1
|
Wen Z, Li S, Liu Y, Liu X, Qiu H, Che Y, Bian L, Zhou M. An engineered M2 macrophage-derived exosomes-loaded electrospun biomimetic periosteum promotes cell recruitment, immunoregulation, and angiogenesis in bone regeneration. Bioact Mater 2025; 50:95-115. [PMID: 40242509 PMCID: PMC12002949 DOI: 10.1016/j.bioactmat.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The periosteum, a fibrous tissue membrane covering bone surfaces, is critical to osteogenesis and angiogenesis in bone reconstruction. Artificial periostea have been widely developed for bone defect repair, but most of these are lacking of periosteal bioactivity. Herein, a biomimetic periosteum (termed PEC-Apt-NP-Exo) is prepared based on an electrospun membrane combined with engineered exosomes (Exos). The electrospun membrane is fabricated using poly(ε-caprolactone) (core)-periosteal decellularized extracellular matrix (shell) fibers via coaxial electrospinning, to mimic the fibrous structure, mechanical property, and tissue microenvironment of natural periosteum. The engineered Exos derived from M2 macrophages are functionalized by surface modification of bone marrow mesenchymal stem cell (BMSC)-specific aptamers to further enhance cell recruitment, immunoregulation, and angiogenesis in bone healing. The engineered Exos are covalently bonded to the electrospun membrane, to achieve rich loading and long-term effects of Exos. In vitro experiments demonstrate that the biomimetic periosteum promotes BMSC migration and osteogenic differentiation via Rap1/PI3K/AKT signaling pathway, and enhances vascular endothelial growth factor secretion from BMSCs to facilitate angiogenesis. In vivo studies reveal that the biomimetic periosteum promotes new bone formation in large bone defect repair by inducing M2 macrophage polarization, endogenous BMSC recruitment, osteogenic differentiation, and vascularization. This research provides valuable insights into the development of a multifunctional biomimetic periosteum for bone regeneration.
Collapse
Affiliation(s)
- Zhuohao Wen
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shuyi Li
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yi Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Xueyan Liu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China
| | - Huiguo Qiu
- Zhuhai Stomatological Hospital, Zhuhai, 519000, China
| | - Yuejuan Che
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Miao Zhou
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
2
|
Hoveidaei AH, Sadat-Shojai M, Nabavizadeh SS, Niakan R, Shirinezhad A, MosalamiAghili S, Tabaie S. Clinical challenges in bone tissue engineering - A narrative review. Bone 2025; 192:117363. [PMID: 39638083 DOI: 10.1016/j.bone.2024.117363] [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: 09/15/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Bone tissue engineering (BTE) has emerged as a promising approach to address large bone defects caused by trauma, infections, congenital malformations, and tumors. This review focuses on scaffold design, cell sources, growth factors, and vascularization strategies, highlighting their roles in developing effective treatments. We explore the complexities of balancing mechanical properties, porosity, and biocompatibility in scaffold materials, alongside optimizing mesenchymal stem cell delivery methods. The critical role of growth factors in bone regeneration and the need for controlled release systems are discussed. Vascularization remains a significant hurdle, with strategies such as angiogenic factors, co-culture systems, and bioprinting under investigation. Mechanical challenges, tissue responses, and inflammation management are examined, alongside gene therapy's potential for enhancing osteogenesis and angiogenesis via both viral and non-viral delivery methods. The review emphasizes the impact of patient-specific factors on bone healing outcomes and the importance of personalized approaches. Future directions are described, emphasizing the necessity of interdisciplinary cooperation to advance the field of BTE and convert laboratory results into clinically feasible solutions.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Sara S Nabavizadeh
- Otolaryngology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Niakan
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Sean Tabaie
- Department of Orthopaedic Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
3
|
Shah SA, Sohail M, Nakielski P, Rinoldi C, Zargarian SS, Kosik-Kozioł A, Ziai Y, Haghighat Bayan MA, Zakrzewska A, Rybak D, Bartolewska M, Pierini F. Integrating Micro- and Nanostructured Platforms and Biological Drugs to Enhance Biomaterial-Based Bone Regeneration Strategies. Biomacromolecules 2025; 26:140-162. [PMID: 39621708 PMCID: PMC11733931 DOI: 10.1021/acs.biomac.4c01133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 01/14/2025]
Abstract
Bone defects resulting from congenital anomalies and trauma pose significant clinical challenges for orthopedics surgeries, where bone tissue engineering (BTE) aims to address these challenges by repairing defects that fail to heal spontaneously. Despite numerous advances, BTE still faces several challenges, i.e., difficulties in detecting and tracking implanted cells, high costs, and regulatory approval hurdles. Biomaterials promise to revolutionize bone grafting procedures, heralding a new era of regenerative medicine and advancing patient outcomes worldwide. Specifically, novel bioactive biomaterials have been developed that promote cell adhesion, proliferation, and differentiation and have osteoconductive and osteoinductive characteristics, stimulating tissue regeneration and repair, particularly in complex skeletal defects caused by trauma, degeneration, and neoplasia. A wide array of biological therapeutics for bone regeneration have emerged, drawing from the diverse spectrum of gene therapy, immune cell interactions, and RNA molecules. This review will provide insights into the current state and potential of future strategies for bone regeneration.
Collapse
Affiliation(s)
- Syed Ahmed Shah
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
- Faculty
of Pharmacy, The Superior University, Lahore 54000, Punjab, Pakistan
| | - Muhammad Sohail
- Faculty
of Pharmacy, Cyprus International University, Nicosia 99258, North Cyprus
| | - Paweł Nakielski
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Chiara Rinoldi
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Alicja Kosik-Kozioł
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Yasamin Ziai
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Mohammad Ali Haghighat Bayan
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Anna Zakrzewska
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Daniel Rybak
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Magdalena Bartolewska
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, Warsaw 02-106, Poland
| |
Collapse
|
4
|
Rambhia KJ, Sun H, Feng K, Kannan R, Doleyres Y, Holzwarth JM, Doepker M, Franceschi RT, Ma PX. Nanofibrous 3D scaffolds capable of individually controlled BMP and FGF release for the regulation of bone regeneration. Acta Biomater 2024:S1742-7061(24)00636-6. [PMID: 39486780 DOI: 10.1016/j.actbio.2024.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The current clinical applications of bone morphogenetic proteins (BMPs) are limited to only a few specific indications. Locally controlled delivery of combinations of growth factors can be a promising strategy to improve BMP-based bone repair. However, the success of this approach requires the development of an effective release system and the correct choice of growth factors capable of enhancing BMP activity. Basic fibroblast growth factor (bFGF, also known as FGF-2) has shown promise in promoting bone repair, although conflicting results have been reported. Considering the complex biological activities of FGF-2, we hypothesized that FGF-2 can promote BMP-induced bone regeneration only if the dosage and kinetic parameters of the two factors are individually tailored. In this study, we conducted systematic in vitro studies on cell proliferation, differentiation, and mineralization in response to factor dose, delivery mode (sequential or simultaneous), and release rate. Subsequently, we designed individually controlled BMP-7 and FGF-2 release poly(lactide-co-glycolide) (PLGA) nanospheres attached to the poly(l-lactic acid) (PLLA) nanofibrous scaffolds. The data showed that BMP-7-induced bone formation was accelerated by a relatively higher FGF-2 dose (100 ng/scaffold) delivered at a faster release rate, or by a relatively lower FGF-2 dose (10 ng/scaffold) at a slower release rate in an in vivo bone regeneration model. In contrast, a very high dose of FGF-2 (1000 ng/scaffold) inhibited bone regeneration under all conditions. In vitro and in vivo data suggest that FGF-2 improved BMP-7-induced bone regeneration by coordinating FGF-2 dosage and release kinetics to enhance stem cell migration, proliferation, and angiogenesis. STATEMENT OF SIGNIFICANCE: Bone morphogenetic proteins (BMPs) are the most potent growth/differentiation factors in bone development and regeneration. However, the clinical applications of BMPs have been limited to only a few specific indications due to the required supraphysiological dosages with the current BMP products and their side effects. Locally controlled delivery of BMPs and additional growth factors that can enhance their osteogenic potency are highly desired. However, different growth factors act with different mechanisms. Here we report a nanofibrous scaffold that mimics collagen in size and geometry and is immobilized with biodegradable nanospheres to achieve local and distinct release profiles of BMP7 and FGF2. Systematic studies demonstrated low dose BMP7 and FGF2 with different temporal release profiles can optimally enhance bone regeneration.
Collapse
Affiliation(s)
- Kunal J Rambhia
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hongli Sun
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Feng
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rahasudha Kannan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yasmine Doleyres
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeremy M Holzwarth
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mikayla Doepker
- Department of Biology, Kalamazoo College, Kalamazoo, MI 49006, USA
| | - Renny T Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Muniz TDTP, Rossi MC, de Vasconcelos Machado VM, Alves ALG. Mesenchymal Stem Cells and Tissue Bioengineering Applications in Sheep as Ideal Model. Stem Cells Int 2024; 2024:5176251. [PMID: 39465229 PMCID: PMC11511598 DOI: 10.1155/2024/5176251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
The most common technologies in tissue engineering include growth factor therapies; metal implants, such as titanium; 3D bioprinting; nanoimprinting for ceramic/polymer scaffolds; and cell therapies, such as mesenchymal stem cells (MSCs). Cell therapy is a promising alternative to organ grafts and transplants in the treatment of numerous musculoskeletal diseases. MSCs have increasingly been used in generative medicine due to their specialized self-renewal, immunomodulation, multiplication, and differentiation properties. To further expand the potential of these cells in tissue repair, significant efforts are currently dedicated to the production of biomaterials with desirable short- and long-term biophysical properties that can aid the differentiation and expansion of MSCs. Biomaterials support MSC differentiation by modulating their characteristics, such as composition, mechanical properties, porosity, and topography. This review aimed to describe recent MSC approaches, including those associated with biomaterials, from experimental, clinical, and preclinical studies with sheep models.
