1
|
El Hajj S, Ntaté MB, Breton C, Siadous R, Aid R, Dupuy M, Letourneur D, Amédée J, Duval H, David B. Bone Spheroid Development Under Flow Conditions with Mesenchymal Stem Cells and Human Umbilical Vein Endothelial Cells in a 3D Porous Hydrogel Supplemented with Hydroxyapatite. Gels 2024; 10:666. [PMID: 39451319 PMCID: PMC11506954 DOI: 10.3390/gels10100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Understanding the niche interactions between blood and bone through the in vitro co-culture of osteo-competent cells and endothelial cells is a key factor in unraveling therapeutic potentials in bone regeneration. This can be additionally supported by employing numerical simulation techniques to assess local physical factors, such as oxygen concentration, and mechanical stimuli, such as shear stress, that can mediate cellular communication. In this study, we developed a Mesenchymal Stem Cell line (MSC) and a Human Umbilical Vein Endothelial Cell line (HUVEC), which were co-cultured under flow conditions in a three-dimensional, porous, natural pullulan/dextran scaffold that was supplemented with hydroxyapatite crystals that allowed for the spontaneous formation of spheroids. After 2 weeks, their viability was higher under the dynamic conditions (>94%) than the static conditions (<75%), with dead cells central in the spheroids. Mineralization and collagen IV production increased under the dynamic conditions, correlating with osteogenesis and vasculogenesis. The endothelial cells clustered at the spheroidal core by day 7. Proliferation doubled in the dynamic conditions, especially at the scaffold peripheries. Lattice Boltzmann simulations showed negligible wall shear stress in the hydrogel pores but highlighted highly oxygenated zones coinciding with cell proliferation. A strong oxygen gradient likely influenced endothelial migration and cell distribution. Hypoxia was minimal, explaining high viability and spheroid maturation in the dynamic conditions.
Collapse
Affiliation(s)
- Soukaina El Hajj
- Laboratoire de Mécanique Paris-Saclay, CNRS, CentraleSupélec, ENS Paris-Saclay, Université Paris-Saclay, 91190 Gif-sur-Yvette, France;
| | - Martial Bankoué Ntaté
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Cyril Breton
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Robin Siadous
- Laboratoire de Bioingénierie Tissulaire (BioTis), INSERM U1026, Université de Bordeaux, 33076 Bordeaux, France; (R.S.); (J.A.)
| | - Rachida Aid
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Paris Cité, 75018 Paris, France; (R.A.); (D.L.)
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Magali Dupuy
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Didier Letourneur
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Paris Cité, 75018 Paris, France; (R.A.); (D.L.)
- Laboratoire de Recherche Vasculaire Translationnelle (LVTS), INSERM U1148, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Joëlle Amédée
- Laboratoire de Bioingénierie Tissulaire (BioTis), INSERM U1026, Université de Bordeaux, 33076 Bordeaux, France; (R.S.); (J.A.)
| | - Hervé Duval
- Laboratoire de Génie des Procédés et Matériaux, CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (M.B.N.); (C.B.); (M.D.); (H.D.)
