1
|
Li H, Gong Y, Wang Y, Sang W, Wang C, Zhang Y, Zhang H, Liu P, Liu M, Sun H. β-Sitosterol modulates osteogenic and adipogenic balance in BMSCs to suppress osteoporosis via regulating mTOR-IMP1-Adipoq axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156400. [PMID: 39848018 DOI: 10.1016/j.phymed.2025.156400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Osteoporosis (OP) is a prevalent global health concern, impacting millions of individuals, especially the elderly. The etiology of senile OP is associated with the imbalance of osteogenic and adipogenic differentiation in the bone marrow mesenchymal stem cells (BMSCs). The imbalance of BMSCs differentiation fate will leading to bone loss and lipids accumulation. β-sitosterol, a naturally occurring phytosterol which is abundant in plants and has a similar structure to cholesterol, demonstrates diverse bioactivities, including lipid-lowering effect and osteogenesis-inducing effects. These effects indicate that β-sitosterol might have anti-OP effects. Nevertheless, the precise mechanism underlying β-sitosterol's anti-osteoporotic efficacy via modulating BMSCs differentiation fate remains obscure. PURPOSE This study endeavors to elucidate whether β-sitosterol has the potential to augment the osteogenic differentiation of BMSCs while mitigating their adipogenic differentiation, thereby exerting an anti-OP effect; and to reveal its molecular mechanisms of action. METHODS In this study, a dosage form HP-β-cyclodextrin-coated β-sitosterol was developed for intragastric administration in mice to enhancing its bioavailability. Subsequently by using an integrative approach encompassing bioinformatics, computer molecular simulations, high-throughput sequencing, and in vitro/vivo as well as in-tube experiments, we investigated the anti-osteoporotic and bone healing effects of β-sitosterol and delineated its underlying mechanisms. RESULTS Our findings demonstrate that β-sitosterol exhibits anti-osteoporotic and bone healing effects both in vitro and in vivo by modulating the osteogenic and adipogenic differentiation of BMSCs. Mechanistically, these effects are mediated through the direct inhibition of mTOR's kinase activity independent of mediating autophagy, leading to the suppression of the mTOR-IMP1-Adipoq axis in BMSCs. CONCLUSION These results unveil β-sitosterol as a promising therapeutic agent for OP, shedding light on its underlying mechanisms. This research contributes potential candidates for diagnostic and therapeutic interventions in the realm of OP.
Collapse
Affiliation(s)
- Hao Li
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Ying Gong
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yanna Wang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wanyu Sang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yukun Zhang
- Three Gorges Medical College, Wanzhou, Chongqing, China
| | - Hanrui Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Peixuan Liu
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Mozhen Liu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Huijun Sun
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.
| |
Collapse
|
2
|
Yuan Y, Mao Y, Sun B, Chen C. Injectable Fibrocartilage-Forming Cores Enhance Bone-Tendon Healing in a Rat Rotator Cuff Model. Am J Sports Med 2025; 53:66-79. [PMID: 39741471 DOI: 10.1177/03635465241300138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
BACKGROUND After surgical repair of rotator cuff (RC) tears, the torn tendon heals unsatisfactorily to the greater tuberosity owing to limited regeneration of the bone-tendon (BT) insertion. This situation motivates the need for new interventions to enhance BT healing in the RC repair site. PURPOSE To develop injectable fibrocartilage-forming cores by tethering fibroblast growth factor 18 (FGF18) on acellular fibrocartilage matrix microparticles (AFM-MPs) and evaluate their efficacy on BT healing. STUDY DESIGN Controlled laboratory study. METHODS We harvested normal fibrocartilage tissue from the porcine RC insertion, after which it was decellularized and then micronized for fabricating AFM-MPs. The collagen-binding domain was fused into the N-terminus of FGF18 to synthesize recombinant FGF18 (CBD-FGF18), which was tethered to the collagen fibers of AFM-MPs to prepare the injectable fibrocartilage-forming cores (CBD-FGF18@AFM-MPs). After examining the influence of the CBD-FGF18@AFM-MPs on the viability and chondrogenic differentiation of bone marrow mesenchymal stem cells in vitro, we determined the function of the CBD-FGF18@AFM-MPs on BT healing in a rat RC tear model. A total of 80 Sprague-Dawley rats with RC injuries were randomly assigned to 4 supplemental treatments during RC repair: saline injection (control group), AFM-MPs injection, natural FGF18@AFM-MPs injection, and CBD-FGF18@AFM-MPs injection. At 4 and 8 weeks postoperatively, the harvested RC specimens were evaluated via micro-computed tomography, histologic staining, and mechanical testing. RESULTS In vitro, the CBD-FGF18@AFM-MPs were highly biomimetic, suitable for cell growth and proliferation, and superior in stimulating chondrogenesis. In vivo micro-computed tomography results showed that the CBD-FGF18@AFM-MPs group had significantly more new bone formation and better bone remodeling than the other 3 groups. Histologically, at 4 and 8 weeks postoperatively, the CBD-FGF18@AFM-MPs group had the best continuity of the BT insertion with regular collagen alignment and extensive fibrocartilage regeneration. Importantly, at 8 weeks postoperatively, the RC specimens from the CBD-FGF18@AFM-MPs group presented the highest failure load and stiffness. CONCLUSION The injectable fibrocartilage-forming cores provide a new biological intervention to promote RC healing. CLINICAL RELEVANCE The injectable fibrocartilage-forming cores may be a new complementary treatment for surgical repair of RC tears.
Collapse
Affiliation(s)
- Yuhao Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yiyang Mao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Buhua Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Can Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Chen Z, Zhang J, Lee FY, Kyriakides TR. Bone-derived extracellular matrix hydrogel from thrombospondin-2 knock-out mice for bone repair. Acta Biomater 2024; 186:85-94. [PMID: 39134130 PMCID: PMC11500023 DOI: 10.1016/j.actbio.2024.08.011] [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: 04/10/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Bone extracellular matrix (ECM) has been shown to mimic aspects of the tissue's complex microenvironment, suggesting its potential role in promoting bone repair. However, current ECM-based therapies suffer from limitations such as inefficient scale-up, lack of mechanical integrity, and sub-optimal efficacy. Here, we fabricated hydrogels from decellularized ECM (dECM) from wild type (WT) and thrombospondin-2 knock-out (TSP2KO) mouse bones. TSP2KO bone ECM hydrogel was found to have distinct mechanical properties and collagen fibril assembly from WT. Furthermore, TSP2KO hydrogel promoted mesenchymal stem cell (MSC) attachment, spreading, and invasion in vitro. Similarly, it promoted formation of tube-like structures by human umbilical vein endothelial cells (HUVECs). When applied to a murine calvarial defect model, TSP2KO hydrogel enhanced repair, in part, due to increased angiogenesis. Our study suggests the pro-angiogenic therapeutic potential of TSP2KO bone ECM hydrogel in bone repair. STATEMENT OF SIGNIFICANCE: The study describes the first successful preparation of a novel hydrogel made from decellularized bones from wild-type mice and mice lacking thrombospondin-2 (TSP2). Hydrogels from TSP2 knock-out (TSP2KO) bones have unique characteristics in structure and biomechanics. These gels interacted well with cells in vitro and helped repair damaged bone in a mouse model. Therefore, TSP2KO bone-derived hydrogel has translational potential for accelerating repair of bone defects that are otherwise difficult to heal. This study not only creates a new material with promise for accelerated healing, but also validates tunability of native biomaterials by genetic engineering.