Collapse
Affiliation(s)
- Talita D'Paula Tavares Pereira Muniz
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Mariana Correa Rossi
- Materials Engineering Department (DEMa), São Carlos Federal University (UFSCar), 13.565-905, São Carlos, Sao Paulo, Brazil
| | - Vânia Maria de Vasconcelos Machado
- Department of Veterinary Surgery and Animal Reproduction, Imaging Diagnostic Sector, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Ana Liz Garcia Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| |
Collapse
|
6
|
Chow SKH, Gao Q, Pius A, Morita M, Ergul Y, Murayama M, Shinohara I, Cekuc MS, Ma C, Susuki Y, Goodman SB. The Advantages and Shortcomings of Stem Cell Therapy for Enhanced Bone Healing. Tissue Eng Part C Methods 2024; 30:415-430. [PMID: 39311464 DOI: 10.1089/ten.tec.2024.0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
This review explores the regenerative potential of key progenitor cell types and therapeutic strategies to improve healing of complex fractures and bone defects. We define, summarize, and discuss the differentiation potential of totipotent, pluripotent, and multipotent stem cells, emphasizing the advantages and shortcomings of cell therapy for bone repair and regeneration. The fundamental role of mesenchymal stem cells is highlighted due to their multipotency to differentiate into the key lineage cells including osteoblasts, osteocytes, and chondrocytes, which are crucial for bone formation and remodeling. Hematopoietic stem cells (HSCs) also play a significant role; immune cells such as macrophages and T-cells modulate inflammation and tissue repair. Osteoclasts are multinucleated cells that are important to bone remodeling. Vascular progenitor (VP) cells are critical to oxygen and nutrient supply. The dynamic interplay among these lineages and their microenvironment is essential for effective bone restoration. Therapies involving cells that are more than "minimally manipulated" are controversial and include embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). ESCs, derived from early-stage embryos, possess pluripotent capabilities and have shown promise in preclinical studies for bone healing. iPSCs, reprogrammed from somatic cells, offer personalized medicine applications and can differentiate into various tissue-specific cell lines. Minimally manipulative cell therapy approaches such as the use of bone marrow aspirate concentrate (BMAC), exosomes, and various biomaterials for local delivery are explored for their effectiveness in bone regeneration. BMAC, which contains mostly immune cells but few mesenchymal and VPs, probably improves bone healing by facilitating paracrine-mediated intercellular communication. Exosome isolation harnesses the biological signals and cellular by-products that are a primary source for cell crosstalk and activation. Safe, efficacious, and cost-effective strategies to enhance bone healing using novel cellular therapies are part of a changing paradigm to modulate the inflammatory, repair, and regenerative pathways to achieve earlier more robust tissue healing and improved physical function. Impact Statement Stem cell therapy holds immense potential for bone healing due to its ability to regenerate damaged tissue. Nonmanipulated bone marrow aspirate contains mesenchymal stem cells that promote bone repair and reduce healing time. Induced pluripotent stem cells offer the advantage of creating patient-specific cells that can differentiate into osteoblasts, aiding in bone regeneration. Other delivery methods, such as scaffold-based techniques, enhance stem cell integration and function. Collectively, these approaches can improve treatment outcomes, reduce recovery periods, and advance our understanding of bone healing mechanisms, making them pivotal in orthopedic research and regenerative medicine.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Alexa Pius
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mayu Morita
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yasemin Ergul
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mehmet Sertac Cekuc
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Chao Ma
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
7
|
Abdollahi F, Saghatchi M, Paryab A, Malek Khachatourian A, Stephens ED, Toprak MS, Badv M. Angiogenesis in bone tissue engineering via ceramic scaffolds: A review of concepts and recent advancements. BIOMATERIALS ADVANCES 2024; 159:213828. [PMID: 38479240 DOI: 10.1016/j.bioadv.2024.213828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
Due to organ donor shortages, long transplant waitlists, and the complications/limitations associated with auto and allotransplantation, biomaterials and tissue-engineered models are gaining attention as feasible alternatives for replacing and reconstructing damaged organs and tissues. Among various tissue engineering applications, bone tissue engineering has become a promising strategy to replace or repair damaged bone. We aimed to provide an overview of bioactive ceramic scaffolds in bone tissue engineering, focusing on angiogenesis and the effect of different biofunctionalization strategies. Different routes to angiogenesis, including chemical induction through signaling molecules immobilized covalently or non-covalently, in situ secretion of angiogenic growth factors, and the degradation of inorganic scaffolds, are described. Physical induction mechanisms are also discussed, followed by a review of methods for fabricating bioactive ceramic scaffolds via microfabrication methods, such as photolithography and 3D printing. Finally, the strengths and weaknesses of the commonly used methodologies and future directions are discussed.
Collapse
Affiliation(s)
- Farnoosh Abdollahi
- Department of Dentistry, Kashan University of Medical Science, Kashan, Iran
| | - Mahshid Saghatchi
- School of Metallurgy & Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Amirhosein Paryab
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Emma D Stephens
- Department of Biomedical Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Muhammet S Toprak
- Department of Applied Physics, Biomedical and X-ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden
| | - Maryam Badv
- Department of Biomedical Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Libin Cardiovascular Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
8
|
Wang D, Guo Y, Heng BC, Zhang X, Wei Y, He Y, Xu M, Xia B, Deng X. Cell membrane vesicles derived from hBMSCs and hUVECs enhance bone regeneration. Bone Res 2024; 12:23. [PMID: 38594236 PMCID: PMC11003965 DOI: 10.1038/s41413-024-00325-9] [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: 04/12/2023] [Revised: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Bone tissue renewal can be enhanced through co-transplantation of bone mesenchymal stem cells (BMSCs) and vascular endothelial cells (ECs). However, there are apparent limitations in stem cell-based therapy which hinder its clinic translation. Hence, we investigated the potential of alternative stem cell substitutes for facilitating bone regeneration. In this study, we successfully prepared cell membrane vesicles (CMVs) from BMSCs and ECs. The results showed that BMSC-derived cell membrane vesicles (BMSC-CMVs) possessed membrane receptors involved in juxtacrine signaling and growth factors derived from their parental cells. EC-derived cell membrane vesicles (EC-CMVs) also contained BMP2 and VEGF derived from their parental cells. BMSC-CMVs enhanced tube formation and migration ability of hUVECs, while EC-CMVs promoted the osteogenic differentiation of hBMSCs in vitro. Using a rat skull defect model, we found that co-transplantation of BMSC-CMVs and EC-CMVs could stimulate angiogenesis and bone formation in vivo. Therefore, our research might provide an innovative and feasible approach for cell-free therapy in bone tissue regeneration.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Boon Chin Heng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Bin Xia
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China.
| |
Collapse
|
9
|
Mo Y, He W, Hu S, Guo H, Li S, Zhang J, Wang X. 3D printed Si-CaP scaffold released SiO 32- and Ca 2+ to synergistically promote angiogenesis. J Biomater Appl 2024; 38:784-793. [PMID: 37963098 DOI: 10.1177/08853282231216546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
BACKGROUND AND PURPOSE Structuring scaffold with both osteogenic and angiogenesis capabilities is a challenge for bone tissue engineering. Powder structured Si-CaP materials have shown excellent osteogenic properties and induction of stem cell differentiation. Our research group have successful produced 3D printed Si-CaP scaffolds by DLP technology. This study aims to explore the angiogenic effects of SiO32- and Ca2+ released by 3D printed Si-CaP scaffold, and whether there is a synergistic effect between the two ions. METHODS The 3D printed Si-CaP scaffolds were immersed in endothelial cell medium solution for 24 h. The Si, Ca ion released was detected by Inductively coupled plasma-optical emission spectrometry. We used detected data as a standard to prepare the simulated solution to investigate the effect of SiO32-, Ca2+ separately. Experiment was divided into control group, Si ion group, Ca ion group and Si + Ca ion group. We evaluated different ionic effect on HUVECs viability, proliferation, migration, gene expression, and tube formation on different groups. RESULTS The concentration of SiO32- was detected as 15.71 ± 0.04 μg/mL, Ca2+ as 67.14 ± 0.95 μg/mL. Na2SiO3 and CaCl2 were used to prepare the simulated solution. There were no statistically difference between simulated solution from ion released by scaffold. Si + Ca group promoted the gene expression significantly compared with the control group, p < .01. Expression of vascular-associated protein in Si + Ca ion group was higher than that in Si ion group, Ca ion group and control group. Si + Ca ion group significantly enhanced endothelial cell on migration and tube formation assay. CONCLUSION The 3D printed Si-CaP scaffold can release effective physiological concentrations of Si, Ca ions. Si and Ca ions have a synergistic effect on promoting angiogenesis of HUVECs. 3D printed Si-CaP scaffold is promising in vascularized bone tissue engineering application.
Collapse
Affiliation(s)
- Yongqiang Mo
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weitao He
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shiqi Hu
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongchun Guo
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuangzuo Li
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingwei Zhang
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintao Wang
- Department of Orthopaedic, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
11
|
Dalfino S, Savadori P, Piazzoni M, Connelly ST, Giannì AB, Del Fabbro M, Tartaglia GM, Moroni L. Regeneration of Critical-Sized Mandibular Defects Using 3D-Printed Composite Scaffolds: A Quantitative Evaluation of New Bone Formation in In Vivo Studies. Adv Healthc Mater 2023; 12:e2300128. [PMID: 37186456 PMCID: PMC11469182 DOI: 10.1002/adhm.202300128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/12/2023] [Indexed: 05/17/2023]
Abstract
Mandibular tissue engineering aims to develop synthetic substitutes for the regeneration of critical size defects (CSD) caused by a variety of events, including tumor surgery and post-traumatic resections. Currently, the gold standard clinical treatment of mandibular resections (i.e., autologous fibular flap) has many drawbacks, driving research efforts toward scaffold design and fabrication by additive manufacturing (AM) techniques. Once implanted, the scaffold acts as a support for native tissue and facilitates processes that contribute to its regeneration, such as cells infiltration, matrix deposition and angiogenesis. However, to fulfil these functions, scaffolds must provide bioactivity by mimicking natural properties of the mandible in terms of structure, composition and mechanical behavior. This review aims to present the state of the art of scaffolds made with AM techniques that are specifically employed in mandibular tissue engineering applications. Biomaterials chemical composition and scaffold structural properties are deeply discussed, along with strategies to promote osteogenesis (i.e., delivery of biomolecules, incorporation of stem cells, and approaches to induce vascularization in the constructs). Finally, a comparison of in vivo studies is made by taking into consideration the amount of new bone formation (NB), the CSD dimensions, and the animal model.
Collapse
Affiliation(s)
- Sophia Dalfino
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilano20122Italy
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology Inspired Regenerative MedicineMaastricht6229 ERThe Netherlands
- Fondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilano20122Italy
| | - Paolo Savadori
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilano20122Italy
- Fondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilano20122Italy
| | - Marco Piazzoni
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilano20122Italy
- Department of PhysicsUniversità degli Studi di MilanoMilano20133Italy
| | - Stephen Thaddeus Connelly
- Department of Oral & Maxillofacial SurgeryUniversity of California San Francisco4150 Clement StSan FranciscoCA94121USA
| | - Aldo Bruno Giannì
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilano20122Italy
- Fondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilano20122Italy
| | - Massimo Del Fabbro
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilano20122Italy
- Fondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilano20122Italy
| | - Gianluca Martino Tartaglia
- Department of BiomedicalSurgical and Dental SciencesUniversità degli Studi di MilanoMilano20122Italy
- Fondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilano20122Italy
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology Inspired Regenerative MedicineMaastricht6229 ERThe Netherlands
| |
Collapse
|
12
|
Zhou Z, Zhou A, Jalil AT, Saleh MM, Huang C. Carbon nanoparticles-based hydrogel nanocomposite induces bone repair in vivo. Bioprocess Biosyst Eng 2023; 46:577-588. [PMID: 36580135 DOI: 10.1007/s00449-022-02843-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
The main objective of the current study is to fabricate a 3D scaffold using alginate hydrogel implemented with carbon nanoparticles (CNPs) as the filler. The SEM imaging revealed that the scaffold possesses a porous internal structure with interconnected pores. The swelling value of the scaffolds (more than 400%) provides a wet niche for bone cell proliferation and migration. The in vitro evaluations showed that the scaffolds were hemocompatible (with hemolysis induction lower than 5%) and cytocompatible (inducing significant proliferative effect (cell viability of 121 ± 4%, p < 0.05) for AlG/CNPs 10%). The in vivo studies showed that the implantation of the fabricated 3D nanocomposite scaffolds induced a bone-forming effect and mediated bone formation into the induced bone defect. In conclusion, these results implied that the fabricated NFC-integrated 3D scaffold exhibited promising characteristics beneficial for bone regeneration and can be applied as the bone tissue engineering scaffold.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Orthopaedic Surgery, Yangzhou Hongquan Hospital, Yangzhou, 225200, China
| | - Ao Zhou
- Department of Bone and Soft Tissue Oncology, Cancer Hospital Affiliated to Chongqing University, Chongqing, 400020, China
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Hilla, 51001, Babylon, Iraq
| | - Marwan Mahmood Saleh
- Department of Biophysics, College of Applied Sciences, University of Anbar, Ramadi, Iraq.,Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Chengjun Huang
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
13
|
A Review of 3D Polymeric Scaffolds for Bone Tissue Engineering: Principles, Fabrication Techniques, Immunomodulatory Roles, and Challenges. Bioengineering (Basel) 2023; 10:bioengineering10020204. [PMID: 36829698 PMCID: PMC9952306 DOI: 10.3390/bioengineering10020204] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Over the last few years, biopolymers have attracted great interest in tissue engineering and regenerative medicine due to the great diversity of their chemical, mechanical, and physical properties for the fabrication of 3D scaffolds. This review is devoted to recent advances in synthetic and natural polymeric 3D scaffolds for bone tissue engineering (BTE) and regenerative therapies. The review comprehensively discusses the implications of biological macromolecules, structure, and composition of polymeric scaffolds used in BTE. Various approaches to fabricating 3D BTE scaffolds are discussed, including solvent casting and particle leaching, freeze-drying, thermally induced phase separation, gas foaming, electrospinning, and sol-gel techniques. Rapid prototyping technologies such as stereolithography, fused deposition modeling, selective laser sintering, and 3D bioprinting are also covered. The immunomodulatory roles of polymeric scaffolds utilized for BTE applications are discussed. In addition, the features and challenges of 3D polymer scaffolds fabricated using advanced additive manufacturing technologies (rapid prototyping) are addressed and compared to conventional subtractive manufacturing techniques. Finally, the challenges of applying scaffold-based BTE treatments in practice are discussed in-depth.