| | - Bertrand David
- Laboratoire de Mécanique Paris-Saclay, CNRS, CentraleSupélec, ENS Paris-Saclay, Université Paris-Saclay, 91190 Gif-sur-Yvette, France;
| |
Collapse
|
2
|
Augustine R, Gezek M, Nikolopoulos VK, Buck PL, Bostanci NS, Camci-Unal G. Stem Cells in Bone Tissue Engineering: Progress, Promises and Challenges. Stem Cell Rev Rep 2024; 20:1692-1731. [PMID: 39028416 DOI: 10.1007/s12015-024-10738-y] [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] [Accepted: 05/17/2024] [Indexed: 07/20/2024]
Abstract
Bone defects from accidents, congenital conditions, and age-related diseases significantly impact quality of life. Recent advancements in bone tissue engineering (TE) involve biomaterial scaffolds, patient-derived cells, and bioactive agents, enabling functional bone regeneration. Stem cells, obtained from numerous sources including umbilical cord blood, adipose tissue, bone marrow, and dental pulp, hold immense potential in bone TE. Induced pluripotent stem cells and genetically modified stem cells can also be used. Proper manipulation of physical, chemical, and biological stimulation is crucial for their proliferation, maintenance, and differentiation. Stem cells contribute to osteogenesis, osteoinduction, angiogenesis, and mineralization, essential for bone regeneration. This review provides an overview of the latest developments in stem cell-based TE for repairing and regenerating defective bones.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Radiology, Stanford Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | | | - Paige Lauren Buck
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
3
|
Jeon O, Park H, Leach JK, Alsberg E. Biofabrication of engineered tissues by 3D bioprinting of tissue specific high cell-density bioinks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612457. [PMID: 39314396 PMCID: PMC11419076 DOI: 10.1101/2024.09.11.612457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Bioprinting of high cell-density bioinks is a promising technique for cellular condensation-based tissue engineering and regeneration medicine. However, it remains difficult to create precisely controlled complex structures and organization of tissues with high cell-density bioink-based bioprinting for tissue specific condensation. In this study, we present newly biofabricated tissues from directly assembled, tissue specific, high cell-density bioinks which have been three-dimensionally printed into a photocrosslinkable and biodegradable hydrogel microparticle supporting bath. Three types of tissue specific high cell-density bioinks have been prepared with individual stem cells or stem cell aggregates by incorporation of growth factor-loaded gelatin microparticles. The bioprinted tissue specific high cell-density bioinks in the photocrosslinked microgel supporting bath condense together and differentiate down tissue-specific lineages to form multi-phase tissues (e.g., osteochondral tissues). By changing the growth factors and cell types, these tissue specific high cell-density bioinks enable engineering of various functional tissues with controlled architecture and organization of cells.
Collapse
|
4
|
Camal Ruggieri IN, Aimone M, Juanes-Gusano D, Ibáñez-Fonseca A, Santiago O, Stur M, Mardegan Issa JP, Missana LR, Alonso M, Rodríguez-Cabello JC, Feldman S. Biocompatibility and bone regeneration with elastin-like recombinamer-based catalyst-free click gels. Sci Rep 2024; 14:20223. [PMID: 39215050 PMCID: PMC11364658 DOI: 10.1038/s41598-024-69658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Large bone defects are a significant health problem today with various origins, including extensive trauma, tumours, or congenital musculoskeletal disorders. Tissue engineering, and in particular bone tissue engineering, aims to respond to this demand. As such, we propose a specific model based on Elastin-Like Recombinamers-based click-chemistry hydrogels given their high biocompatibility and their potent on bone regeneration effect conferred by different bioactive sequences. In this work we demonstrate, using biochemistry, histology, histomorphometry and imaging techniques, the biocompatibility of our matrix and its potent effect on bone regeneration in a model of bone parietal lesion in female New Zealand rabbits.
Collapse
Affiliation(s)
- I N Camal Ruggieri
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina.
| | - M Aimone
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | - D Juanes-Gusano
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
| | - A Ibáñez-Fonseca
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
| | - O Santiago
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | - M Stur
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
- Diagnostic Imaging and Radiology, School of Medicine, National Rosario University, Rosario, Argentina
| | - J P Mardegan Issa
- Ribeirão Preto School of Dentistry, São Paulo University, São Paulo, Brazil
| | - L R Missana
- Experimental Pathology and Tissue Engineering Laboratory, School of Dentistry, National Tucumán University, Tucumán, Argentina
- Tissues Laboratory, IMMCA-CONICET, Tucumán, Argentina
| | - M Alonso
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain.
| | | | - S Feldman
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina.