Collapse
Affiliation(s)
- Zhuoyue Chen
- Department of Pathology, Yale University, New Haven, CT 06519, USA; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA
| | - Junqi Zhang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, CT 06519, USA
| | - Themis R Kyriakides
- Department of Pathology, Yale University, New Haven, CT 06519, USA; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA.
| |
Collapse
|
4
|
Wu Y, Ji Y, Lyu Z. 3D printing technology and its combination with nanotechnology in bone tissue engineering. Biomed Eng Lett 2024; 14:451-464. [PMID: 38645590 PMCID: PMC11026358 DOI: 10.1007/s13534-024-00350-x] [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: 06/05/2023] [Revised: 12/18/2023] [Accepted: 12/30/2023] [Indexed: 04/23/2024] Open
Abstract
With the graying of the world's population, the morbidity of age-related chronic degenerative bone diseases, such as osteoporosis and osteoarthritis, is increasing yearly, leading to an increased risk of bone defects, while current treatment methods face many problems, such as shortage of grafts and an incomplete repair. Therefore, bone tissue engineering offers an alternative solution for regenerating and repairing bone tissues by constructing bioactive scaffolds with porous structures that provide mechanical support to damaged bone tissue while promoting angiogenesis and cell adhesion, proliferation, and activity. 3D printing technology has become the primary scaffold manufacturing method due to its ability to precisely control the internal pore structure and complex spatial shape of bone scaffolds. In contrast, the fast development of nanotechnology has provided more possibilities for the internal structure and biological function of scaffolds. This review focuses on the application of 3D printing technology in bone tissue engineering and nanotechnology in the field of bone tissue regeneration and repair, and explores the prospects for the integration of the two technologies.
Collapse
Affiliation(s)
- Yuezhou Wu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001 China
| | - Yucheng Ji
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127 China
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001 China
| |
Collapse
|
5
|
Evrard R, Manon J, Rafferty C, Fieve L, Cornu O, Kirchgesner T, Lecouvet FE, Schubert T, Lengele B. Vascular study of decellularized porcine long bones: Characterization of a tissue engineering model. Bone 2024; 182:117073. [PMID: 38493932 DOI: 10.1016/j.bone.2024.117073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION Massive bone allografts enable the reconstruction of critical bone defects in numerous conditions (e.g. tumoral, infection or trauma). Unfortunately, their biological integration remains insufficient and the reconstruction may suffer from several postoperative complications. Perfusion-decellularization emerges as a tissue engineering potential solution to enhance osseointegration. Therefore, an intrinsic vascular study of this novel tissue engineering tool becomes essential to understand its efficacy and applicability. MATERIAL AND METHODS 32 porcine long bones (humeri and femurs) were used to assess the quality of their vascular network prior and after undergoing a perfusion-decellularization protocol. 12 paired bones were used to assess the vascular matrix prior (N = 6) and after our protocol (N = 6) by immunohistochemistry. Collagen IV, Von Willebrand factor and CD31 were targeted then quantified. The medullary macroscopic vascular network was evaluated with 12 bones: 6 were decellularized and the other 6 were, as control, not treated. All 12 underwent a contrast-agent injection through the nutrient artery prior an angio CT-scan acquisition. The images were processed and the length of medullary vessels filled with contrast agent were measured on angiographic cT images obtained in control and decellularized bones by 4 independent observers to evaluate the vascular network preservation. The microscopic cortical vascular network was evaluated on 8 bones: 4 control and 4 decellularized. After injection of gelatinous fluorochrome mixture (calcein green), non-decalcified fluoroscopic microscopy was performed in order to assess the perfusion quality of cortical vascular lacunae. RESULTS The continuity of the microscopic vascular network was assessed with Collagen IV immunohistochemistry (p-value = 0.805) while the decellularization quality was observed through CD31 and Von Willebrand factor immunohistochemistry (p-values <0.001). The macroscopic vascular network was severely impaired after perfusion-decellularization; nutrient arteries were still patent but the amount of medullary vascular channels measured was significantly higher in the control group compared to the decellularized group (p-value <0.001). On average, the observers show good agreement on these results, except in the decellularized group where more inter-observer discrepancies were observed. The microscopic vascular network was observed with green fluoroscopic signal in almost every canals and lacunae of the bone cortices, in three different bone locations (proximal metaphysis, diaphysis and distal metaphysis). CONCLUSION Despite the aggressiveness of the decellularization protocol on medullary vessels, total porcine long bones decellularized by perfusion retain an acellular cortical microvascular network. By injection through the intact nutrient arteries, this latter vascular network can still be used as a total bone infusion access for bone tissue engineering in order to enhance massive bone allografts prior implantation.
Collapse
Affiliation(s)
- R Evrard
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium.
| | - J Manon
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - C Rafferty
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium
| | - L Fieve
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium
| | - O Cornu
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - T Kirchgesner
- Département d'Imagerie Médicale, Institut de Recherche Expérimentale et Clinique (Pôle IMAG), Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - F E Lecouvet
- Département d'Imagerie Médicale, Institut de Recherche Expérimentale et Clinique (Pôle IMAG), Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - T Schubert
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - B Lengele
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| |
Collapse
|
6
|
Kim YH, Kanczler JM, Lanham S, Rawlings A, Roldo M, Tozzi G, Dawson JI, Cidonio G, Oreffo ROC. Biofabrication of nanocomposite-based scaffolds containing human bone extracellular matrix for the differentiation of skeletal stem and progenitor cells. Biodes Manuf 2024; 7:121-136. [PMID: 38497056 PMCID: PMC10937808 DOI: 10.1007/s42242-023-00265-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/13/2023] [Indexed: 03/19/2024]
Abstract
Autograft or metal implants are routinely used in skeletal repair. However, they fail to provide long-term clinical resolution, necessitating a functional biomimetic tissue engineering alternative. The use of native human bone tissue for synthesizing a biomimetic material ink for three-dimensional (3D) bioprinting of skeletal tissue is an attractive strategy for tissue regeneration. Thus, human bone extracellular matrix (bone-ECM) offers an exciting potential for the development of an appropriate microenvironment for human bone marrow stromal cells (HBMSCs) to proliferate and differentiate along the osteogenic lineage. In this study, we engineered a novel material ink (LAB) by blending human bone-ECM (B) with nanoclay (L, Laponite®) and alginate (A) polymers using extrusion-based deposition. The inclusion of the nanofiller and polymeric material increased the rheology, printability, and drug retention properties and, critically, the preservation of HBMSCs viability upon printing. The composite of human bone-ECM-based 3D constructs containing vascular endothelial growth factor (VEGF) enhanced vascularization after implantation in an ex vivo chick chorioallantoic membrane (CAM) model. The inclusion of bone morphogenetic protein-2 (BMP-2) with the HBMSCs further enhanced vascularization and mineralization after only seven days. This study demonstrates the synergistic combination of nanoclay with biomimetic materials (alginate and bone-ECM) to support the formation of osteogenic tissue both in vitro and ex vivo and offers a promising novel 3D bioprinting approach to personalized skeletal tissue repair. Graphic abstract Supplementary Information The online version contains supplementary material available at 10.1007/s42242-023-00265-z.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
| | - Janos M. Kanczler
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
| | - Stuart Lanham
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
| | - Andrew Rawlings
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
| | - Marta Roldo
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, PO1 2DT UK
| | - Gianluca Tozzi
- School of Engineering, Faculty of Engineering and Science, University of Greenwich, Greenwich, ME4 4TB UK
| | - Jonathan I. Dawson
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
| | - Gianluca Cidonio
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
- Center for Life Nano- and Neuro-Science (CLN2S), Italian Institute of Technology, 00161 Rome, Italy
| | - Richard O. C. Oreffo
- Faculty of Medicine, Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|
7
|
Dehghani S, Aghaee Z, Soleymani S, Tafazoli M, Ghabool Y, Tavassoli A. An overview of the production of tissue extracellular matrix and decellularization process. Cell Tissue Bank 2024; 25:369-387. [PMID: 37812368 DOI: 10.1007/s10561-023-10112-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/09/2023] [Indexed: 10/10/2023]
Abstract
Thousands of patients need an organ transplant yearly, while only a tiny percentage have this chance to receive a tissue/organ transplant. Nowadays, decellularized animal tissue is one of the most widely used methods to produce engineered scaffolds for transplantation. Decellularization is defined as physically or chemically removing cellular components from tissues while retaining structural and functional extracellular matrix (ECM) components and creating an ECM-derived scaffold. Then, decellularized scaffolds could be reseeded with different cells to fabricate an autologous graft. Effective decellularization methods preserve ECM structure and bioactivity through the application of the agents and techniques used throughout the process. The most valuable agents for the decellularization process depend on biological properties, cellular density, and the thickness of the desired tissue. ECM-derived scaffolds from various mammalian tissues have been recently used in research and preclinical applications in tissue engineering. Many studies have shown that decellularized ECM-derived scaffolds could be obtained from tissues and organs such as the liver, cartilage, bone, kidney, lung, and skin. This review addresses the significance of ECM in organisms and various decellularization agents utilized to prepare the ECM. Also, we describe the current knowledge of the decellularization of different tissues and their applications.