Collapse
|
14
|
Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue-engineered endochondral ossification approaches. iScience 2022; 25:105370. [PMID: 36339269 PMCID: PMC9626746 DOI: 10.1016/j.isci.2022.105370] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In order to solve the clinical challenges related to bone grafting, several tissue engineering (TE) strategies have been proposed to repair critical-sized defects. Generally, the classical TE approaches are designed to promote bone repair via intramembranous ossification. Although promising, strategies that direct the osteogenic differentiation of mesenchymal stem/stromal cells are usually characterized by a lack of functional vascular supply, often resulting in necrotic cores. A less explored alternative is engineering bone constructs through a cartilage-mediated approach, resembling the embryological process of endochondral ossification. The remodeling of an intermediary hypertrophic cartilaginous template triggers vascular invasion and bone tissue deposition. Thus, employing this knowledge can be a promising direction for the next generation of bone TE constructs. This review highlights the most recent biomimetic strategies for applying endochondral ossification in bone TE while discussing the plethora of cell types, culture conditions, and biomaterials essential to promote a successful bone regeneration process.
Collapse
|
15
|
Rudiansyah M, El-Sehrawy AA, Ahmad I, Terefe EM, Abdelbasset WK, Bokov DO, Salazar A, Rizaev JA, Muthanna FMS, Shalaby MN. Osteoporosis treatment by mesenchymal stromal/stem cells and their exosomes: Emphasis on signaling pathways and mechanisms. Life Sci 2022; 306:120717. [PMID: 35792178 DOI: 10.1016/j.lfs.2022.120717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/30/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023]
Abstract
Osteoporosis is the loss of bone density, which is one of the main problems in developed and developing countries and is more common in the elderly. Because this disease is often not diagnosed until a bone fracture, it can become a life-threatening disease and cause hospitalization. With the increase of older people in a population, this disease's personal and social costs increase year by year and affect different communities. Most current treatments focus on pain relief and usually do not lead to bone tissue recovery and regeneration. But today, the use of stem cell therapy is recommended to treat and improve this disease recovery, which helps restore bone tissue by improving the imbalance in the osteoblast-osteoclast axis. Due to mesenchymal stromal/stem cells (MSCs) characteristics and their exosomes, these cells and vesicles are excellent sources for treating and preventing the progression and improvement of osteoporosis. Due to the ability of MSCs to differentiate into different cells and migrate to the site of injury, these cells are used in tissue regenerative medicine. Also, due to their contents, the exosomes of these cells help regenerate and treat various tissue injuries by affecting the injury site's cells. In this article, we attempted to review new studies in which MSCs and their exosomes were used to treat osteoporosis.
Collapse
Affiliation(s)
- Mohammad Rudiansyah
- Division of Nephrology & Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Lambung Mangkurat/Ulin Hospital, Banjarmasin, Indonesia
| | - Amr A El-Sehrawy
- Department of Internal Medicine, Mansoura Specialized Medical Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ermias Mergia Terefe
- School of pharmacy and Health science, United States International University, Nairobi, Kenya
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia; Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow 119991, Russian Federation; Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow 109240, Russian Federation
| | - Aleli Salazar
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City, Mexico
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector of Samarkand State Medical Institute, Samarkand, Uzbekistan
| | | | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Egypt
| |
Collapse
|
16
|
Azari Matin A, Fattah K, Saeidpour Masouleh S, Tavakoli R, Houshmandkia SA, Moliani A, Moghimimonfared R, Pakzad S, Dalir Abdolahinia E. Synthetic electrospun nanofibers as a supportive matrix in osteogenic differentiation of induced pluripotent stem cells. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1469-1493. [PMID: 35321624 DOI: 10.1080/09205063.2022.2056941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Continuous remodeling is not able to repair large bone defects. Bone tissue engineering is aimed to repair these defects by creating bone grafts. To do this, several technologies and biomaterials have been employed to fabricate an in vivo-like supportive matrix. Electrospinning is a versatile technique to fabricate porous matrices with interconnected pores and high surface area, replicating in vivo microenvironment. Electrospun scaffolds have been used in a large number of studies to provide a matrix for bone regeneration and osteogenic differentiation of stem cells such as induced pluripotent stem cells (iPSCs). Electrospinning uses both natural and synthetic polymers, either alone or in combination, to fabricate scaffolds. Among them, synthetic polymers have had a great promise in bone regeneration and repair. They allow the fabrication of biocompatible and biodegradable scaffolds with high mechanical properties, suitable for bone engineering. Furthermore, several attempts have done to increase the osteogenic properties of these scaffolds. This paper reviewed the potential of synthetic electrospun scaffolds in osteogenic differentiation of iPSCs. In addition, the approaches to improve the osteogenic differentiation of these scaffolds are addressed.
Collapse
Affiliation(s)
- Arash Azari Matin
- Department of Biology, California State University, Northridge, CA, USA
| | - Khashayar Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Reza Tavakoli
- Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Afshin Moliani
- Isfahan Medical Students Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Moghimimonfared
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Sahar Pakzad
- Department of Oral and Maxillofacial Surgery, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Zhang Z, Chen W, Tiemessen DM, Oosterwijk E, Kouwer PHJ. A Temperature-Based Easy-Separable (TempEasy) 3D Hydrogel Coculture System. Adv Healthc Mater 2022; 11:e2102389. [PMID: 35029325 PMCID: PMC11469334 DOI: 10.1002/adhm.202102389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/10/2021] [Indexed: 12/13/2022]
Abstract
Interactions between different cell types are crucial for their behavior in tissues, but are rarely considered in 3D in vitro cell culture experiments. One reason is that such coculture experiments are sometimes difficult to perform in 3D or require specialized equipment or know-how. Here, a new 3D cell coculture system is introduced, TempEasy, which is readily applied in any cell culture lab. The matrix material is based on polyisocyanide hydrogels, which closely resemble the mechanical characteristics of the natural extracellular matrix. Gels with different gelation temperatures, seeded with different cells, are placed on top of each other to form an indirect coculture. Cooling reverses gelation, allowing cell harvesting from each layer separately, which benefits downstream analysis. To demonstrate the potential of TempEasy , human adipose stem cells (hADSCs) with vaginal epithelial fibroblasts are cocultured. The analysis of a 7-day coculture shows that hADSCs promote cell-cell interaction of fibroblasts, while fibroblasts promote proliferation and differentiation of hADSCs. TempEasy provides a straightforward operational platform for indirect cocultures of cells of different lineages in well-defined microenvironments.
Collapse
Affiliation(s)
- Zhaobao Zhang
- Institute for Molecules and MaterialsRadboud University NijmegenHeyendaalseweg 135Nijmegen6525 AJThe Netherlands
| | - Wen Chen
- Institute for Molecules and MaterialsRadboud University NijmegenHeyendaalseweg 135Nijmegen6525 AJThe Netherlands
| | - Dorien M. Tiemessen
- Department of UrologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525 GAThe Netherlands
| | - Egbert Oosterwijk
- Department of UrologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525 GAThe Netherlands
| | - Paul H. J. Kouwer
- Institute for Molecules and MaterialsRadboud University NijmegenHeyendaalseweg 135Nijmegen6525 AJThe Netherlands
| |
Collapse
|
18
|
Impact of Fluid Dynamics on the Viability and Differentiation Capacity of 3D-Cultured Jaw Periosteal Cells. Int J Mol Sci 2022; 23:ijms23094682. [PMID: 35563073 PMCID: PMC9099539 DOI: 10.3390/ijms23094682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Perfused bioreactor systems are considered to be a promising approach for the 3D culturing of stem cells by improving the quality of the tissue-engineered grafts in terms of better cell proliferation and deeper penetration of used scaffold materials. Our study aims to establish an optimal perfusion culture system for jaw periosteal cell (JPC)-seeded scaffolds. For this purpose, we used beta-tricalcium phosphate (β-TCP) scaffolds as a three-dimensional structure for cell growth and osteogenic differentiation. Experimental set-ups of tangential and sigmoidal fluid configurations with medium flow rates of 100 and 200 µL/min were applied within the perfusion system. Cell metabolic activities of 3D-cultured JPCs under dynamic conditions with flow rates of 100 and 200 µL/min were increased in the tendency after 1, and 3 days of culture, and were significantly increased after 5 days. Significantly higher cell densities were detected under the four perfused conditions compared to the static condition at day 5. However, cell metabolic and proliferation activity under dynamic conditions showed flow rate independency in our study. In this study, dynamic conditions increased the expression of osteogenic markers (ALPL, COL1A1, RUNX2, and OCN) compared to static conditions and the tangential configuration showed a stronger osteogenic effect than the sigmoidal flow configuration.
Collapse
|
19
|
Li J, Cui X, Lindberg G, Alcala-Orozco CR, Hooper GJ, Lim K, Woodfield TBF. Hybrid fabrication of photo-clickable vascular hydrogels with additive manufactured titanium implants for enhanced osseointegration and vascularized bone formation. Biofabrication 2022; 14. [PMID: 35320796 DOI: 10.1088/1758-5090/ac6051] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/23/2022] [Indexed: 11/11/2022]
Abstract
Bone regeneration of critical-sized bone defects, bone fractures or joint replacements remains a significant unmet clinical challenge. Although there has been rapid advancement in both the fields of bone tissue engineering and additive manufacturing (AM), functional bone implants with rapid vascularization capacity to ensure osseointegration and long-term biological fixation in large bone defects remains limited in clinics. In this study, we developed an in vitro vascularized bone implant by combining cell-laden hydrogels with direct metal printed (DMP) porous titanium alloys (Ti-6Al-4V). 5wt% allylated gelatin (GelAGE), was utilized to co-encapsulate human mesenchymal stromal cells (hMSCs) and human umbilical vein endothelial cells (HUVECs) to investigate concurrent osteogenic and vasculogenic performance. DMP macro-porous Ti-6Al-4V scaffolds were subsequently infused/enriched with cell-laden GelAGE to examine the feasibility to deliver cells and engineer vascular-like networks in the hybrid implant. Furthermore, as a proof of concept, a full-scale porous Ti-6Al-4V acetabular cup was impregnated with cell-laden hydrogel to validate the clinical potential of this strategy. The vasculogenic potential was evaluated by examining micro-capillary formation coupled with capillary network maturation and stabilization. Osteogenic differentiation was assessed via ALP activity as well as osteocalcin and osteopontin expression. Our results suggested that GelAGE supported HUVECs spreading and vascular-like network formation, along with osteogenesis of hMSCs. Titanium hybrid constructs with cell-laden hydrogel demonstrated enhanced osteogenesis with similar vasculogenic capability compared to the cell-laden hydrogel alone constructs. The full-scale implant with cell-laden hydrogel coating similarly showed cell distribution and spreading, implying the potential for further clinical application. Our study presents the feasibility of integrating bio-functional hydrogels with porous titanium implants to fabricate a vascularized hybrid construct with both mechanical support and preferable biological functionality (osteogenesis/vasculogenesis), which paves the way for improved strategies to enhance bone regeneration in complex large bone defects achieving long-term bone-implant fixation.