- Research Council of the National Rosario University (CIUNR) and CONICET, Rosario, Argentina.
| |
Collapse
|
5
|
Mirsky NA, Ehlen QT, Greenfield JA, Antonietti M, Slavin BV, Nayak VV, Pelaez D, Tse DT, Witek L, Daunert S, Coelho PG. Three-Dimensional Bioprinting: A Comprehensive Review for Applications in Tissue Engineering and Regenerative Medicine. Bioengineering (Basel) 2024; 11:777. [PMID: 39199735 PMCID: PMC11351251 DOI: 10.3390/bioengineering11080777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Since three-dimensional (3D) bioprinting has emerged, it has continuously to evolved as a revolutionary technology in surgery, offering new paradigms for reconstructive and regenerative medical applications. This review highlights the integration of 3D printing, specifically bioprinting, across several surgical disciplines over the last five years. The methods employed encompass a review of recent literature focusing on innovations and applications of 3D-bioprinted tissues and/or organs. The findings reveal significant advances in the creation of complex, customized, multi-tissue constructs that mimic natural tissue characteristics, which are crucial for surgical interventions and patient-specific treatments. Despite the technological advances, the paper introduces and discusses several challenges that remain, such as the vascularization of bioprinted tissues, integration with the host tissue, and the long-term viability of bioprinted organs. The review concludes that while 3D bioprinting holds substantial promise for transforming surgical practices and enhancing patient outcomes, ongoing research, development, and a clear regulatory framework are essential to fully realize potential future clinical applications.
Collapse
Affiliation(s)
| | - Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | - Blaire V. Slavin
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Pelaez
- Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David T. Tse
- Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Paulo G. Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
6
|
Zhang J, Suttapreyasri S, Leethanakul C, Samruajbenjakun B. Fabrication of vascularized tissue-engineered bone models using triaxial bioprinting. J Biomed Mater Res A 2024; 112:1093-1106. [PMID: 38411369 DOI: 10.1002/jbm.a.37694] [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: 09/15/2023] [Revised: 01/01/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
Bone tissue is a highly vascularized tissue. When constructing tissue-engineered bone models, both the osteogenic and angiogenic capabilities of the construct should be carefully considered. However, fabricating a vascularized tissue-engineered bone to promote vascular formation and bone generation, while simultaneously establishing nutrition channels to facilitate nutrient exchange within the constructs, remains a significant challenge. Triaxial bioprinting, which not only allows the independent encapsulation of different cell types while simultaneously forming nutrient channels, could potentially emerge as a strategy for fabricating vascularized tissue-engineered bone. Moreover, bioinks should also be applied in combination to promote both osteogenesis and angiogenesis. In this study, employing triaxial bioprinting, we used a blend bioink of gelatin methacryloyl (GelMA), sodium alginate (Alg), and different concentrations of nano beta-tricalcium phosphate (nano β-TCP) encapsulated MC3T3-E1 preosteoblasts as the outer layer, a mixed bioink of GelMA and Alg loaded with human umbilical vein endothelial cells (HUVEC) as the middle layer, and gelatin as a sacrificial material to form nutrient channels in the inner layer to fabricate vascularized bone constructs simulating the microenvironment for bone and vascular tissues. The results showed that the addition of nano β-TCP could adjust the mechanical, swelling, and degradation properties of the constructs. Biological assessments revealed the cell viability of constructs containing different concentrations of nano β-TCP was higher than 90% on day 7, The cell-laden constructs containing 3% (w/v) nano β-TCP exhibited better osteogenic (higher Alkaline phosphatase activity and larger Osteocalcin positive area) and angiogenic (the gradual increased CD31 positive area) potential. Therefore, using triaxial bioprinting technology and employing GelMA, Alg, and nano β-TCP as bioink components could fabricate vascularized bone tissue constructs, offering a novel strategy for vascularized bone tissue engineering.