Collapse
Affiliation(s)
- Shima Dehghani
- Department of Biology, Kavian Institute of Higher Education, Mashhad, Iran
| | - Zahra Aghaee
- Department of Biology, Kavian Institute of Higher Education, Mashhad, Iran
| | - Safoura Soleymani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974, Iran
| | - Maryam Tafazoli
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974, Iran
| | - Yasin Ghabool
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Amin Tavassoli
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, 9177948974, Iran.
| |
Collapse
|
8
|
Mohammadi A, Koruji M, Azami M, Shabani R, Mohandesnezhad S, Bashiri Z, Asgari H. Polycaprolactone/Testicular Extracellular Matrix/Graphene Oxide-Based Electrospun Tubular Scaffolds for Reproductive Medicine: Biomimetic Architecture of Seminiferous Tubules. Macromol Biosci 2024; 24:e2300342. [PMID: 37729950 DOI: 10.1002/mabi.202300342] [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: 07/26/2023] [Revised: 09/06/2023] [Indexed: 09/22/2023]
Abstract
Numerous scaffolds are developed in the field of testicular bioengineering. However, effectively replicating the spatial characteristics of native tissue, poses a challenge in maintaining the requisite cellular arrangement essential for spermatogenesis. In order to mimic the structural properties of seminiferous tubules, the objective is to fabricate a biocompatible tubular scaffold. Following the decellularization process of the testicular tissue, validation of cellular remnants' elimination from the specimens is conducted using 4',6-diamidino-2-phenylindole staining, hematoxylin and eosin staining, and DNA content analysis. The presence of extracellular matrix (ECM) components is confirmed through Alcian blue, Orcein, and Masson's trichrome staining techniques. The electrospinning technique is employed to synthesize the scaffolds using polycaprolactone (PCL), extracted ECM, and varying concentrations of graphene oxide (GO) (0.5%, 1%, and 2%). Subsequently, comprehensive evaluations are performed to assess the properties of the synthetic scaffolds. These evaluations encompass Fourier-transform infrared spectroscopy, scanning electron microscopy imaging, scaffold degradation testing, mechanical behavior analysis, methylthiazolyldiphenyl-tetrazolium bromide assay, and in vivo biocompatibility assessment. The PCL/decellularized extracellular matrix with 0.5% GO formulation exhibits superior fiber morphology and enhanced mechanical properties, and outperforms other groups in terms of in vitro biocompatibility. Consequently, these scaffolds present a viable option for implementation in "in vitro spermatogenesis" procedures, holding promise for future sperm production from spermatogonial cells.
Collapse
Affiliation(s)
- Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 88770048, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 88770048, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
- Joint Reconstruction Research Center (JRRC), Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Ronak Shabani
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Sanam Mohandesnezhad
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, 6517789971, Iran
- Omid Fertility & Infertility Clinic, Hamedan, 6516796198, Iran
| | - Hamidreza Asgari
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 88770048, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| |
Collapse
|
9
|
Yuan Y, Xu Y, Mao Y, Liu H, Ou M, Lin Z, Zhao R, Long H, Cheng L, Sun B, Zhao S, Zeng M, Lu B, Lu H, Zhu Y, Chen C. Three Birds, One Stone: An Osteo-Microenvironment Stage-Regulative Scaffold for Bone Defect Repair through Modulating Early Osteo-Immunomodulation, Middle Neovascularization, and Later Osteogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306428. [PMID: 38060833 PMCID: PMC10853759 DOI: 10.1002/advs.202306428] [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: 09/06/2023] [Revised: 10/28/2023] [Indexed: 02/10/2024]
Abstract
In order to repair critical-sized bone defects, various polylactic acid-glycolic acid (PLGA)-based hybrid scaffolds are successfully developed as bone substitutes. However, the byproducts of these PLGA-based scaffolds are known to acidify the implanted site, inducing tiresome acidic inflammation. Moreover, these degradation productions cannot offer an osteo-friendly microenvironment at the implanted site, matching natural bone healing. Herein, inspired by bone microenvironment atlas of natural bone-healing process, an osteo-microenvironment stage-regulative scaffold (P80/D10/M10) is fabricated by incorporating self-developed decellularized bone matrix microparticles (DBM-MPs) and multifunctional magnesium hydroxide nanoparticles (MH-NPs) into PLGA with an optimized proportion using low-temperature rapid prototyping (LT-RP) 3D-printing technology. The cell experiments show that this P80/D10/M10 exhibits excellent properties in mechanics, biocompatibility, and biodegradability, meanwhile superior stimulations in osteo-immunomodulation, angiogenesis, and osteogenesis. Additionally, the animal experiments determined that this P80/D10/M10 can offer an osteo-friendly microenvironment in a stage-matched pattern for enhanced bone regeneration, namely, optimization of early inflammation, middle neovascularization, and later bone formation. Furthermore, transcriptomic analysis suggested that the in vivo performance of P80/D10/M10 on bone defect repair is mostly attributed to regulating artery development, bone development, and bone remodeling. Overall, this study reveals that the osteo-microenvironment stage-regulative scaffold provides a promising treatment for bone defect repair.
Collapse
Affiliation(s)
- Yuhao Yuan
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yan Xu
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
- Department of Sports MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yiyang Mao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
| | - Hongbin Liu
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Minning Ou
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zhangyuan Lin
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ruibo Zhao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Haitao Long
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Liang Cheng
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Buhua Sun
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Shushan Zhao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ming Zeng
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Bangbao Lu
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hongbin Lu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
- Department of Sports MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yong Zhu
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Can Chen
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
| |
Collapse
|
10
|
Evrard R, Manon J, Maistriaux L, Rafferty C, Fieve L, Heller U, Cornu O, Gianello P, Schubert T, Lengele B. Decellularization of Massive Bone Allografts By Perfusion: A New Protocol for Tissue Engineering. Tissue Eng Part A 2024; 30:31-44. [PMID: 37698880 DOI: 10.1089/ten.tea.2023.0182] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023] Open
Abstract
In terms of large bone defect reconstructions, massive bone allografts may sometimes be the only solution. However, they are still burdened with a high postoperative complication rate. Our hypothesis is that the immunogenicity of residual cells in the graft is involved in this issue. Decellularization by perfusion might therefore be the answer to process and create more biologically effective massive bone allografts. Seventy-two porcine bones were used to characterize the efficiency of our sodium hydroxide-based decellularization protocol. A sequence of solvent perfusion through each nutrient artery was set up to ensure the complete decellularization of whole long bones. Qualitative (histology and immunohistochemistry [IHC]) and quantitative (fluoroscopic absorbance and enzyme-linked immunosorbent assay) evaluations were performed to assess the decellularization and the preservation of the extracellular matrix in the bone grafts. Cytotoxicity and compatibility were also tested. Comparatively to nontreated bones, our experiments showed a very high decellularization quality, demonstrating that perfusion is mandatory to achieve an entire decellularization. Moreover, results showed a good preservation of the bone composition and microarchitecture, Haversian systems and vascular network included. This protocol reduces the human leukocyte antigen antigenic load of the graft by >50%. The majority of measured growth factors is still present in the same amount in the decellularized bones compared to the nontreated bones. Histology and IHC show that the bones were cell compatible, noncytotoxic, and capable of inducing osteoblastic differentiation of mesenchymal stem cells. Our decellularization/perfusion protocol allowed to create decellularized long bone graft models, thanks to their inner vascular network, ready for in vivo implantation or to be further used as seeding matrices.