Collapse
Affiliation(s)
- Jun Li
- Dept. of Orthopaedic Surgery , University of Otago, 2 Riccarton Avenue, Christchurch, Christchurch, Canterbury, 8011, NEW ZEALAND
| | - Xiaolin Cui
- University of Otago Christchurch, 2 Riccarton Avenue, Christchurch, 8011, NEW ZEALAND
| | - Gabriella Lindberg
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch, 8011, NEW ZEALAND
| | - Cesar R Alcala-Orozco
- Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, 2 Riccarton Avenue, Christchurch, Christchurch, 8011, NEW ZEALAND
| | - Gary J Hooper
- Christchurch Regenerative Medicine and Tissue Engineering Group Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, PO Box 4345, Christchurch 8140, Christchurch, 8140, NEW ZEALAND
| | - Khoon Lim
- Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, 2 Riccarton Avenue, Christchurch, 8011, NEW ZEALAND
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering Group Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, PO Box 4345, Christchurch 8140, Christchurch, 8140, NEW ZEALAND
| |
Collapse
|
20
|
Maji S, Lee H. Engineering Hydrogels for the Development of Three-Dimensional In Vitro Models. Int J Mol Sci 2022; 23:2662. [PMID: 35269803 PMCID: PMC8910155 DOI: 10.3390/ijms23052662] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 02/06/2023] Open
Abstract
The superiority of in vitro 3D cultures over conventional 2D cell cultures is well recognized by the scientific community for its relevance in mimicking the native tissue architecture and functionality. The recent paradigm shift in the field of tissue engineering toward the development of 3D in vitro models can be realized with its myriad of applications, including drug screening, developing alternative diagnostics, and regenerative medicine. Hydrogels are considered the most suitable biomaterial for developing an in vitro model owing to their similarity in features to the extracellular microenvironment of native tissue. In this review article, recent progress in the use of hydrogel-based biomaterial for the development of 3D in vitro biomimetic tissue models is highlighted. Discussions of hydrogel sources and the latest hybrid system with different combinations of biopolymers are also presented. The hydrogel crosslinking mechanism and design consideration are summarized, followed by different types of available hydrogel module systems along with recent microfabrication technologies. We also present the latest developments in engineering hydrogel-based 3D in vitro models targeting specific tissues. Finally, we discuss the challenges surrounding current in vitro platforms and 3D models in the light of future perspectives for an improved biomimetic in vitro organ system.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
- Department of Smart Health Science and Technology, Kangwon National University (KNU), Chuncheon 24341, Korea
| |
Collapse
|
21
|
Malekahmadi B, Esfahanian V, Ejeian F, Dastgurdi ME, Agheb M, Kaveian F, Rafienia M, Nasr-Esfahani MH. In Vitro Study of the Recruitment and Expansion of Mesenchymal Stem Cells at the Interface of a Cu-Doped PCL-Bioglass Scaffold. Biomimetics (Basel) 2022; 7:biomimetics7010019. [PMID: 35225912 PMCID: PMC8883986 DOI: 10.3390/biomimetics7010019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/01/2022] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
Developing new barrier membranes with improved biomechanical characteristics has acquired much interest owing to their crucial role in the field of periodontal tissue regeneration. In this regard, we enriched the electrospun polycaprolactone (PCL)/gelatin (Gel) membranes by adding bioglass (BG) or Cu-doped bioglass (CuBG) and examined their cellular adhesion and proliferation potential in the presence of alveolar bone marrow-derived mesenchymal stem cells (aBMSCs). The membranes were fabricated and characterized using mechanical strength, SEM, FTIR, EDX, and ICP assay. Besides, aBMSCs were isolated, characterized, and seeded with a density of 35,000 cells in each experimental group. Next, the cellular morphology, cell adhesion capacity, proliferation rate, and membrane antibacterial activity were assessed. The results displayed a significant improvement in the wettability, pore size, and Young’s modulus of the PCL membrane following the incorporation of gelatin and CuBG particles. Moreover, all scaffolds exhibited reasonable biocompatibility and bioactivity in physiological conditions. Although the PCL/Gel/CuBG membrane revealed the lowest primary cell attachment, cells were grown properly and reached the confluent state after seven days. In conclusion, we found a reasonable level of attachment and proliferation of aBMSCs on all modified membranes. Meanwhile, a trace amount of Cu provided superiority for PCL/Gel/CuBG in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Behnaz Malekahmadi
- Isfahan (Khorasgan) Branch, Dental School, Islamic Azad University, Isfahan 8155139998, Iran;
- Cell Science Research Center, Department of Animal Biotechnology, Royan Institute for Biotechnology, ACECR, Isfahan 8159358686, Iran;
| | - Vahid Esfahanian
- Isfahan (Khorasgan) Branch, Department of Periodontics, Dental School, Islamic Azad University, Isfahan 8155139998, Iran
- Correspondence: (V.E.); (M.H.N.-E.); Tel.: +98-31-95015680 (M.H.N.-E.); Fax: +98-31-95015687 (M.H.N.-E.)
| | - Fatemeh Ejeian
- Cell Science Research Center, Department of Animal Biotechnology, Royan Institute for Biotechnology, ACECR, Isfahan 8159358686, Iran;
| | | | - Maria Agheb
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Faranak Kaveian
- Biosensor Research Center, Isfahan University of Medical Science, Isfahan 8174673461, Iran; (F.K.); (M.R.)
| | - Mohammad Rafienia
- Biosensor Research Center, Isfahan University of Medical Science, Isfahan 8174673461, Iran; (F.K.); (M.R.)
| | - Mohammad Hossein Nasr-Esfahani
- Cell Science Research Center, Department of Animal Biotechnology, Royan Institute for Biotechnology, ACECR, Isfahan 8159358686, Iran;
- Correspondence: (V.E.); (M.H.N.-E.); Tel.: +98-31-95015680 (M.H.N.-E.); Fax: +98-31-95015687 (M.H.N.-E.)
| |
Collapse
|
22
|
Li Y, Fraser D, Mereness J, Van Hove A, Basu S, Newman M, Benoit DSW. Tissue Engineered Neurovascularization Strategies for Craniofacial Tissue Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:20-39. [PMID: 35014834 PMCID: PMC9016342 DOI: 10.1021/acsabm.1c00979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Craniofacial tissue injuries, diseases, and defects, including those within bone, dental, and periodontal tissues and salivary glands, impact an estimated 1 billion patients globally. Craniofacial tissue dysfunction significantly reduces quality of life, and successful repair of damaged tissues remains a significant challenge. Blood vessels and nerves are colocalized within craniofacial tissues and act synergistically during tissue regeneration. Therefore, the success of craniofacial regenerative approaches is predicated on successful recruitment, regeneration, or integration of both vascularization and innervation. Tissue engineering strategies have been widely used to encourage vascularization and, more recently, to improve innervation through host tissue recruitment or prevascularization/innervation of engineered tissues. However, current scaffold designs and cell or growth factor delivery approaches often fail to synergistically coordinate both vascularization and innervation to orchestrate successful tissue regeneration. Additionally, tissue engineering approaches are typically investigated separately for vascularization and innervation. Since both tissues act in concert to improve craniofacial tissue regeneration outcomes, a revised approach for development of engineered materials is required. This review aims to provide an overview of neurovascularization in craniofacial tissues and strategies to target either process thus far. Finally, key design principles are described for engineering approaches that will support both vascularization and innervation for successful craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David Fraser
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Amy Van Hove
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Sayantani Basu
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States.,Materials Science Program, University of Rochester, Rochester, New York 14627, United States.,Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
23
|
Chaturvedi I, Jandyal A, Wazir I, Raina A, Ul Haq MI. Biomimetics and 3D printing - Opportunities for design applications. SENSORS INTERNATIONAL 2022. [DOI: 10.1016/j.sintl.2022.100191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
24
|
McGue CM, Mañón VA, Viet CT. Advances in Tissue Engineering and Implications for Oral and Maxillofacial Reconstruction. JOURNAL OF THE CALIFORNIA DENTAL ASSOCIATION 2021; 49:685-694. [PMID: 34887651 PMCID: PMC8653764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Reconstructive surgery in the oral and maxillofacial region poses many challenges due to the complexity of the facial skeleton and the presence of composite defects involving soft tissue, bone and nerve defects. METHODS Current methods of reconstruction include autologous grafting techniques with local or regional rotational flaps or microvascular free flaps, allografts, xenografts and prosthetic devices. RESULTS Tissue engineering therapies utilizing stem cells provide promise for enhancing the current reconstructive options. CONCLUSIONS This article is a review on tissue engineering strategies applicable to specialists who treat oral and maxillofacial defects. PRACTICAL IMPLICATIONS We review advancements in hard tissue regeneration for dental rehabilitation, soft tissue engineering, nerve regeneration and innovative strategies for reconstruction of major defects.
Collapse
Affiliation(s)
- Caitlyn M McGue
- Department of oral and maxillofacial surgery at the Loma Linda University School of Dentistry
| | - Victoria A Mañón
- Department of oral and maxillofacial surgery at the University of Texas Health Science Center at Houston School of Dentistry
| | - Chi T Viet
- Department of oral and maxillofacial surgery at the Loma Linda University School of Dentistry
| |
Collapse
|
25
|
Guo L, Liang Z, Yang L, Du W, Yu T, Tang H, Li C, Qiu H. The role of natural polymers in bone tissue engineering. J Control Release 2021; 338:571-582. [PMID: 34481026 DOI: 10.1016/j.jconrel.2021.08.055] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/31/2022]
Abstract
Bone is a dynamic self-healing organ and a continuous remodeling ensures the restoration of the bone structure and function over time. However, bone remodeling is not able to repair large traumatic injuries. Therefore, surgical interventions and bone substitutes are required. The aim of bone tissue engineering is to repair and regenerate tissues and engineered a bone graft as a bone substitute. To met this goal, several natural or synthetic polymers have been used to develop a biocompatible and biodegradable polymeric construct. Among the polymers, natural polymers have higher biocompatibility, excellent biodegradability, and no toxicity. So far, collagen, chitosan, gelatin, silk fibroin, alginate, cellulose, and starch, alone or in combination, have been widely used in bone tissue engineering. These polymers have been used as scaffolds, hydrogels, and micro-nanospheres. The functionalization of the polymer with growth factors and bioactive glasses increases the potential use of polymers for bone regeneration. As bone is a dynamic highly vascularized tissue, the vascularization of the polymeric scaffolds is vital for successful bone regeneration. Several in vivo and in vitro strategies have been used to vascularize the polymeric scaffolds. In this review, the application of the most commonly used natural polymers is discussed.