Collapse
Affiliation(s)
- Junbiao Zhang
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
- Guiyang Hospital of Stomatology, Guiyang, People's Republic of China
| | - Srisurang Suttapreyasri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| | - Bancha Samruajbenjakun
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
7
|
Nwokoye PN, Abilez OJ. Blood vessels in a dish: the evolution, challenges, and potential of vascularized tissues and organoids. Front Cardiovasc Med 2024; 11:1336910. [PMID: 38938652 PMCID: PMC11210405 DOI: 10.3389/fcvm.2024.1336910] [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: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/29/2024] Open
Abstract
Vascular pathologies are prevalent in a broad spectrum of diseases, necessitating a deeper understanding of vascular biology, particularly in overcoming the oxygen and nutrient diffusion limit in tissue constructs. The evolution of vascularized tissues signifies a convergence of multiple scientific disciplines, encompassing the differentiation of human pluripotent stem cells (hPSCs) into vascular cells, the development of advanced three-dimensional (3D) bioprinting techniques, and the refinement of bioinks. These technologies are instrumental in creating intricate vascular networks essential for tissue viability, especially in thick, complex constructs. This review provides broad perspectives on the past, current state, and advancements in key areas, including the differentiation of hPSCs into specific vascular lineages, the potential and challenges of 3D bioprinting methods, and the role of innovative bioinks mimicking the native extracellular matrix. We also explore the integration of biophysical cues in vascularized tissues in vitro, highlighting their importance in stimulating vessel maturation and functionality. In this review, we aim to synthesize these diverse yet interconnected domains, offering a broad, multidisciplinary perspective on tissue vascularization. Advancements in this field will help address the global organ shortage and transform patient care.
Collapse
Affiliation(s)
- Peter N. Nwokoye
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Oscar J. Abilez
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
- Division of Pediatric CT Surgery, Stanford University, Stanford, CA, United States
- Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, United States
- Bio-X Program, Stanford University, Stanford, CA, United States
| |
Collapse
|
8
|
Das S, Valoor R, Ratnayake P, Basu B. Low-Concentration Gelatin Methacryloyl Hydrogel with Tunable 3D Extrusion Printability and Cytocompatibility: Exploring Quantitative Process Science and Biophysical Properties. ACS APPLIED BIO MATERIALS 2024; 7:2809-2835. [PMID: 38602318 DOI: 10.1021/acsabm.3c01194] [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: 04/12/2024]
Abstract
Three-dimensional (3D) bioprinting of hydrogels with a wide spectrum of compositions has been widely investigated. Despite such efforts, a comprehensive understanding of the correlation among the process science, buildability, and biophysical properties of the hydrogels for a targeted clinical application has not been developed in the scientific community. In particular, the quantitative analysis across the entire developmental path for 3D extrusion bioprinting of such scaffolds is not widely reported. In the present work, we addressed this gap by using widely investigated biomaterials, such as gelatin methacryloyl (GelMA), as a model system. Using extensive experiments and quantitative analysis, we analyzed how the individual components of methacrylated carboxymethyl cellulose (mCMC), needle-shaped nanohydroxyapatite (nHAp), and poly(ethylene glycol)diacrylate (PEGDA) with GelMA as baseline matrix of the multifunctional bioink can influence the biophysical properties, printability, and cellular functionality. The complex interplay among the biomaterial ink formulations, viscoelastic properties, and printability toward the large structure buildability (structurally stable cube scaffolds with 15 mm edge) has been explored. Intriguingly, the incorporation of PEGDA into the GelMA/mCMC matrix offered improved compressive modulus (∼40-fold), reduced swelling ratio (∼2-fold), and degradation rates (∼30-fold) compared to pristine GelMA. The correlation among microstructural pore architecture, biophysical properties, and cytocompatibility is also established for the biomaterial inks. These photopolymerizable bio(material)inks served as the platform for the growth and development of bone and cartilage matrix when human mesenchymal stem cells (hMSCs) are either seeded on two-dimensional (2D) substrates or encapsulated on 3D scaffolds. Taken together, this present study unequivocally establishes a significant step forward in the development of a broad spectrum of shape-fidelity compliant bioink for the 3D bioprinting of multifunctional scaffolds and emphasizes the need for invoking more quantitative analysis in establishing process-microstructure-property correlation.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Remya Valoor