Collapse
Affiliation(s)
- Robin Evrard
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Julie Manon
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Louis Maistriaux
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Chiara Rafferty
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Lies Fieve
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
| | - Ugo Heller
- Centre Hospitalo-Universitaire Necker Enfants Malades, Service de Chirurgie Maxillo-Faciale et Reconstructrice, Paris, France
| | - Olivier Cornu
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Pierre Gianello
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Bruxelles, Belgique
| | - Thomas Schubert
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| | - Benoit Lengele
- Secteur des Sciences de la Santé, Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Bruxelles, Belgique
- Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, Bruxelles, Belgique
| |
Collapse
|
11
|
Cowell K, Statham P, Sagoo GS, Chandler JH, Herbert A, Rooney P, Wilcox RK, Fermor HL. Cost-effectiveness of decellularised bone allograft compared with fresh-frozen bone allograft for acetabular impaction bone grafting during a revision hip arthroplasty in the UK. BMJ Open 2023; 13:e067876. [PMID: 37802609 PMCID: PMC10565200 DOI: 10.1136/bmjopen-2022-067876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
OBJECTIVES Fresh-frozen allograft is the gold-standard bone graft material used during revision hip arthroplasty. However, new technology has been developed to manufacture decellularised bone with potentially better graft incorporation. As these grafts cost more to manufacture, the aim of this cost-effectiveness study was to estimate whether the potential health benefit of decellularised bone allograft outweighs their increased cost. STUDY DESIGN A Markov model was constructed to estimate the costs and the quality-adjusted life years of impaction bone grafting during a revision hip arthroplasty. SETTING This study took the perspective of the National Health Service in the UK. PARTICIPANTS The Markov model includes patients undergoing a revision hip arthroplasty in the UK. INTERVENTION Impaction bone grafting during a revision hip arthroplasty using either decellularised bone allograft or fresh-frozen allograft. MEASURES Outcome measures included: total costs and quality-adjusted life years of both interventions over the lifetime of the model; and incremental cost-effectiveness ratios for both graft types, using base case parameters, univariate sensitivity analysis and probabilistic analysis. RESULTS The incremental cost-effectiveness ratio for the base case model was found to be £270 059 per quality-adjusted life year. Univariate sensitivity analysis found that changing the discount rate, the decellularised bone graft cost, age of the patient cohort and the revision rate all had a significant effect on the incremental cost-effectiveness ratio. CONCLUSIONS As there are no clinical studies of impaction bone grafting using a decellularised bone allograft, there is a high level of uncertainty around the costs of producing a decellularised bone allograft and the potential health benefits. However, if a decellularised bone graft was manufactured for £2887 and lowered the re-revision rate to less than 64 cases per year per 10 000 revision patients, then it would most likely be cost-effective compared with fresh-frozen allograft.
Collapse
Affiliation(s)
- Kern Cowell
- Institute of Medical and Biological Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK
| | - Patrick Statham
- Institute of Medical and Biological Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK
| | - Gurdeep Singh Sagoo
- Academic Unit of Health Economics, University of Leeds, Leeds, UK
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - James H Chandler
- Institute of Design, Robotics and Optimisation, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK
| | - Anthony Herbert
- Institute of Medical and Biological Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK
| | - Paul Rooney
- Research and Development, NHS Blood and Transplant Tissue and Eye Services, Speke, UK
| | - Ruth K Wilcox
- Institute of Medical and Biological Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, UK
| | - Hazel L Fermor
- Institute of Medical and Biological Eningeering, School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
12
|
Liang C, Liao L, Tian W. Advances Focusing on the Application of Decellularized Extracellular Matrix in Periodontal Regeneration. Biomolecules 2023; 13:673. [PMID: 37189420 PMCID: PMC10136219 DOI: 10.3390/biom13040673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
The decellularized extracellular matrix (dECM) is capable of promoting stem cell proliferation, migration, adhesion, and differentiation. It is a promising biomaterial for application and clinical translation in the field of periodontal tissue engineering as it most effectively preserves the complex array of ECM components as they are in native tissue, providing ideal cues for regeneration and repair of damaged periodontal tissue. dECMs of different origins have different advantages and characteristics in promoting the regeneration of periodontal tissue. dECM can be used directly or dissolved in liquid for better flowability. Multiple ways were developed to improve the mechanical strength of dECM, such as functionalized scaffolds with cells that harvest scaffold-supported dECM through decellularization or crosslinked soluble dECM that can form injectable hydrogels for periodontal tissue repair. dECM has found recent success in many periodontal regeneration and repair therapies. This review focuses on the repairing effect of dECM in periodontal tissue engineering, with variations in cell/tissue sources, and specifically discusses the future trend of periodontal regeneration and the future role of soluble dECM in entire periodontal tissue regeneration.
Collapse
Affiliation(s)
| | - Li Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Sichuan 610041, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Sichuan 610041, China
| |
Collapse
|
13
|
Deng Z, Zhu W, Lu B, Li M, Xu D. A Slotted Decellularized Osteochondral Scaffold With Layer-Specific Release of Stem Cell Differentiation Stimulators Enhances Cartilage and Bone Regeneration in Osteochondral Defects in a Rabbit Model. Am J Sports Med 2022; 50:3390-3405. [PMID: 36122351 DOI: 10.1177/03635465221114412] [Citation(s) in RCA: 4] [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 Owing to the disappointing regenerative ability of osteochondral tissue, without treatment an osteochondral defect would progress to osteoarthritis. This situation motivates the need for new strategies to enhance the regeneration of osteochondral defects. PURPOSE To develop a tissue-engineering scaffold by tethering bone morphogenetic protein 2 (BMP2) and transforming growth factor beta 3 (TGFβ3) in a layer-specific manner on a slotted decellularized osteochondral matrix (SDOM) and to evaluate the efficacy of this scaffold for osteochondral regeneration. STUDY DESIGN Controlled laboratory study. METHODS Normal osteochondral tissue from the rabbit patellofemoral groove was sectioned into a slot shape and decellularized for fabricating an SDOM. The collagen-binding domain (CBD) was fused into the N-terminus of BMP2 or TGFβ3 to synthesize 2 recombinant growth factors (GFs) (CBD-BMP2 or CBD-TGFβ3), which were tethered to the bone layer and cartilage layer, respectively, of the SDOM to prepare a tissue-engineering scaffold (namely, CBD-GFs/SDOM). After examining the influence of the CBD-GFs/SDOM on the viability and layer-specific differentiation of bone marrow mesenchymal stem cells in vitro, we determined the regeneration potential of the CBD-GFs/SDOM on osteochondral regeneration in a rabbit model. A total of 72 New Zealand White rabbits with a cylindrical osteochondral defect in the patellofemoral groove were randomly assigned to 3 groups: defect only (control [CTL] group), defect patched with an SDOM (SDOM group), and defect patched with the CBD-GFs/SDOM (CBD-GFs/SDOM group). At 6 or 12 weeks postoperatively, the rabbits were euthanized to harvest the knee joint, which was then evaluated via gross observation, micro-computed tomography, histological staining, and mechanical testing. RESULTS In vitro, the CBD-GFs/SDOM was noncytotoxic, showed high biomimetics with normal osteochondral tissue, was suitable for cell adhesion and growth, and had good layer-specific ability in inducing stem cell differentiation. Macroscopic images showed that the CBD-GFs/SDOM group had significantly better osteochondral regeneration than the CTL and SDOM groups had. Micro-computed tomography demonstrated that much more bony tissue was formed at the defect sites in the CBD-GFs/SDOM group compared with the defect sites in the CTL or SDOM group. Histological analysis showed that the CBD-GFs/SDOM group had a significant enhancement in osteochondral regeneration at 6 and 12 weeks postoperatively in comparison with the CTL or SDOM group. At 12 weeks postoperatively, the mechanical properties of reparative tissue were significantly better in the CBD-GFs/SDOM group than in the other groups. CONCLUSION The CBD-GFs/SDOM is a promising scaffold for osteochondral regeneration. CLINICAL RELEVANCE The findings of this study indicated that the CBD-GFs/SDOM is an excellent candidate for reconstructing osteochondral defects, which may be translated for clinical use in the future.