Collapse
Affiliation(s)
- Linqi Guo
- Department of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154000, China
| | - Zhihui Liang
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi 154000, China
| | - Liang Yang
- Department of Orthopaedics, The People's Hospital of Daqing, Daqing 163000, China
| | - Wenyan Du
- Department of Orthopaedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154000, China
| | - Tao Yu
- Department of Orthopaedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154000, China
| | - Huayu Tang
- Department of Orthopaedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154000, China
| | - Changde Li
- Department of Orthopaedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154000, China
| | - Hongbin Qiu
- Department of Public Health, Jiamusi University, Jiamusi, 154000, China.
| |
Collapse
|
26
|
Redenski I, Guo S, Machour M, Szklanny A, Landau S, Kaplan B, Lock RI, Gabet Y, Egozi D, Vunjak‐Novakovic G, Levenberg S. Engineered Vascularized Flaps, Composed of Polymeric Soft Tissue and Live Bone, Repair Complex Tibial Defects. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2008687. [DOI: 10.1002/adfm.202008687] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 02/05/2023]
Affiliation(s)
- Idan Redenski
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Shaowei Guo
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
- The First Affiliated Hospital Shantou University Medical College Shantou 515000 China
| | - Majd Machour
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Ariel Szklanny
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Shira Landau
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Ben Kaplan
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Roberta I. Lock
- Department of Biomedical Engineering Columbia University New York NY 10032 USA
| | - Yankel Gabet
- Department of Anatomy and Anthropology Sackler Faculty of Medicine Tel‐Aviv University Tel‐Aviv 6997801 Israel
| | - Dana Egozi
- Department of Plastic and Reconstructive Surgery Kaplan Hospital Rehovot and the Hebrew University Jerusalem 7661041 Israel
| | | | - Shulamit Levenberg
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| |
Collapse
|
27
|
Jamalpoor Z, Taromi N. Pre-vascularization of biomimetic 3-D scaffolds via direct co-culture of human umbilical cord derived osteogenic and angiogenic progenitor cells. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
28
|
Pirosa A, Tankus EB, Mainardi A, Occhetta P, Dönges L, Baum C, Rasponi M, Martin I, Barbero A. Modeling In Vitro Osteoarthritis Phenotypes in a Vascularized Bone Model Based on a Bone-Marrow Derived Mesenchymal Cell Line and Endothelial Cells. Int J Mol Sci 2021; 22:ijms22179581. [PMID: 34502489 PMCID: PMC8430538 DOI: 10.3390/ijms22179581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
The subchondral bone and its associated vasculature play an important role in the onset of osteoarthritis (OA). Integration of different aspects of the OA environment into multi-cellular and complex human, in vitro models is therefore needed to properly represent the pathology. In this study, we exploited a mesenchymal stromal cell line/endothelial cell co-culture to produce an in vitro human model of vascularized osteogenic tissue. A cocktail of inflammatory cytokines, or conditioned medium from mechanically-induced OA engineered microcartilage, was administered to this vascularized bone model to mimic the inflamed OA environment, hypothesizing that these treatments could induce the onset of specific pathological traits. Exposure to the inflammatory factors led to increased network formation by endothelial cells, reminiscent of the abnormal angiogenesis found in OA subchondral bone, demineralization of the constructs, and increased collagen production, signs of OA related bone sclerosis. Furthermore, inflammation led to augmented expression of osteogenic (alkaline phosphatase (ALP) and osteocalcin (OCN)) and angiogenic (vascular endothelial growth factor (VEGF)) genes. The treatment, with a conditioned medium from the mechanically-induced OA engineered microcartilage, also caused increased demineralization and expression of ALP, OCN, ADAMTS5, and VEGF; however, changes in network formation by endothelial cells were not observed in this second case, suggesting a possible different mechanism of action in inducing OA-like phenotypes. We propose that this vascularized bone model could represent a first step for the in vitro study of bone changes under OA mimicking conditions and possibly serve as a tool in testing anti-OA drugs.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Esma Bahar Tankus
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Andrea Mainardi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
- Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
| | - Laura Dönges
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Cornelia Baum
- Department of Orthopaedic Surgery and Traumatology, University Hospital Basel, 4031 Basel, Switzerland;
- Department of Research and Development, Schulthess Klinik Zurich, 8008 Zurich, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
- Correspondence:
| |
Collapse
|
29
|
Lim KT, Patel DK, Dutta SD, Ganguly K. Fluid Flow Mechanical Stimulation-Assisted Cartridge Device for the Osteogenic Differentiation of Human Mesenchymal Stem Cells. MICROMACHINES 2021; 12:927. [PMID: 34442549 PMCID: PMC8398302 DOI: 10.3390/mi12080927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have the potential to differentiate into different types of mesodermal tissues. In vitro proliferation and differentiation of hMSCs are necessary for bone regeneration in tissue engineering. The present study aimed to design and develop a fluid flow mechanically-assisted cartridge device to enhance the osteogenic differentiation of hMSCs. We used the fluorescence-activated cell-sorting method to analyze the multipotent properties of hMSCs and found that the cultured cells retained their stemness potential. We also evaluated the cell viabilities of the cultured cells via water-soluble tetrazolium salt 1 (WST-1) assay under different rates of flow (0.035, 0.21, and 0.35 mL/min) and static conditions and found that the cell growth rate was approximately 12% higher in the 0.035 mL/min flow condition than the other conditions. Moreover, the cultured cells were healthy and adhered properly to the culture substrate. Enhanced mineralization and alkaline phosphatase activity were also observed under different perfusion conditions compared to the static conditions, indicating that the applied conditions play important roles in the proliferation and differentiation of hMSCs. Furthermore, we determined the expression levels of osteogenesis-related genes, including the runt-related protein 2 (Runx2), collagen type I (Col1), osteopontin (OPN), and osteocalcin (OCN), under various perfusion vis-à-vis static conditions and found that they were significantly affected by the applied conditions. Furthermore, the fluorescence intensities of OCN and OPN osteogenic gene markers were found to be enhanced in the 0.035 mL/min flow condition compared to the control, indicating that it was a suitable condition for osteogenic differentiation. Taken together, the findings of this study reveal that the developed cartridge device promotes the proliferation and differentiation of hMSCs and can potentially be used in the field of tissue engineering.
Collapse
Affiliation(s)
- Ki-Taek Lim
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
- Biomechagen Co., Ltd., Chuncheon 24341, Korea
| | - Dinesh-K. Patel
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| | - Sayan-Deb Dutta
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| | - Keya Ganguly
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| |
Collapse
|
30
|
Vascularization Strategies in Bone Tissue Engineering. Cells 2021; 10:cells10071749. [PMID: 34359919 PMCID: PMC8306064 DOI: 10.3390/cells10071749] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.
Collapse
|
31
|
Whelan IT, Moeendarbary E, Hoey DA, Kelly DJ. Biofabrication of vasculature in microphysiological models of bone. Biofabrication 2021; 13. [PMID: 34034238 DOI: 10.1088/1758-5090/ac04f7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022]
Abstract
Bone contains a dense network of blood vessels that are essential to its homoeostasis, endocrine function, mineral metabolism and regenerative functions. In addition, bone vasculature is implicated in a number of prominent skeletal diseases, and bone has high affinity for metastatic cancers. Despite vasculature being an integral part of bone physiology and pathophysiology, it is often ignored or oversimplified inin vitrobone models. However, 3D physiologically relevant vasculature can now be engineeredin vitro, with microphysiological systems (MPS) increasingly being used as platforms for engineering this physiologically relevant vasculature. In recent years, vascularised models of bone in MPSs systems have been reported in the literature, representing the beginning of a possible technological step change in how bone is modelledin vitro. Vascularised bone MPSs is a subfield of bone research in its nascency, however given the impact of MPSs has had inin vitroorgan modelling, and the crucial role of vasculature to bone physiology, these systems stand to have a substantial impact on bone research. However, engineering vasculature within the specific design restraints of the bone niche is significantly challenging given the different requirements for engineering bone and vasculature. With this in mind, this paper aims to serve as technical guidance for the biofabrication of vascularised bone tissue within MPS devices. We first discuss the key engineering and biological considerations for engineering more physiologically relevant vasculaturein vitrowithin the specific design constraints of the bone niche. We next explore emerging applications of vascularised bone MPSs, and conclude with a discussion on the current status of vascularised bone MPS biofabrication and suggest directions for development of next generation vascularised bone MPSs.
Collapse
|
32
|
Masson-Meyers DS, Tayebi L. Vascularization strategies in tissue engineering approaches for soft tissue repair. J Tissue Eng Regen Med 2021; 15:747-762. [PMID: 34058083 DOI: 10.1002/term.3225] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Insufficient vascularization during tissue repair is often associated with poor clinical outcomes. This is a concern especially when patients have critical-sized injuries, where the size of the defect restricts vascularity, or even in small defects that have to be treated under special conditions, such as after radiation therapy (relevant to tumor resection) that hinders vascularity. In fact, poor vascularization is one of the major obstacles for clinical application of tissue engineering methods in soft tissue repair. As a key issue, lack of graft integration, caused by inadequate vascularization after implantation, can lead to graft failure. Moreover, poor vascularization compromises the viability of cells seeded in deep portions of scaffolds/graft materials, due to hypoxia and insufficient nutrient supply. In this article we aim to review vascularization strategies employed in tissue engineering techniques to repair soft tissues. For this purpose, we start by providing a brief overview of the main events during the physiological wound healing process in soft tissues. Then, we discuss how tissue repair can be achieved through tissue engineering, and considerations with regards to the choice of scaffold materials, culture conditions, and vascularization techniques. Next, we highlight the importance of vascularization, along with strategies and methods of prevascularization of soft tissue equivalents, particularly cell-based prevascularization. Lastly, we present a summary of commonly used in vitro methods during the vascularization of tissue-engineered soft tissue constructs.
Collapse
Affiliation(s)
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| |
Collapse
|
33
|
Bouland C, Philippart P, Dequanter D, Corrillon F, Loeb I, Bron D, Lagneaux L, Meuleman N. Cross-Talk Between Mesenchymal Stromal Cells (MSCs) and Endothelial Progenitor Cells (EPCs) in Bone Regeneration. Front Cell Dev Biol 2021; 9:674084. [PMID: 34079804 PMCID: PMC8166285 DOI: 10.3389/fcell.2021.674084] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Bone regeneration is a complex, well-orchestrated process based on the interactions between osteogenesis and angiogenesis, observed in both physiological and pathological situations. However, specific conditions (e.g., bone regeneration in large quantity, immunocompromised regenerative process) require additional support. Tissue engineering offers novel strategies. Bone regeneration requires a cell source, a matrix, growth factors and mechanical stimulation. Regenerative cells, endowed with proliferation and differentiation capacities, aim to recover, maintain, and improve bone functions. Vascularization is mandatory for bone formation, skeletal development, and different osseointegration processes. The latter delivers nutrients, growth factors, oxygen, minerals, etc. The development of mesenchymal stromal cells (MSCs) and endothelial progenitor cells (EPCs) cocultures has shown synergy between the two cell populations. The phenomena of osteogenesis and angiogenesis are intimately intertwined. Thus, cells of the endothelial line indirectly foster osteogenesis, and conversely, MSCs promote angiogenesis through different interaction mechanisms. In addition, various studies have highlighted the importance of the microenvironment via the release of extracellular vesicles (EVs). These EVs stimulate bone regeneration and angiogenesis. In this review, we describe (1) the phenomenon of bone regeneration by different sources of MSCs. We assess (2) the input of EPCs in coculture in bone regeneration and describe their contribution to the osteogenic potential of MSCs. We discuss (3) the interaction mechanisms between MSCs and EPCs in the context of osteogenesis: direct or indirect contact, production of growth factors, and the importance of the microenvironment via the release of EVs.