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Praneeth Ratnayake
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
9
|
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
|
10
|
Das S, Jegadeesan JT, Basu B. Gelatin Methacryloyl (GelMA)-Based Biomaterial Inks: Process Science for 3D/4D Printing and Current Status. Biomacromolecules 2024; 25:2156-2221. [PMID: 38507816 DOI: 10.1021/acs.biomac.3c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Tissue engineering for injured tissue replacement and regeneration has been a subject of investigation over the last 30 years, and there has been considerable interest in using additive manufacturing to achieve these goals. Despite such efforts, many key questions remain unanswered, particularly in the area of biomaterial selection for these applications as well as quantitative understanding of the process science. The strategic utilization of biological macromolecules provides a versatile approach to meet diverse requirements in 3D printing, such as printability, buildability, and biocompatibility. These molecules play a pivotal role in both physical and chemical cross-linking processes throughout the biofabrication, contributing significantly to the overall success of the 3D printing process. Among the several bioprintable materials, gelatin methacryloyl (GelMA) has been widely utilized for diverse tissue engineering applications, with some degree of success. In this context, this review will discuss the key bioengineering approaches to identify the gelation and cross-linking strategies that are appropriate to control the rheology, printability, and buildability of biomaterial inks. This review will focus on the GelMA as the structural (scaffold) biomaterial for different tissues and as a potential carrier vehicle for the transport of living cells as well as their maintenance and viability in the physiological system. Recognizing the importance of printability toward shape fidelity and biophysical properties, a major focus in this review has been to discuss the qualitative and quantitative impact of the key factors, including microrheological, viscoelastic, gelation, shear thinning properties of biomaterial inks, and printing parameters, in particular, reference to 3D extrusion printing of GelMA-based biomaterial inks. Specifically, we emphasize the different possibilities to regulate mechanical, swelling, biodegradation, and cellular functionalities of GelMA-based bio(material) inks, by hybridization techniques, including different synthetic and natural biopolymers, inorganic nanofillers, and microcarriers. At the close, the potential possibility of the integration of experimental data sets and artificial intelligence/machine learning approaches is emphasized to predict the printability, shape fidelity, or biophysical properties of GelMA bio(material) inks for clinically relevant tissues.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| | | | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| |
Collapse
|
11
|
Mamidi N, Ijadi F, Norahan MH. Leveraging the Recent Advancements in GelMA Scaffolds for Bone Tissue Engineering: An Assessment of Challenges and Opportunities. Biomacromolecules 2024; 25:2075-2113. [PMID: 37406611 DOI: 10.1021/acs.biomac.3c00279] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
The field of bone tissue engineering has seen significant advancements in recent years. Each year, over two million bone transplants are performed globally, and conventional treatments, such as bone grafts and metallic implants, have their limitations. Tissue engineering offers a new level of treatment, allowing for the creation of living tissue within a biomaterial framework. Recent advances in biomaterials have provided innovative approaches to rebuilding bone tissue function after damage. Among them, gelatin methacryloyl (GelMA) hydrogel is emerging as a promising biomaterial for supporting cell proliferation and tissue regeneration, and GelMA has exhibited exceptional physicochemical and biological properties, making it a viable option for clinical translation. Various methods and classes of additives have been used in the application of GelMA for bone regeneration, with the incorporation of nanofillers or other polymers enhancing its resilience and functional performance. Despite promising results, the fabrication of complex structures that mimic the bone architecture and the provision of balanced physical properties for both cell and vasculature growth and proper stiffness for load bearing remain as challenges. In terms of utilizing osteogenic additives, the priority should be on versatile components that promote angiogenesis and osteogenesis while reinforcing the structure for bone tissue engineering applications. This review focuses on recent efforts and advantages of GelMA-based composite biomaterials for bone tissue engineering, covering the literature from the last five years.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Fatemeh Ijadi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| |
Collapse