Collapse
Affiliation(s)
- Zhenhan Deng
- Department of Sports Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Weimin Zhu
- Department of Sports Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Muzhi Li
- Department of Rehabilitation, Second Xiangya Hospital, Central South University, Changsha, China
| | - Daqi Xu
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China.,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Neunaber C, Dalinghaus C, Bundkirchen K, Toumpaniari S, Gladitz LM, Joda A, Morticelli L, Krettek C, Korossis S. Towards the development of osteochondral allografts with reduced immunogenicity. J Mech Behav Biomed Mater 2022; 133:105359. [PMID: 35841749 DOI: 10.1016/j.jmbbm.2022.105359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022]
Abstract
Nowadays, repair and replacement of hyaline articular cartilage still challenges orthopedic surgery. Using a graft of decellularized articular cartilage as a structural scaffold is considered as a promising therapy. So far, successful cell removal has only been possible for small samples with destruction of the macrostructure or loss of biomechanics. Our aim was to develop a mild, enzyme-free chemical decellularization procedure while preserving the biomechanical properties of cartilage. Porcine osteochondral cylinders (diameter: 12 mm; height: 10 mm) were divided into four groups: Native plugs (NA), decellularized plugs treated with PBS, Triton-X-100 and SDS (DC), and plugs additionally treated with freeze-thaw-cycles of -20 °C, -80 °C or shock freezing in nitrogen (N2) before decellularization. In a non-decalcified HE stain the decellularization efficiency (cell removal, cell size, depth of decellularization) was calculated. For biomechanics the elastic and compression modulus, transition and failure strain as well as transition and failure stress were evaluated. The -20 °C, -80 °C, and N2 groups showed a complete decellularization of the superficial and middle zone. In the deep zone cells could not be removed in any experimental group. The biomechanical analysis showed only a reduced elastic modulus in all decellularized samples. No significant differences were found for the other biomechanical parameters.
Collapse
Affiliation(s)
- Claudia Neunaber
- Trauma Department, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Catharina Dalinghaus
- Trauma Department, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Katrin Bundkirchen
- Trauma Department, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Sotiria Toumpaniari
- Cardiopulmonary Regenerative Engineering (CARE) Group, Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Holywell Park, Loughborough University, LE11 3TU, Loughborough, UK.
| | - Luisa Marilena Gladitz
- Trauma Department, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Akram Joda
- Cardiopulmonary Regenerative Engineering (CARE) Group, Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Holywell Park, Loughborough University, LE11 3TU, Loughborough, UK; Higher Colleges of Technology, Faculty of Engineering Technology and Science, Dubai, United Arab Emirates.
| | - Lucrezia Morticelli
- Lower Saxony Centre for Biomedical Engineering Implant Research & Development, Hannover Medical School, Stadtfelddamm 34, 30625, Hannover, Germany.
| | - Christian Krettek
- Trauma Department, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Sotirios Korossis
- Cardiopulmonary Regenerative Engineering (CARE) Group, Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Holywell Park, Loughborough University, LE11 3TU, Loughborough, UK; Lower Saxony Centre for Biomedical Engineering Implant Research & Development, Hannover Medical School, Stadtfelddamm 34, 30625, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
15
|
Browe DC, Díaz-Payno PJ, Freeman FE, Schipani R, Burdis R, Ahern DP, Nulty JM, Guler S, Randall LD, Buckley CT, Brama PA, Kelly DJ. Bilayered extracellular matrix derived scaffolds with anisotropic pore architecture guide tissue organization during osteochondral defect repair. Acta Biomater 2022; 143:266-281. [PMID: 35278686 DOI: 10.1016/j.actbio.2022.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022]
Abstract
While some clinical advances in cartilage repair have occurred, osteochondral (OC) defect repair remains a significant challenge, with current scaffold-based approaches failing to recapitulate the complex, hierarchical structure of native articular cartilage (AC). To address this need, we fabricated bilayered extracellular matrix (ECM)-derived scaffolds with aligned pore architectures. By modifying the freeze-drying kinetics and controlling the direction of heat transfer during freezing, it was possible to produce anisotropic scaffolds with larger pores which supported homogenous cellular infiltration and improved sulfated glycosaminoglycan deposition. Neo-tissue organization in vitro could also be controlled by altering scaffold pore architecture, with collagen fibres aligning parallel to the long-axis of the pores within scaffolds containing aligned pore networks. Furthermore, we used in vitro and in vivo assays to demonstrate that AC and bone ECM derived scaffolds could preferentially direct the differentiation of mesenchymal stromal cells (MSCs) towards either a chondrogenic or osteogenic lineage respectively, enabling the development of bilayered ECM scaffolds capable of spatially supporting unique tissue phenotypes. Finally, we implanted these scaffolds into a large animal model of OC defect repair. After 6 months in vivo, scaffold implantation was found to improve cartilage matrix deposition, with collagen fibres preferentially aligning parallel to the long axis of the scaffold pores, resulting in a repair tissue that structurally and compositionally was more hyaline-like in nature. These results demonstrate how scaffold architecture and composition can be spatially modulated to direct the regeneration of complex interfaces such as the osteochondral unit, enabling their use as cell-free, off-the-shelf implants for joint regeneration. STATEMENT OF SIGNIFICANCE: The architecture of the extracellular matrix, while integral to tissue function, is often neglected in the design and evaluation of regenerative biomaterials. In this study we developed a bilayered scaffold for osteochondral defect repair consisting of tissue-specific extracellular matrix (ECM)-derived biomaterials to spatially direct stem/progenitor cell differentiation, with a tailored pore microarchitecture to promote the development of a repair tissue that recapitulates the hierarchical structure of native AC. The use of this bilayered scaffold resulted in improved tissue repair outcomes in a large animal model, specifically the ability to guide neo-tissue organization and therefore recapitulate key aspects of the zonal structure of native articular cartilage. These bilayer scaffolds have the potential to become a new therapeutic option for osteochondral defect repair.
