifunctional microfibres with a spatially controlled co-culture of encapsulated cells.
Collapse
Affiliation(s)
- Edward Kang
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
33
|
Ravindran S, Roam JL, Nguyen PK, Hering TM, Elbert DL, McAlinden A. Changes of chondrocyte expression profiles in human MSC aggregates in the presence of PEG microspheres and TGF-β3. Biomaterials 2011; 32:8436-45. [PMID: 21820171 DOI: 10.1016/j.biomaterials.2011.07.056] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 07/17/2011] [Indexed: 11/27/2022]
Abstract
Biomaterial microparticles are commonly utilized as growth factor delivery vehicles to induce chondrogenic differentiation of mesenchymal stem/stromal cells (MSCs). To address whether the presence of microparticles could themselves affect differentiation of MSCs, a 3D co-aggregate system was developed containing an equal volume of human primary bone marrow-derived MSCs and non-degradable RGD-conjugated poly(ethylene glycol) microspheres (PEG-μs). Following TGF-β3 induction, differences in cell phenotype, gene expression and protein localization patterns were found when compared to MSC aggregate cultures devoid of PEG-μs. An outer fibrous layer always found in differentiated MSC aggregate cultures was not formed in the presence of PEG-μs. Type II collagen protein was synthesized by cells in both culture systems, although increased levels of the long (embryonic) procollagen isoforms were found in MSC/PEG-μs aggregates. Ubiquitous deposition of type I and type X collagen proteins was found in MSC/PEG-μs cultures while the expression patterns of these collagens was restricted to specific areas in MSC aggregates. These findings show that MSCs respond differently to TGF-β3 when in a PEG-μs environment due to effects of cell dilution, altered growth factor diffusion and/or cellular interactions with the microspheres. Although not all of the expression patterns pointed toward improved chondrogenic differentiation in the MSC/PEG-μs cultures, the surprisingly large impact of the microparticles themselves should be considered when designing drug delivery/scaffold strategies.
Collapse
Affiliation(s)
- Soumya Ravindran
- Department of Orthopaedic Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
34
|
Sheehy EJ, Buckley CT, Kelly DJ. Chondrocytes and bone marrow-derived mesenchymal stem cells undergoing chondrogenesis in agarose hydrogels of solid and channelled architectures respond differentially to dynamic culture conditions. J Tissue Eng Regen Med 2011; 5:747-58. [DOI: 10.1002/term.385] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 10/13/2010] [Indexed: 12/18/2022]
|
35
|
On the Horizon From the ORS. J Am Acad Orthop Surg 2011; 19:59-62. [PMID: 29329125 DOI: 10.5435/00124635-201101000-00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
36
|
Moran CJ, Shannon FJ, Barry FP, O’Byrne JM, O’Brien T, Curtin W. Translation of science to surgery. ACTA ACUST UNITED AC 2010; 92:1195-202. [DOI: 10.1302/0301-620x.92b9.23651] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Orthopaedic surgery is in an exciting transitional period as modern surgical interventions, implants and scientific developments are providing new therapeutic options. As advances in basic science and technology improve our understanding of the pathology and repair of musculoskeletal tissue, traditional operations may be replaced by newer, less invasive procedures which are more appropriately targeted at the underlying pathophysiology. However, evidence-based practice will remain a basic requirement of care. Orthopaedic surgeons can and should remain at the forefront of the development of novel therapeutic interventions and their application. Progression of the potential of bench research into an improved array of orthopaedic treatments in an effective yet safe manner will require the development of a subgroup of specialists with extended training in research to play an important role in bridging the gap between laboratory science and clinical practice. International regulations regarding the introduction of new biological treatments will place an additional burden on the mechanisms of this translational process, and orthopaedic surgeons who are trained in science, surgery and the regulatory environment will be essential. Training and supporting individuals with these skills requires special consideration and discussion by the orthopaedic community. In this paper we review some traditional approaches to the integration of orthopaedic science and surgery, the therapeutic potential of current regenerative biomedical science for cartilage repair and ways in which we may develop surgeons with the skills required to translate scientific discovery into effective and properly assessed orthopaedic treatments.
Collapse
Affiliation(s)
- C. J. Moran
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - F. J. Shannon
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - F. P. Barry
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - J. M. O’Byrne
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - T. O’Brien
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - W. Curtin
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| |
Collapse
|