|
12
|
Hassanpour P, Sadeghsoltani F, Haiaty S, Zakeri Z, Saghebasl S, Izadpanah M, Boroumand S, Mota A, Rahmati M, Rahbarghazi R, Talebi M, Rabbani S, Tafti SHA. Mitochondria-loaded alginate-based hydrogel accelerated angiogenesis in a rat model of acute myocardial infarction. Int J Biol Macromol 2024; 260:129633. [PMID: 38253146 DOI: 10.1016/j.ijbiomac.2024.129633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/13/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024]
Abstract
Here, mitochondria were isolated from mesenchymal stem cells (MSCs) after being treated with mitochondria-stimulating substrates, 50 μM metformin (Met), and 40 μM dichloroacetic acid (DCA). The isolated mitochondria (2 × 107 particles) were characterized and encapsulated inside 100 μl hydrogel composed of alginate (3 % w/v; Alg)/gelatin (Gel; 1 % w/v) enriched with 1 μM pyrrole (Pyr) solidified in the presence of 0.2 M FeCl3. The physicochemical properties and cytocompatibility of prepared hydrogels were assessed using FTIR, swelling, biodegradation, porosity assays, and scanning electron microscopy (SEM). The mitochondria-bearing hydrogel was injected into the ischemic area of rat hearts. FTIR absorption bands represented that the addition of FeCl3 led to polypyrrole (PPy) formation, polysaccharide oxidation, and interaction between Alg and Gel. SEM images exhibited porous structure and the size of pores was reduced in Alg/Gel + PPy group compared to Alg + PPy hydrogel. Based on the data, both Alg + PPy and Alg/Gel + PPy hydrogels can preserve the integrity and morphology of loaded mitochondria. It was noted that Alg/Gel + PPy hydrogel possessed a higher swelling ratio, degradation, and porosity compared to Alg + PPy group. Data confirmed that Alg/Gel + PPy hydrogel containing 1 μM Pyr yielded the highest survival rate compared to groups with 2 and 4 μM Pyr (p < 0.05). Injection of mitochondria-loaded Alg/Gel + PPy hydrogel yielded significant restoration of left ventricle thickness compared to the infarction, mitochondria, and Alg/Gel + PPy hydrogel groups 14 days post-injection (p < 0.05). Histological analyses revealed a significant increase of vWF+ capillaries and α-SMA+ arterioles in the mitochondria-loaded Alg/Gel + PPy hydrogel group (p < 0.05). Immunofluorescence imaging revealed the ability of rat cardiomyocytes to uptake mitochondria alone or after being loaded into Alg/Gel + PPy hydrogel. These effects were evident in the Alg/Gel + PPy group. Taken together, electroconductive Alg-based hydrogels are suitable platforms for the transplantation of cells and organelles and the regeneration of ischemic heart changes.
Collapse
Affiliation(s)
- Parisa Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ziba Zakeri
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Melika Izadpanah
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safieh Boroumand
- Research Center for Advanced Technologies In Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Mota
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Rahmati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Talebi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies In Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies In Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Wang R, Zha X, Chen J, Fu R, Fu Y, Xiang J, Yang W, Zhao L. Hierarchical Composite Scaffold with Deferoxamine Delivery System to Promote Bone Regeneration via Optimizing Angiogenesis. Adv Healthc Mater 2024:e2304232. [PMID: 38375993 DOI: 10.1002/adhm.202304232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/18/2024] [Indexed: 02/21/2024]
Abstract
A bone defect refers to the loss of bone tissue caused by trauma or lesion. Bone defects result in high morbidity and deformity rates worldwide. Autologous bone grafting has been widely applied in clinics as the gold standard of treatment; however, it has limitations. Hence, bone tissue engineering has been proposed and developed as a novel therapeutic strategy for treating bone defects. Rapid and effective vascularization is essential for bone regeneration. In this study, a hierarchical composite scaffold with deferoxamine (DFO) delivery system, DFO@GMs-pDA/PCL-HNTs (DGPN), is developed, focusing on vascularized bone regeneration. The hierarchical structure of DGPN imitates the microstructure of natural bone and interacts with the local extracellular matrix, facilitating cell adhesion and proliferation. The addition of 1 wt% of halloysite nanotubes (HNTs) improves the material properties. Hydrophilic and functional groups conferred by polydopamine (pDA) modifications strengthen the scaffold bioactivity. Gelatin microspheres (GMs) protect the pharmacological activity of DFO, achieving local application and sustained release for 7 days. DFO effectively promotes angiogenesis by activating the signaling pathway of hypoxia inducible factor-1 α. In addition, DFO synergizes with HNTs to promote osteogenic differentiation and matrix mineralization. These results indicate that DGPN promotes bone regeneration and accelerates cranial defect healing.