Collapse
Affiliation(s)
- Cyril Bouland
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Philippart
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Stomatology and Maxillofacial Surgery, IRIS South Hospital, Brussels, Belgium
| | - Didier Dequanter
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Florent Corrillon
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Loeb
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Dominique Bron
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
34
|
Louvrier A, Terranova L, Meyer C, Meyer F, Euvrard E, Kroemer M, Rolin G. Which experimental models and explorations to use in regenerative endodontics? A comprehensive review on standard practices. Mol Biol Rep 2021; 48:3799-3812. [PMID: 33761086 DOI: 10.1007/s11033-021-06299-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/18/2021] [Indexed: 01/09/2023]
Abstract
Since the discovery of dental pulp stem cells, a lot of teams have expressed an interest in dental pulp regeneration. Many approaches, experimental models and biological explorations have been developed, each including the use of stem cells and scaffolds with the final goal being clinical application in humans. In this review, the authors' objective was to compare the experimental models and strategies used for the development of biomaterials for tissue engineering of dental pulp with stem cells. Electronic queries were conducted on PubMed using the following terms: pulp regeneration, scaffold, stem cells, tissue engineering and biomaterial. The extracted data included the following information: the strategy envisaged, the type of stem cells, the experimental models, the exploration or analysis methods, the cytotoxicity or viability or proliferation cellular tests, the tests of scaffold antibacterial properties and take into account the vascularization of the regenerated dental pulp. From the 71 selected articles, 59% focused on the "cell-transplantation" strategy, 82% used in vitro experimentation, 58% in vivo animal models and only one described an in vivo in situ human clinical study. 87% used dental pulp stem cells. A majority of the studies reported histology (75%) and immunohistochemistry explorations (66%). 73% mentioned the use of cytotoxicity, proliferation or viability tests. 48% took vascularization into account but only 6% studied the antibacterial properties of the scaffolds. This article gives an overview of the methods used to regenerate dental pulp from stem cells and should help researchers create the best development strategies for research in this field.
Collapse
Affiliation(s)
- A Louvrier
- Chirurgie Maxillo-Faciale, stomatologie et odontologie hospitalière, CHU Besançon, 25000, Besançon, France.
- UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, 25000, Besançon, France.
| | - L Terranova
- UMR_S 1121 Biomatériaux et Bioingénierie, Université de Strasbourg, INSERM, FMTS, Strasbourg, France
| | - C Meyer
- Chirurgie Maxillo-Faciale, stomatologie et odontologie hospitalière, CHU Besançon, 25000, Besançon, France
- Laboratoire Nano Médecine, Imagerie, Thérapeutique, Univ. Bourgogne Franche-Comté, EA 4662, 25000, Besançon, France
| | - F Meyer
- UMR_S 1121 Biomatériaux et Bioingénierie, Université de Strasbourg, INSERM, FMTS, Strasbourg, France
| | - E Euvrard
- Chirurgie Maxillo-Faciale, stomatologie et odontologie hospitalière, CHU Besançon, 25000, Besançon, France
- Laboratoire Nano Médecine, Imagerie, Thérapeutique, Univ. Bourgogne Franche-Comté, EA 4662, 25000, Besançon, France
| | - M Kroemer
- UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, 25000, Besançon, France
- Pharmacie Centrale, CHU Besançon, 25000, Besançon, France
| | - G Rolin
- UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, 25000, Besançon, France
- INSERM CIC-1431, CHU Besançon, 25000, Besançon, France
| |
Collapse
|
35
|
Pirosa A, Gottardi R, Alexander PG, Puppi D, Chiellini F, Tuan RS. An in vitro chondro-osteo-vascular triphasic model of the osteochondral complex. Biomaterials 2021; 272:120773. [PMID: 33798958 DOI: 10.1016/j.biomaterials.2021.120773] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 01/06/2023]
Abstract
The generation of engineered models of the osteochondral complex to study its pathologies and develop possible treatments is hindered by the distinctly different properties of articular cartilage and subchondral bone, with the latter characterized by vascularization. In vitro models of the osteochondral complex have been mainly engineered as biphasic constructs containing just cartilage and bone cells, a condition very dissimilar from the in vivo environment. The different cellular components of the osteochondral complex are governed by interacting biochemical signaling; hence, to study the crosstalk among chondrocytes, osteoblasts, and endothelial cells, we have developed a novel triphasic model of the osteochondral tissue interface. Wet-spun poly(ε-caprolactone) (PCL) and PCL/hydroxyapatite (HA) scaffolds in combination with a methacrylated gelatin (gelMA) hydrogel were used as the polymeric backbone of the constructs. The scaffold components were engineered with human bone marrow derived mesenchymal stem cells (hMSCs) and human umbilical vein endothelial cells (HUVECs), and differentiated using a dual chamber microphysiological system (MPS) bioreactor that allows the simultaneous, separate flow of media of different compositions for induced differentiation of each compartment towards a cartilaginous or osseous lineage. Within the engineered Microphysiological Vascularized Osteochondral System, hMSCs showed spatially distinct chondrogenic and osteogenic markers in terms of histology and gene expression. HUVECs formed a stable capillary-like network in the engineered bone compartment and enhanced both chondrogenic and osteogenic differentiation of hMSCs, resulting in the generation of an in vitro system that mimics a vascularized osteochondral interface tissue.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Ri.MED Foundation, Palermo, Italy
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dario Puppi
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Federica Chiellini
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
36
|
Hu L, Wang Y, Pan H, Kadir K, Wen J, Li S, Zhang C. Apoptosis repressor with caspase recruitment domain (ARC) promotes bone regeneration of bone marrow-derived mesenchymal stem cells by activating Fgf-2/PI3K/Akt signaling. Stem Cell Res Ther 2021; 12:185. [PMID: 33726822 PMCID: PMC7962397 DOI: 10.1186/s13287-021-02253-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/28/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives This study aims to investigate whether apoptosis repressor with caspase recruitment domain (ARC) could promote survival and enhance osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Materials and methods The lentivirus transfection method was used to establish ARC-overexpressing BMSCs. The CCK-8 method was used to detect cell proliferation. The BD Pharmingen™ APC Annexin V Apoptosis Detection kit was used to detect cell apoptosis. The osteogenic capacity was investigated by OCN immunofluorescence staining, ALP analysis, ARS assays, and RT-PCR analysis. Cells were seeded into calcium phosphate cement (CPC) scaffolds and then inserted subcutaneously into nude mice and the defect area of the rat calvarium. Histological analysis was conducted to evaluate the in vivo cell apoptosis and new bone formation of the ARC-overexpressing BMSCs. RNA-seq was used to detect the possible mechanism of the effect of ARC on BMSCs. Results ARC promoted BMSC proliferation and inhibited cell apoptosis. ARC enhanced BMSC osteogenic differentiation in vitro. An in vivo study revealed that ARC can inhibit BMSC apoptosis and increase new bone formation. ARC regulates BMSCs mainly by activating the Fgf-2/PI3K/Akt pathway. Conclusions The present study suggests that ARC is a powerful agent for promoting bone regeneration of BMSCs and provides a promising method for bone tissue engineering.
Collapse
Affiliation(s)
- Longwei Hu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology& Shanghai Research Institute of Stomatology, Shanghai, 200011, People's Republic of China
| | - Yang Wang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology& Shanghai Research Institute of Stomatology, Shanghai, 200011, People's Republic of China
| | - Hongya Pan
- Linno Pharmaceuticals Inc., Shanghai, 200011, People's Republic of China
| | - Kathreena Kadir
- Department of Oral & Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jin Wen
- Department of Prosthodontics, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, People's Republic of China
| | - Siyi Li
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology& Shanghai Research Institute of Stomatology, Shanghai, 200011, People's Republic of China.
| | - Chenping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology& Shanghai Research Institute of Stomatology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
37
|
Fu L, Zhang L, Zhang X, Chen L, Cai Q, Yang X. Roles of oxygen level and hypoxia-inducible factor signaling pathway in cartilage, bone and osteochondral tissue engineering. Biomed Mater 2021; 16:022006. [PMID: 33440367 DOI: 10.1088/1748-605x/abdb73] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The repair and treatment of articular cartilage injury is a huge challenge of orthopedics. Currently, most of the clinical methods applied in treating cartilage injuries are mainly to relieve pains rather than to cure them, while the strategy of tissue engineering is highly expected to achieve the successful repair of osteochondral defects. Clear understandings of the physiological structures and mechanical properties of cartilage, bone and osteochondral tissues have been established, but the understanding of their physiological heterogeneity still needs further investigation. Apart from the gradients in the micromorphology and composition of cartilage-to-bone extracellular matrixes, an oxygen gradient also exists in natural osteochondral tissue. The response of hypoxia-inducible factor (HIF)-mediated cells to oxygen would affect the differentiation of stem cells and the maturation of osteochondral tissue. This article reviews the roles of oxygen level and HIF signaling pathway in the development of articular cartilage tissue, and their prospective applications in bone and cartilage tissue engineering. The strategies for regulating HIF signaling pathway and how these strategies finding their potential applications in the regeneration of integrated osteochondral tissue are also discussed.
Collapse
Affiliation(s)
- Lei Fu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People's Republic of China
| | | | | | | | | | | |
Collapse
|
38
|
Poustchi F, Amani H, Ahmadian Z, Niknezhad SV, Mehrabi S, Santos HA, Shahbazi M. Combination Therapy of Killing Diseases by Injectable Hydrogels: From Concept to Medical Applications. Adv Healthc Mater 2021; 10:e2001571. [PMID: 33274841 DOI: 10.1002/adhm.202001571] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/13/2020] [Indexed: 01/16/2023]
Abstract
The complexity of hard-to-treat diseases strongly undermines the therapeutic potential of available treatment options. Therefore, a paradigm shift from monotherapy toward combination therapy has been observed in clinical research to improve the efficiency of available treatment options. The advantages of combination therapy include the possibility of synchronous alteration of different biological pathways, reducing the required effective therapeutic dose, reducing drug resistance, and lowering the overall costs of treatment. The tunable physical properties, excellent biocompatibility, facile preparation, and ease of administration with minimal invasiveness of injectable hydrogels (IHs) have made them excellent candidates to solve the clinical and pharmacological limitations of present systems for multitherapy by direct delivery of therapeutic payloads and improving therapeutic responses through the formation of depots containing drugs, genes, cells, or a combination of them in the body after a single injection. In this review, currently available methods for the design and fabrication of IHs are systematically discussed in the first section. Next, as a step toward establishing IHs for future multimodal synergistic therapies, recent advances in cancer combination therapy, wound healing, and tissue engineering are addressed in detail in the following sections. Finally, opportunities and challenges associated with IHs for multitherapy are listed and further discussed.