Collapse
|
16
|
Repair of osteochondral defects mediated by double-layer scaffolds with natural osteochondral-biomimetic microenvironment and interface. Mater Today Bio 2022; 14:100234. [PMID: 35308043 PMCID: PMC8924418 DOI: 10.1016/j.mtbio.2022.100234] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 12/12/2022]
Abstract
Tissue engineering provides a new approach for the treatment of osteochondral defects. However, the lack of an ideal double-layer scaffold with osteochondral-biomimetic microenvironment and interface similar to native articular tissue greatly limits clinical translation. Our current study developed a double-layer acellular osteochondral matrix (AOM) scaffold with natural osteochondral-biomimetic microenvironment and interface by integrating ultraviolet (UV) laser and decellularization techniques. The laser parameters were optimized to achieve a proper pore depth close to the osteochondral interface, which guaranteed complete decellularization, sufficient space for cell loading, and relative independence of the chondrogenic and osteogenic microenvironments. Gelatin-methacryloyl (GelMA) hydrogel was further used as the cell carrier to significantly enhance the efficiency and homogeneity of cell loading in the AOM scaffold with large pore structure. Additionally, in vitro results demonstrated that the components of the AOM scaffold could efficiently regulate the chondrogenic/osteogenic differentiations of bone marrow stromal cells (BMSCs) by activating the chondrogenic/osteogenic related pathways. Importantly, the AOM scaffolds combined with BMSC-laden GelMA hydrogel successfully realized tissue-specific repair of the osteochondral defects in a knee joint model of rabbit. The current study developed a novel double-layer osteochondral biomimetic scaffold and feasible strategy, providing strong support for the tissue-specific repair of osteochondral defects and its future clinical translation.
Collapse
|
17
|
Wan HY, Shin RLY, Chen JCH, Assunção M, Wang D, Nilsson SK, Tuan RS, Blocki A. Dextran sulfate-amplified extracellular matrix deposition promotes osteogenic differentiation of mesenchymal stem cells. Acta Biomater 2022; 140:163-177. [PMID: 34875356 DOI: 10.1016/j.actbio.2021.11.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022]
Abstract
The development of bone-like tissues in vitro that exhibit key features similar to those in vivo is needed to produce tissue models for drug screening and the study of bone physiology and disease pathogenesis. Extracellular matrix (ECM) is a predominant component of bone in vivo; however, as ECM assembly is sub-optimal in vitro, current bone tissue engineering approaches are limited by an imbalance in ECM-to-cell ratio. We amplified the deposition of osteoblastic ECM by supplementing dextran sulfate (DxS) into osteogenically induced cultures of human mesenchymal stem cells (MSCs). DxS, previously implicated to act as a macromolecular crowder, was recently demonstrated to aggregate and co-precipitate major ECM components, including collagen type I, thereby amplifying its deposition. This effect was re-confirmed for MSC cultures undergoing osteogenic induction, where DxS supplementation augmented collagen type I deposition, accompanied by extracellular osteocalcin accumulation. The resulting differentiated osteoblasts exhibited a more mature osteogenic gene expression profile, indicated by a strong upregulation of the intermediate and late osteogenic markers ALP and OCN, respectively. The associated cellular microenvironment was also enriched in bone morphogenetic protein 2 (BMP-2). Interestingly, the resulting decellularized matrices exhibited the strongest osteo-inductive effects on re-seeded MSCs, promoted cell proliferation, osteogenic marker expression and ECM calcification. Taken together, these findings suggest that DxS-mediated enhancement of osteogenic differentiation by MSCs is mediated by the amplified ECM, which is enriched in osteo-inductive factors. We have thus established a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with sequestration of osteo-inductive factors. STATEMENT OF SIGNIFICANCE: As extracellular matrix (ECM) assembly is significantly retarded in vitro, the imbalance in ECM-to-cell ratio hampers current in vitro bone tissue engineering approaches in their ability to faithfully resemble their in vivo counterpart. We addressed this limitation by leveraging a poly-electrolyte mediated co-assembly and amplified deposition of ECM during osteogenic differentiation of human mesenchymal stem cells (MSCs). The resulting pericelluar space in culture was enriched in organic and inorganic bone ECM components, as well as osteo-inductive factors, which promoted the differentiation of MSCs towards a more mature osteoblastic phenotype. These findings thus demonstrated a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with a closer recapitulation of the in vivo tissue niche.
Collapse
|
18
|
Zhang C, Xia D, Li J, Zheng Y, Weng B, Mao H, Mei J, Wu T, Li M, Zhao J. BMSCs and Osteoblast-Engineered ECM Synergetically Promotes Osteogenesis and Angiogenesis in an Ectopic Bone Formation Model. Front Bioeng Biotechnol 2022; 10:818191. [PMID: 35127662 PMCID: PMC8814575 DOI: 10.3389/fbioe.2022.818191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/04/2022] [Indexed: 12/16/2022] Open
Abstract
Bone mesenchymal stem cells (BMSCs) have been extensively used in bone tissue engineering because of their potential to differentiate into multiple cells, secrete paracrine factors, and attenuate immune responses. Biomaterials are essential for the residence and activities of BMSCs after implantation in vivo. Recently, extracellular matrix (ECM) modification with a favorable regenerative microenvironment has been demonstrated to be a promising approach for cellular activities and bone regeneration. The aim of the present study was to evaluate the effects of BMSCs combined with cell-engineered ECM scaffolds on osteogenesis and angiogenesis in vivo. The ECM scaffolds were generated by osteoblasts on the small intestinal submucosa (SIS) under treatment with calcium (Ca)-enriched medium and icariin (Ic) after decellularization. In a mouse ectopic bone formation model, the SIS scaffolds were demonstrated to reduce the immune response, and lower the levels of immune cells compared with those in the sham group. Ca/Ic-ECM modification inhibited the degradation of the SIS scaffolds in vivo. The generated Ca/Ic-SIS scaffolds ectopically promoted osteogenesis according to the results of micro-CT and histological staining. Moreover, BMSCs on Ca/Ic-SIS further increased the bone volume percentage (BV/TV) and bone density. Moreover, angiogenesis was also enhanced by the Ca/Ic-SIS scaffolds, resulting in the highest levels of neovascularization according to the data ofCD31 staining. In conclusion, osteoblast-engineered ECM under directional induction is a promising strategy to modify biomaterials for osteogenesis and angiogenesis. BMSCs synergetically improve the properties of ECM constructs, which may contribute to the repair of large bone defects.
Collapse
Affiliation(s)
- Chi Zhang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
- Medical Research Center, Ningbo City First Hospital, Ningbo, China
| | - Dongdong Xia
- Orthopedic Department, Ningbo City First Hospital, Ningbo, China
| | - Jiajing Li
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Yanan Zheng
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Bowen Weng
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Jing Mei
- Medical Research Center, Ningbo City First Hospital, Ningbo, China
| | - Tao Wu
- Cardiovascular Center, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Mei Li
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
- Ningbo Institute of Medical Sciences, Ningbo, China
- *Correspondence: Mei Li, ; Jiyuan Zhao,
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
- *Correspondence: Mei Li, ; Jiyuan Zhao,
| |
Collapse
|
19
|
Erickson CB, Hill R, Pascablo D, Kazakia G, Hansen K, Bahney C. A timeseries analysis of the fracture callus extracellular matrix proteome during bone fracture healing. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2021; 3:1-30. [PMID: 35765657 PMCID: PMC9236279 DOI: 10.36069/jols/20220601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
While most bones fully self-heal, certain diseases require bone allograft to assist with fracture healing. Bone allografts offer promise as treatments for such fractures due to their osteogenic properties. However, current bone allografts made of decellularized bone extracellular matrix (ECM) have high failure rates, and thus grafts which improve fracture healing outcomes are needed. Understanding specific changes to the ECM proteome during normal fracture healing would enable the identification of key proteins that could be used enhance osteogenicity of bone allograft. Here, we performed a timeseries analysis of the fracture callus in mice to investigate proteomic and mineralization changes to the ECM at key stages of fracture healing. We found that changes to the ECM proteome largely coincide with the distinct phases of fracture healing. Basement membrane proteins (AGRN, COL4, LAMA), cartilage proteins (COL2A1, ACAN), and collagen crosslinking enzymes (LOXL, PLOD, ITIH) were initially upregulated, followed by bone specific proteoglycans and collagens (IBSP, COL1A1). Various tissue proteases (MMP2, 9, 13, 14; CTSK, CTSG, ELANE) were expressed at different levels throughout fracture healing. These changes coordinated with mineralization of the fracture callus, which increased steeply during the initial stages of healing. Interestingly the later timepoint was characterized by a response to wound healing and high expression of clotting factors (F2, 7, 9, 10). We identified ELANE and ITIH2 as tissue remodeling enzymes having no prior known involvement with fracture healing. This data can be further mined to identify regenerative proteins for enhanced bone graft design.