Collapse
Affiliation(s)
- Fatemeh Poustchi
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki FI‐00014 Finland
- Department of Nanotechnology University of Guilan Rasht Guilan 41996‐13765 Iran
| | - Hamed Amani
- Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology Iran University of Medical Science Tehran 14496‐14535 Iran
| | - Zainab Ahmadian
- Department of Pharmaceutics School of Pharmacy Zanjan University of Medical Science Zanjan 45139‐56184 Iran
| | - Seyyed Vahid Niknezhad
- Burn and Wound Healing Research Center Shiraz University of Medical Sciences Shiraz 71987‐54361 Iran
| | - Soraya Mehrabi
- Faculty of Medicine, Department of Physiology Iran University of Medical Sciences Tehran 14496‐14535 Iran
| | - Hélder A. Santos
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki FI‐00014 Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki Helsinki FI‐00014 Finland
| | - Mohammad‐Ali Shahbazi
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki FI‐00014 Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC) Zanjan University of Medical Sciences Zanjan 45139‐56184 Iran
| |
Collapse
|
39
|
Masuda H, Arisaka Y, Hakariya M, Iwata T, Yoda T, Yui N. Synergy of molecularly mobile polyrotaxane surfaces with endothelial cell co-culture for mesenchymal stem cell mineralization. RSC Adv 2021; 11:18685-18692. [PMID: 35480955 PMCID: PMC9033494 DOI: 10.1039/d1ra01296g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/16/2021] [Indexed: 11/26/2022] Open
Abstract
Stem cell-based bone tissue engineering is a promising strategy for the treatment of bone defects. Since regeneration of bone tissue takes a long time, promoting osteogenesis of stem cells is desired for earlier recovery from dysfunctions caused by bone defects. Here, we combined endothelial cell co-culture using the molecularly mobile sulfonated polyrotaxane (PRX) surfaces to enhance the mineralization of human bone marrow derived mesenchymal stem cells (HBMSCs). Sulfonated PRXs are composed of sulfopropyl ether-modified α-cyclodextrins (α-CDs) threaded on a polyethylene glycol chain. The molecular mobility of PRX, α-CDs moving along the polymer, can be modulated by the number of α-CDs. When osteoblastic differentiation was induced in HBMSCs and human umbilical vein endothelial cells (HUVECs), co-culture groups on sulfonated PRX surfaces with low molecular mobility showed the highest mineralization, which is about two times as high as co-culture groups on sulfonated PRX surfaces with high molecular mobility. Nuclear accumulation of yes-associated proteins in HBMSCs and cell–cell communication via cytokines or cadherin may play an important role in synergistically induced mineralization of HBMSCs. Molecular mobility of polyrotaxane surfaces promoted mineralization in a co-culture system of mesenchymal stem cells and endothelial cells.![]()
Collapse
Affiliation(s)
- Hiroki Masuda
- Department of Maxillofacial Surgery
- Graduate School of Medical and Dental Sciences
- Tokyo Medical and Dental University (TMDU)
- Bunkyo
- Japan
| | - Yoshinori Arisaka
- Department of Organic Biomaterials
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University (TMDU)
- Chiyoda
- Japan
| | - Masahiro Hakariya
- Department of Periodontology
- Graduate School of Medical and Dental Sciences
- Tokyo Medical and Dental University (TMDU)
- Bunkyo
- Japan
| | - Takanori Iwata
- Department of Periodontology
- Graduate School of Medical and Dental Sciences
- Tokyo Medical and Dental University (TMDU)
- Bunkyo
- Japan
| | - Tetsuya Yoda
- Department of Maxillofacial Surgery
- Graduate School of Medical and Dental Sciences
- Tokyo Medical and Dental University (TMDU)
- Bunkyo
- Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University (TMDU)
- Chiyoda
- Japan
| |
Collapse
|
40
|
Li J, Zhu Y, Li N, Wu T, Zheng X, Heng BC, Zou D, Xu J. Upregulation of ETV2 Expression Promotes Endothelial Differentiation of Human Dental Pulp Stem Cells. Cell Transplant 2021; 30:963689720978739. [PMID: 33522307 PMCID: PMC7863555 DOI: 10.1177/0963689720978739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/30/2020] [Accepted: 12/12/2020] [Indexed: 11/26/2022] Open
Abstract
The lack of vasculogenesis often hampers the survivability and integration of newly engineered tissue grafts within the host. Autologous endothelial cells (ECs) are an ideal cell source for neovascularization, but they are limited by their scarcity, lack of proliferative capacity, and donor site morbidity upon isolation. The objective of this study was to determine whether differentiation of human dental pulp stem cells (DPSCs) into the endothelial lineage can be enhanced by recombinant ETV2 overexpression. DPSCs were extracted from fresh dental pulp tissues. ETV2 overexpression in DPSCs was achieved by lentiviral infection and cellular morphological changes were evaluated. The mRNA and protein expression levels of endothelial-specific markers were assessed through quantitative real-time polymerase chain reaction, western blot, immunofluorescence staining, and flow cytometry. The tube formation assay and Matrigel plug assay were also performed to evaluate the angiogenic potential of the ETV2-transduced cells in vitro and in vivo, respectively. Additionally, proteomic analysis was performed to analyze global changes in protein expression following ETV2 overexpression. After lentiviral infection, ETV2-overexpressing DPSCs showed endothelial-like morphology. Compared with control DPSCs, significantly higher mRNA and protein expression levels of endothelial-specific genes, including CD31, VE-Cadherin, VEGFR1, and VEGFR2, were detected in ETV2-overexpressing DPSCs. Moreover, ETV2 overexpression enhanced capillary-like tube formation on Matrigel in vitro, as well as neovascularization in vivo. In addition, comparative proteomic profiling showed that ETV2 overexpression upregulated the expression of vascular endothelial growth factor (VEGF) receptors, which was indicative of increased VEGF signaling. Taken together, our results indicate that ETV2 overexpression significantly enhanced the endothelial differentiation of DPSCs. Thus, this study shows that DPSCs can be a promising candidate cell source for tissue engineering applications.
Collapse
Affiliation(s)
- Jing Li
- Department of Orthodontics, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, P. R. China
| | - Youming Zhu
- Department of Orthodontics, Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, P. R. China
| | - Na Li
- Department of Orthodontics, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, P. R. China
| | - Tao Wu
- Department of Orthodontics, Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, P. R. China
| | - Xianyu Zheng
- Department of Orthodontics, Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, P. R. China
| | - Boon chin Heng
- Central Laboratories, School of Stomatology, Peking University, Beijing, P. R. China
| | - Duohong Zou
- Department of Oral Surgery, Shanghai Key Laboratory of Stomatology, National Clinical Research Center of Stomatology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Jianguang Xu
- Department of Orthodontics, Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, P. R. China
| |
Collapse
|
41
|
Jin Q, Li P, Yuan K, Zhao F, Zhu X, Zhang P, Huang Z. Extracellular vesicles derived from human dental pulp stem cells promote osteogenesis of adipose-derived stem cells via the MAPK pathway. J Tissue Eng 2020; 11:2041731420975569. [PMID: 33312494 PMCID: PMC7716067 DOI: 10.1177/2041731420975569] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
Recent studies have shown that co-culture systems play an important role in bone tissue engineering. In this study, human dental pulp stem cells (hDPSCs) were co-cultured with human adipose-derived stem cells (hADSCs), and osteoblastic phenotypes were found to be enhanced in co-cultures compared with monocultures of hDPSCs or hADSCs. Furthermore, GW4869, an inhibitor of extracellular vesicle (EV) formation, suppressed the mineralization of co-cultured cells. Studies indicate that the therapeutic potential of DPSCs is realized through paracrine action, in which EVs play an important role. To study their role, we successfully obtained and identified hDPSC-derived extracellular vesicles (hDPSC-EVs), and further investigated their effects on hADSCs and the underlying mechanism. hADSCs were stimulated with hDPSC-EVs, which were found to promote the migration and mineralization of hADSCs. Moreover, hDPSC-EVs promoted osteogenic differentiation by enhancing the phosphorylation of ERK 1/2 and JNK in hADSCs. To investigate the specific proteins in EVs that might play a role in hADSC osteogenic differentiation, we performed proteomic analysis of hDPSC-EVs. We determined the top 30 enriched pathways, which notably included the insulin signaling pathway. The number of genes enriched in the insulin signaling pathway was the largest, in addition to the “protein processing in endoplasmic reticulum” term. The MAPK cascade is a typical downstream pathway mediating insulin signaling. To further study the effects of hDPSC-EVs on maxillofacial bone regeneration, we used hDPSC-EVs as a cell-free biomaterial in a model of mandibular defects in rats. To assess the therapeutic potential of EVs, we analyzed their proteome. Animal experiments demonstrated that hDPSC-EVs promoted the regeneration of bone defects. Overall, these results highlight the potential of hDPSC-EVs to induce lineage specific differentiation of hADSCs. The results also indicated the importance of considering hDPSC-EVs as biomimetic materials for clinical translation of treatments for oral maxillofacial defects.
Collapse
Affiliation(s)
- Qiaoqiao Jin
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Peilun Li
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Keyong Yuan
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Fen Zhao
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xiaohan Zhu
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Pengfei Zhang
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhengwei Huang
- Department of Endodontics, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
42
|
Chen Y, Ouyang X, Wu Y, Guo S, Xie Y, Wang G. Co-culture and Mechanical Stimulation on Mesenchymal Stem Cells and Chondrocytes for Cartilage Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:54-60. [PMID: 31660820 DOI: 10.2174/1574888x14666191029104249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023]
Abstract
Defects in articular cartilage injury and chronic osteoarthritis are very widespread and common, and the ability of injured cartilage to repair itself is limited. Stem cell-based cartilage tissue engineering provides a promising therapeutic option for articular cartilage damage. However, the application of the technique is limited by the number, source, proliferation, and differentiation of stem cells. The co-culture of mesenchymal stem cells and chondrocytes is available for cartilage tissue engineering, and mechanical stimulation is an important factor that should not be ignored. A combination of these two approaches, i.e., co-culture of mesenchymal stem cells and chondrocytes under mechanical stimulation, can provide sufficient quantity and quality of cells for cartilage tissue engineering, and when combined with scaffold materials and cytokines, this approach ultimately achieves the purpose of cartilage repair and reconstruction. In this review, we focus on the effects of co-culture and mechanical stimulation on mesenchymal stem cells and chondrocytes for articular cartilage tissue engineering. An in-depth understanding of the impact of co-culture and mechanical stimulation of mesenchymal stem cells and chondrocytes can facilitate the development of additional strategies for articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yawen Chen
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Xinli Ouyang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yide Wu
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Shaojia Guo
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yongfang Xie
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Guohui Wang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| |
Collapse
|
43
|
Kim J, Kong JS, Han W, Kim BS, Cho DW. 3D Cell Printing of Tissue/Organ-Mimicking Constructs for Therapeutic and Drug Testing Applications. Int J Mol Sci 2020; 21:E7757. [PMID: 33092184 PMCID: PMC7589604 DOI: 10.3390/ijms21207757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The development of artificial tissue/organs with the functional maturity of their native equivalents is one of the long-awaited panaceas for the medical and pharmaceutical industries. Advanced 3D cell-printing technology and various functional bioinks are promising technologies in the field of tissue engineering that have enabled the fabrication of complex 3D living tissue/organs. Various requirements for these tissues, including a complex and large-volume structure, tissue-specific microenvironments, and functional vasculatures, have been addressed to develop engineered tissue/organs with native relevance. Functional tissue/organ constructs have been developed that satisfy such criteria and may facilitate both in vivo replenishment of damaged tissue and the development of reliable in vitro testing platforms for drug development. This review describes key developments in technologies and materials for engineering 3D cell-printed constructs for therapeutic and drug testing applications.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Wonil Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
44
|
Vis MAM, Ito K, Hofmann S. Impact of Culture Medium on Cellular Interactions in in vitro Co-culture Systems. Front Bioeng Biotechnol 2020; 8:911. [PMID: 32850750 PMCID: PMC7417654 DOI: 10.3389/fbioe.2020.00911] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/15/2020] [Indexed: 12/24/2022] Open
Abstract
Co-culturing of cells in in vitro tissue models is widely used to study how they interact with each other. These models serve to represent a variety of processes in the human body such as development, homeostasis, regeneration, and disease. The success of a co-culture is dependent on a large number of factors which makes it a complex and ambiguous task. This review article addresses co-culturing challenges regarding the cell culture medium used in these models, in particular concerning medium composition, volume, and exchange. The effect of medium exchange on cells is often an overlooked topic but particularly important when cell communication via soluble factors and extracellular vesicles, the so-called cell secretome (CS) is being studied. Culture medium is regularly exchanged to supply new nutrients and to eliminate waste products produced by the cells. By removing medium, important CSs are also removed. After every medium change, the cells must thus restore their auto- and paracrine communication through these CSs. This review article will also discuss the possibility to integrate biosensors into co-cultures, in particular to provide real-time information regarding media composition. Overall, the manner in which culture medium is currently used will be re-evaluated. Provided examples will be on the subject of bone tissue engineering.
Collapse
Affiliation(s)
- Michelle A M Vis
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
45
|
Borciani G, Montalbano G, Baldini N, Cerqueni G, Vitale-Brovarone C, Ciapetti G. Co-culture systems of osteoblasts and osteoclasts: Simulating in vitro bone remodeling in regenerative approaches. Acta Biomater 2020; 108:22-45. [PMID: 32251782 DOI: 10.1016/j.actbio.2020.03.043] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023]
Abstract
Bone is an extremely dynamic tissue, undergoing continuous remodeling for its whole lifetime, but its regeneration or augmentation due to bone loss or defects are not always easy to obtain. Bone tissue engineering (BTE) is a promising approach, and its success often relies on a "smart" scaffold, as a support to host and guide bone formation through bone cell precursors. Bone homeostasis is maintained by osteoblasts (OBs) and osteoclasts (OCs) within the basic multicellular unit, in a consecutive cycle of resorption and formation. Therefore, a functional scaffold should allow the best possible OB/OC cooperation for bone remodeling, as happens within the bone extracellular matrix in the body. In the present work OB/OC co-culture models, with and without scaffolds, are reviewed. These experimental systems are intended for different targets, including bone remodeling simulation, drug testing and the assessment of biomaterials and 3D scaffolds for BTE. As a consequence, several parameters, such as cell type, cell ratio, culture medium and inducers, culture times and setpoints, assay methods, etc. vary greatly. This review identifies and systematically reports the in vitro methods explored up to now, which, as they allow cellular communication, more closely resemble bone remodeling and/or the regeneration process in the framework of BTE. STATEMENT OF SIGNIFICANCE: Bone is a dynamic tissue under continuous remodeling, but spontaneous healing may fail in the case of excessive bone loss which often requires valid alternatives to conventional treatments to restore bone integrity, like bone tissue engineering (BTE). Pre-clinical evaluation of scaffolds for BTE requires in vitro testing where co-cultures combining innovative materials with osteoblasts (OBs) and osteoclasts (OCs) closely mimic the in vivo repair process. This review considers the direct and indirect OB/OC co-cultures relevant to BTE, from the early mouse-cell models to the recent bone regenerative systems. The co-culture modeling of bone microenvironment provides reliable information on bone cell cross-talk. Starting from improved knowledge on bone remodeling, bone disease mechanisms may be understood and new BTE solutions are designed.
Collapse
Affiliation(s)
- Giorgia Borciani
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Giorgia Montalbano
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Nicola Baldini
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60020, Italy
| | - Chiara Vitale-Brovarone
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy.
| | - Gabriela Ciapetti
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
46
|
Safarova Y, Umbayev B, Hortelano G, Askarova S. Mesenchymal stem cells modifications for enhanced bone targeting and bone regeneration. Regen Med 2020; 15:1579-1594. [PMID: 32297546 DOI: 10.2217/rme-2019-0081] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In pathological bone conditions (e.g., osteoporotic fractures or critical size bone defects), increasing the pool of osteoblast progenitor cells is a promising therapeutic approach to facilitate bone healing. Since mesenchymal stem cells (MSCs) give rise to the osteogenic lineage, a number of clinical trials investigated the potential of MSCs transplantation for bone regeneration. However, the engraftment of transplanted cells is often hindered by insufficient oxygen and nutrients supply and the tendency of MSCs to home to different sites of the body. In this review, we discuss various approaches of MSCs transplantation for bone regeneration including scaffold and hydrogel constructs, genetic modifications and surface engineering of the cell membrane aimed to improve homing and increase cell viability, proliferation and differentiation.
Collapse
Affiliation(s)
- Yuliya Safarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,School of Engineering & Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bauyrzhan Umbayev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Gonzalo Hortelano
- School of Sciences & Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Sholpan Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
47
|
Shahabipour F, Oskuee RK, Dehghani H, Shokrgozar MA, Aninwene GE, Bonakdar S. Cell-cell interaction in a coculture system consisting of CRISPR/Cas9 mediated GFP knock-in HUVECs and MG-63 cells in alginate-GelMA based nanocomposites hydrogel as a 3D scaffold. J Biomed Mater Res A 2020; 108:1596-1606. [PMID: 32180319 DOI: 10.1002/jbm.a.36928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
The interaction between osteogenic and angiogenic cells through a coculturing system in biocompatible materials has been considered for successfully engineering vascularized bone tissue equivalents. In this study, we developed a hydrogel-blended scaffold consisted of gelatin methacryloyl (GelMA) and alginate enriched with hydroxyapatite nanoparticles (HAP) to model an in vitro prevascularized bone construct. The hydrogel-based scaffold revealed a higher mechanical stiffness than those of pure (GelMA), alginate, and (GelMA+ HAP) hydrogels. In the present study, we generated a green fluorescent protein (GFP) knock-in umbilical vein endothelial cells (HUVECs) cell line using the CRISPR/Cas9 technology. The GFP was inserted into the human-like ROSA locus of HUVECs genome. HUVECs expressing GFP were cocultured with OB-like cells (MG-63) within three-dimensionally (3D) fabricated hydrogel to investigate the response of cocultured osteoblasts and endothelial cells in a 3D structure. Cell viability under the 3D cocultured gel was higher than the 3D monocultured. Compared to the 3D monocultured condition, the cells were aligned and developed into the vessel-like structures. During 14 days of culture periods, the cells displayed actin protrusions by the formation of spike-like filopodia in the 3D cocultured model. Angiogenic and osteogenic-related genes such as CD31, vWF, and osteocalcin showed higher expression in the cocultured versus the monocultured. These results have collectively indicated that the 3D cocultured hydrogel facilitates interaction among cells, thereby having a greater effect on angiogenic and osteogenic properties in the absence of induction media.
Collapse
Affiliation(s)
| | - Reza K Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Basic Science, Faculty of Veterinary medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - George E Aninwene
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
48
|
Böhrnsen F, Melsheimer P, Natorp M, Rolf H, Schminke B, Kauffmann P, Wolfer S, Schliephake H. Cotransplantation of mesenchymal stromal cells and endothelial cells on calcium carbonate and hydroxylapatite scaffolds in vivo. J Craniomaxillofac Surg 2020; 49:238-245. [PMID: 33483245 DOI: 10.1016/j.jcms.2020.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/06/2020] [Accepted: 03/08/2020] [Indexed: 10/24/2022] Open
Abstract
This study investigated the cotransplantation of bone marrow mesenchymal stromal cells (BMSC) and human umbilical cord endothelial cells (HUVEC), and evaluated their contribution to vascular and bone tissue engineering in vivo. To evaluate the success of osteogenic differentiation and timely vascularization of different osteoconductive scaffolds in vivo, we transferred BMSC and HUVEC pre-cultivated calcium carbonate (CaCO3) and hydroxylapatite (HA) matrices into immunocompromised RNU-rats, and analyzed mineralization, expression of osteopontin, and vascular integration via new vessel formation. After in vivo transplantation, pre-cultivated scaffolds demonstrated overall improved mineralization of 44% for CaCO3 (p = 0.01, SD ± 14.3) and 34% for HA (p = 0.001, SD ± 17.8), as well as improved vascularization of 5.6 vessels/0.1 mm2 on CaCO3 (p < 0.0001, SD ± 2.0) and 5.3 vessels/0.1 mm2 on HA (p < 0.0001, SD ± 2.4) compared with non-pre-cultivated controls. However, no significant differences between the implantation of BMSC-only, HUVEC-only, or BMSC + HUVEC cocultures could be observed. There is an increasing demand for improved bone regeneration in tissue engineering. Cotransplantation of mesenchymal stromal cells and endothelial cells often demonstrates synergistic improvements in vitro. However, the benefits or superiority of cotransplantation was not evident in vivo and so will require further investigation.
Collapse
Affiliation(s)
- Florian Böhrnsen
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany.
| | - Petra Melsheimer
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Mareike Natorp
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Hans Rolf
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Philipp Kauffmann
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Susanne Wolfer
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Henning Schliephake
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| |
Collapse
|
49
|
Kim SHL, Lee SS, Kim I, Kwon J, Kwon S, Bae T, Hur J, Lee H, Hwang NS. Ectopic transient overexpression of OCT-4 facilitates BMP4-induced osteogenic transdifferentiation of human umbilical vein endothelial cells. J Tissue Eng 2020; 11:2041731420909208. [PMID: 32201555 PMCID: PMC7066588 DOI: 10.1177/2041731420909208] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/07/2020] [Indexed: 01/05/2023] Open
Abstract
Limitation in cell sources for autologous cell therapy has been a recent focus in stem cell therapy and tissue engineering. Among various research advances, direct conversion, or transdifferentiation, is a notable and feasible strategy for the generation and acquirement of wanted cell source. So far, utilizing cell transdifferentiation technology in tissue engineering was mainly restricted at achieving single wanted cell type from diverse cell types with high efficiency. However, regeneration of a complete tissue always requires multiple cell types which poses an intrinsic complexity. In this study, enhanced osteogenic differentiation was achieved by transient ectopic expression of octamer-binding transcription factor 4 (OCT-4) gene followed by bone morphogenetic protein 4 treatment on human umbilical vein endothelial cells. OCT-4 transfection and bone morphogenetic protein 4 treatment resulted in enhanced expression of osteogenic markers such as core-binding factor alpha 1, alkaline phosphatase, and collagen 1 compared with bone morphogenetic protein 4 treatment alone. Furthermore, we employed gelatin-heparin cryogel in cranial defect model for in vivo bone formation. Micro-computed tomography and histological analysis of in vivo samples showed that OCT-4 transfection followed by bone morphogenetic protein 4 treatment resulted in efficient transdifferentiation of endothelial cells to osteogenic cells. These results suggest that the combination of OCT-4 and bone morphogenetic protein 4 on endothelial cells would be a reliable multicellular transdifferentiation model which could be applied for bone tissue engineering.
Collapse
Affiliation(s)
- Seung Hyun L Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Seunghun S Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Inseon Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea
| | - Janet Kwon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Song Kwon
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea
| | - Taegeun Bae
- BioMAX/N-Bio Institute, Seoul National University, Seoul, Republic of Korea
| | - Junho Hur
- Department of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hwajin Lee
- School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea.,School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea.,BioMAX/N-Bio Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
50
|
Zheng JS, Ruan HR, Shuang-Qiu, Jing-Nie, Hou KW, Rui-Wu. Therapeutic Effects of Revascularisation on the Healing of Free Bone Grafts in Dogs. J Vet Res 2020; 64:175-180. [PMID: 32258815 PMCID: PMC7105986 DOI: 10.2478/jvetres-2020-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 03/02/2020] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION The therapeutic effect of subcutaneous embedding and revascularisation on the repair of canine bone defects caused by open fracture was examined. MATERIAL AND METHODS A total of 12 adult beagle dogs were randomly split into a control group (group C) and a test group (group T). A section of the radius was removed from each dog under general anaesthesia and the deficit supported by an orthopaedic implant. Group T had the section surgically implanted next to the blood vessel-rich saphenous vein and Group C had it cryopreserved at -80°C. After eight weeks, the bone was surgically implanted back into the matching radial deficit. Bone healing was evaluated by gross morphological and X-ray examinations, post-mortem histology, and successive blood measurements of key bone biochemical markers. RESULTS At 12 weeks, the bone healing boundary was disappearing more quickly in group T dogs than in their group C counterparts. X-ray and histological examinations showed that the cortical repair of group T subjects was complete and the bony plate arrangement was more regular than that in group C. The levels of bone biochemical markers also proved that the healing state of group T was better. CONCLUSION The results showed that the degree of healing, osteoclast activity, and bone formation status of group T were better than those of group C, proving that the vascularised bone graft had a significantly shorter healing time than the cryopreserved bone graft.
Collapse
Affiliation(s)
- Jia-San Zheng
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University Daqing High-tech Industrial Development Zone, Daqing, 163319, People’s Republic of China
| | - Hong-Ri Ruan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University Daqing High-tech Industrial Development Zone, Daqing, 163319, People’s Republic of China
| | - Shuang-Qiu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University Daqing High-tech Industrial Development Zone, Daqing, 163319, People’s Republic of China
| | - Jing-Nie
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University Daqing High-tech Industrial Development Zone, Daqing, 163319, People’s Republic of China
| | - Kai-Wen Hou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University Daqing High-tech Industrial Development Zone, Daqing, 163319, People’s Republic of China
| | - Rui-Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University Daqing High-tech Industrial Development Zone, Daqing, 163319, People’s Republic of China
| |
Collapse
|