Collapse
Affiliation(s)
- Christopher B. Erickson
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Ryan Hill
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Donna Pascablo
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA
| | - Galateia Kazakia
- Department of Radiology and Biomedical Imaging, University of California, San Francisco (UCSF), San Francisco, CA
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Chelsea Bahney
- Stedman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine. Vail, CO
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA
| |
Collapse
|
20
|
Shengnan Q, Bennett S, Wen W, Aiguo L, Jiake X. The role of tendon derived stem/progenitor cells and extracellular matrix components in the bone tendon junction repair. Bone 2021; 153:116172. [PMID: 34506992 DOI: 10.1016/j.bone.2021.116172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/22/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022]
Abstract
Fibrocartilage enthesis is the junction between bone and tendon with a typical characteristics of fibrocartilage transition zones. The regeneration of this transition zone is the bottleneck for functional restoration of bone tendon junction (BTJ). Biomimetic approaches, especially decellularized extracellular matrix (ECM) materials, are strategies which aim to mimic the components of tissues to the utmost extent, and are becoming popular in BTJ healing because of their ability not only to provide scaffolds to allow cells to attach and migrate, but also to provide a microenvironment to guide stem/progenitor cells lineage-specific differentiation. However, the cellular and molecular mechanisms of those approaches, especially the ECM proteins, remain unclear. For BTJ reconstruction, fibrocartilage regeneration is the key for good integrity of bone and tendon as well as its mechanical recovery, so the components which can guide stem cells to a chondrogenic commitment in biomimetic approaches might well be the key for fibrocartilage regeneration and eventually for the better BTJ healing. In this review, we firstly discuss the importance of cartilage-like formation in the healing process of BTJ. Next, we explore the possibility of tendon-derived stem/progenitor cells as cell sources for BTJ regeneration due to their multi-differentiation potential. Finally, we summarize the role of extracellular matrix components of BTJ in guiding stem cell fate to a chondrogenic commitment, so as to provide cues for understanding the mechanisms of lineage-specific potential of biomimetic approaches as well as to inspire researchers to incorporate unique ECM components that facilitate BTJ repair into design.
Collapse
Affiliation(s)
- Qin Shengnan
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Samuel Bennett
- School of Biomedical Sciences, The University of Western Australia, Perth, Australia
| | - Wang Wen
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Li Aiguo
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China.
| | - Xu Jiake
- School of Biomedical Sciences, The University of Western Australia, Perth, Australia.
| |
Collapse
|
21
|
Characterization of Osteogenesis and Chondrogenesis of Human Decellularized Allogeneic Bone with Mesenchymal Stem Cells Derived from Bone Marrow, Adipose Tissue, and Wharton's Jelly. Int J Mol Sci 2021; 22:ijms22168987. [PMID: 34445692 PMCID: PMC8396436 DOI: 10.3390/ijms22168987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/29/2022] Open
Abstract
Allogeneic bone grafts are a promising material for bone implantation due to reduced operative trauma, reduced blood loss, and no donor-site morbidity. Although human decellularized allogeneic bone (hDCB) can be used to fill bone defects, the research of revitalizing hDCB blocks with human mesenchymal stem cells (hMSCs) for osteochondral regeneration is missing. The hMSCs derived from bone marrow, adipose tissue, and Wharton’s jelly (BMMSCs, ADMSCs, and UMSCs, respectively) are potential candidates for bone regeneration. This study characterized the potential of hDCB as a scaffold for osteogenesis and chondrogenesis of BMMSCs, ADMSCs, and UMSCs. The pore sizes and mechanical strength of hDCB were characterized. Cell survival and adhesion of hMSCs were investigated using MTT assay and F-actin staining. Alizarin Red S and Safranin O staining were conducted to demonstrate calcium deposition and proteoglycan production of hMSCs after osteogenic and chondrogenic differentiation, respectively. A RT-qPCR was performed to analyze the expression levels of osteogenic and chondrogenic markers in hMSCs. Results indicated that BMMSCs and ADMSCs exhibited higher osteogenic potential than UMSCs. Furthermore, ADMSCs and UMSCs had higher chondrogenic potential than BMMSCs. This study demonstrated that chondrogenic ADMSCs- or UMSCs-seeded hDCB might be potential osteochondral constructs for osteochondral regeneration.
Collapse
|
22
|
Pereira AR, Lipphaus A, Ergin M, Salehi S, Gehweiler D, Rudert M, Hansmann J, Herrmann M. Modeling of the Human Bone Environment: Mechanical Stimuli Guide Mesenchymal Stem Cell-Extracellular Matrix Interactions. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4431. [PMID: 34442954 PMCID: PMC8398413 DOI: 10.3390/ma14164431] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023]
Abstract
In bone tissue engineering, the design of in vitro models able to recreate both the chemical composition, the structural architecture, and the overall mechanical environment of the native tissue is still often neglected. In this study, we apply a bioreactor system where human bone-marrow hMSCs are seeded in human femoral head-derived decellularized bone scaffolds and subjected to dynamic culture, i.e., shear stress induced by continuous cell culture medium perfusion at 1.7 mL/min flow rate and compressive stress by 10% uniaxial load at 1 Hz for 1 h per day. In silico modeling revealed that continuous medium flow generates a mean shear stress of 8.5 mPa sensed by hMSCs seeded on 3D bone scaffolds. Experimentally, both dynamic conditions improved cell repopulation within the scaffold and boosted ECM production compared with static controls. Early response of hMSCs to mechanical stimuli comprises evident cell shape changes and stronger integrin-mediated adhesion to the matrix. Stress-induced Col6 and SPP1 gene expression suggests an early hMSC commitment towards osteogenic lineage independent of Runx2 signaling. This study provides a foundation for exploring the early effects of external mechanical stimuli on hMSC behavior in a biologically meaningful in vitro environment, opening new opportunities to study bone development, remodeling, and pathologies.
Collapse
Affiliation(s)
- Ana Rita Pereira
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, 97070 Wuerzburg, Germany; (A.R.P.); (M.E.)
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, 97074 Wuerzburg, Germany
| | - Andreas Lipphaus
- Biomechanics Research Group, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Mert Ergin
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, 97070 Wuerzburg, Germany; (A.R.P.); (M.E.)
- Department of Biomaterials, Center of Energy Technology und Materials Science (TAO), University of Bayreuth, 95447 Bayreuth, Germany;
| | - Sahar Salehi
- Department of Biomaterials, Center of Energy Technology und Materials Science (TAO), University of Bayreuth, 95447 Bayreuth, Germany;
| | | | - Maximilian Rudert
- Department of Orthopedic Surgery, Koenig-Ludwig-Haus, University of Wuerzburg, 97074 Wuerzburg, Germany;
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research, Translational Center for Regenerative Therapies, 97082 Wuerzburg, Germany;
| | - Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, 97070 Wuerzburg, Germany; (A.R.P.); (M.E.)
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, 97074 Wuerzburg, Germany
| |
Collapse
|
23
|
Wei B, Wang W, Liu X, Xu C, Wang Y, Wang Z, Xu J, Guan J, Zhou P, Mao Y. Gelatin methacrylate hydrogel scaffold carrying resveratrol-loaded solid lipid nanoparticles for enhancement of osteogenic differentiation of BMSCs and effective bone regeneration. Regen Biomater 2021; 8:rbab044. [PMID: 34394955 PMCID: PMC8358478 DOI: 10.1093/rb/rbab044] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/30/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Critical-sized bone defects caused by traumatic fractures, tumour resection and congenital malformation are unlikely to heal spontaneously. Bone tissue engineering is a promising strategy aimed at developing in vitro replacements for bone transplantation and overcoming the limitations of natural bone grafts. In this study, we developed an innovative bone engineering scaffold based on gelatin methacrylate (GelMA) hydrogel, obtained via a two-step procedure: first, solid lipid nanoparticles (SLNs) were loaded with resveratrol (Res), a drug that can promote osteogenic differentiation and bone formation; these particles were then encapsulated at different concentrations (0.01%, 0.02%, 0.04% and 0.08%) in GelMA to obtain the final Res-SLNs/GelMA scaffolds. The effects of these scaffolds on osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and bone regeneration in rat cranial defects were evaluated using various characterization assays. Our in vitro and in vivo investigations demonstrated that the different Res-SLNs/GelMA scaffolds improved the osteogenic differentiation of BMSCs, with the ideally slow and steady release of Res; the optimal scaffold was 0.02 Res-SLNs/GelMA. Therefore, the 0.02 Res-SLNs/GelMA hydrogel is an appropriate release system for Res with good biocompatibility, osteoconduction and osteoinduction, thereby showing potential for application in bone tissue engineering.
Collapse
Affiliation(s)
- Bangguo Wei
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, China
| | - Wenrui Wang
- School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Xiangyu Liu
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, China
| | - Chenxi Xu
- School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Yanan Wang
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, China
| | - Ziqi Wang
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Jinnuo Xu
- School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Jianzhong Guan
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, China
| | - Pinghui Zhou
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, China
| | - Yingji Mao
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
24
|
Zhang Y, Wu D, Zhao X, Pakvasa M, Tucker AB, Luo H, Qin KH, Hu DA, Wang EJ, Li AJ, Zhang M, Mao Y, Sabharwal M, He F, Niu C, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Chen C, Wagstaff W, Reid RR, Athiviraham A, Ho S, Lee MJ, Hynes K, Strelzow J, He TC, El Dafrawy M. Stem Cell-Friendly Scaffold Biomaterials: Applications for Bone Tissue Engineering and Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:598607. [PMID: 33381499 PMCID: PMC7767872 DOI: 10.3389/fbioe.2020.598607] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic organ with high regenerative potential and provides essential biological functions in the body, such as providing body mobility and protection of internal organs, regulating hematopoietic cell homeostasis, and serving as important mineral reservoir. Bone defects, which can be caused by trauma, cancer and bone disorders, pose formidable public health burdens. Even though autologous bone grafts, allografts, or xenografts have been used clinically, repairing large bone defects remains as a significant clinical challenge. Bone tissue engineering (BTE) emerged as a promising solution to overcome the limitations of autografts and allografts. Ideal bone tissue engineering is to induce bone regeneration through the synergistic integration of biomaterial scaffolds, bone progenitor cells, and bone-forming factors. Successful stem cell-based BTE requires a combination of abundant mesenchymal progenitors with osteogenic potential, suitable biofactors to drive osteogenic differentiation, and cell-friendly scaffold biomaterials. Thus, the crux of BTE lies within the use of cell-friendly biomaterials as scaffolds to overcome extensive bone defects. In this review, we focus on the biocompatibility and cell-friendly features of commonly used scaffold materials, including inorganic compound-based ceramics, natural polymers, synthetic polymers, decellularized extracellular matrix, and in many cases, composite scaffolds using the above existing biomaterials. It is conceivable that combinations of bioactive materials, progenitor cells, growth factors, functionalization techniques, and biomimetic scaffold designs, along with 3D bioprinting technology, will unleash a new era of complex BTE scaffolds tailored to patient-specific applications.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Xia Zhao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Andrew Blake Tucker
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Eric J. Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Maya Sabharwal
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Fang He
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Laboratory Diagnostic Medicine, The Affiliated Hospital of the University of Chinese Academy of Sciences, Chongqing General Hospital, Chongqing, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine and the Affiliated Hospitals, Chongqing Medical University, Chongqing, China
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Surgery Section of Plastic and Reconstructive Surgery, The University of Chicago Medical Center, Chicago, IL, United States
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
25
|
Mendibil U, Ruiz-Hernandez R, Retegi-Carrion S, Garcia-Urquia N, Olalde-Graells B, Abarrategi A. Tissue-Specific Decellularization Methods: Rationale and Strategies to Achieve Regenerative Compounds. Int J Mol Sci 2020; 21:E5447. [PMID: 32751654 PMCID: PMC7432490 DOI: 10.3390/ijms21155447] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is a complex network with multiple functions, including specific functions during tissue regeneration. Precisely, the properties of the ECM have been thoroughly used in tissue engineering and regenerative medicine research, aiming to restore the function of damaged or dysfunctional tissues. Tissue decellularization is gaining momentum as a technique to obtain potentially implantable decellularized extracellular matrix (dECM) with well-preserved key components. Interestingly, the tissue-specific dECM is becoming a feasible option to carry out regenerative medicine research, with multiple advantages compared to other approaches. This review provides an overview of the most common methods used to obtain the dECM and summarizes the strategies adopted to decellularize specific tissues, aiming to provide a helpful guide for future research development.
Collapse
Affiliation(s)
- Unai Mendibil
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain; (N.G.-U.); (B.O.-G.)
| | - Raquel Ruiz-Hernandez
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
| | - Sugoi Retegi-Carrion
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
| | - Nerea Garcia-Urquia
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain; (N.G.-U.); (B.O.-G.)
| | - Beatriz Olalde-Graells
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain; (N.G.-U.); (B.O.-G.)
| | - Ander Abarrategi
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
26
|
Li Z, Xiang S, Li EN, Fritch MR, Alexander PG, Lin H, Tuan RS. Tissue Engineering for Musculoskeletal Regeneration and Disease Modeling. Handb Exp Pharmacol 2020; 265:235-268. [PMID: 33471201 DOI: 10.1007/164_2020_377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Musculoskeletal injuries and associated conditions are the leading cause of physical disability worldwide. The concept of tissue engineering has opened up novel approaches to repair musculoskeletal defects in a fast and/or efficient manner. Biomaterials, cells, and signaling molecules constitute the tissue engineering triad. In the past 40 years, significant progress has been made in developing and optimizing all three components, but only a very limited number of technologies have been successfully translated into clinical applications. A major limiting factor of this barrier to translation is the insufficiency of two-dimensional cell cultures and traditional animal models in informing the safety and efficacy of in-human applications. In recent years, microphysiological systems, often referred to as organ or tissue chips, generated according to tissue engineering principles, have been proposed as the next-generation drug testing models. This chapter aims to first review the current tissue engineering-based approaches that are being applied to fabricate and develop the individual critical elements involved in musculoskeletal organ/tissue chips. We next highlight the general strategy of generating musculoskeletal tissue chips and their potential in future regenerative medicine research. Exemplary microphysiological systems mimicking musculoskeletal tissues are described. With sufficient physiological accuracy and relevance, the human cell-derived, three-dimensional, multi-tissue systems have been used to model a number of orthopedic disorders and to test new treatments. We anticipate that the novel emerging tissue chip technology will continually reshape and improve our understanding of human musculoskeletal pathophysiology, ultimately accelerating the development of advanced pharmaceutics and regenerative therapies.
Collapse
Affiliation(s)
- Zhong Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shiqi Xiang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eileen N Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Madalyn R Fritch
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA.
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|