Collapse
Affiliation(s)
- Raokaijuan Wang
- West China School of Stomatology Sichuan University, State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| | - Xiangjun Zha
- Liver Transplant Center and Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jouchen Chen
- West China School of Stomatology Sichuan University, State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| | - Ruijie Fu
- West China School of Stomatology Sichuan University, State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| | - Yajun Fu
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Jie Xiang
- West China School of Stomatology Sichuan University, State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| | - Wei Yang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Lixing Zhao
- Department of Orthodontics, West China School of Stomatology Sichuan University, State Key Laboratory of Oral Diseases/National Clinical Research Center for Oral Diseases, Chengdu, 610041, China
| |
Collapse
|
14
|
Tolmacheva N, Bhattacharyya A, Noh I. Calcium Phosphate Biomaterials for 3D Bioprinting in Bone Tissue Engineering. Biomimetics (Basel) 2024; 9:95. [PMID: 38392140 PMCID: PMC10886915 DOI: 10.3390/biomimetics9020095] [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: 12/23/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Three-dimensional bioprinting is a promising technology for bone tissue engineering. However, most hydrogel bioinks lack the mechanical and post-printing fidelity properties suitable for such hard tissue regeneration. To overcome these weak properties, calcium phosphates can be employed in a bioink to compensate for the lack of certain characteristics. Further, the extracellular matrix of natural bone contains this mineral, resulting in its structural robustness. Thus, calcium phosphates are necessary components of bioink for bone tissue engineering. This review paper examines different recently explored calcium phosphates, as a component of potential bioinks, for the biological, mechanical and structural properties required of 3D bioprinted scaffolds, exploring their distinctive properties that render them favorable biomaterials for bone tissue engineering. The discussion encompasses recent applications and adaptations of 3D-printed scaffolds built with calcium phosphates, delving into the scientific reasons behind the prevalence of certain types of calcium phosphates over others. Additionally, this paper elucidates their interactions with polymer hydrogels for 3D bioprinting applications. Overall, the current status of calcium phosphate/hydrogel bioinks for 3D bioprinting in bone tissue engineering has been investigated.
Collapse
Affiliation(s)
- Nelli Tolmacheva
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Amitava Bhattacharyya
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
- Medical Electronics Research Center, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Insup Noh
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
15
|
Mommsen P, März V, Krezdorn N, Aktas G, Sehmisch S, Vogt PM, Großner T, Omar Pacha T. Reconstruction of an Extensive Segmental Radial Shaft Bone Defect by Vascularized 3D-Printed Graft Cage. J Pers Med 2024; 14:178. [PMID: 38392611 PMCID: PMC10890561 DOI: 10.3390/jpm14020178] [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: 01/08/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
We report here a 46-year-old male patient with a 14 cm segmental bone defect of the radial shaft after third degree open infected fracture caused by a shrapnel injury. The patient underwent fixed-angle plate osteosynthesis and bone reconstruction of the radial shaft by a vascularized 3D-printed graft cage, including plastic coverage with a latissimus dorsi flap and an additional central vascular pedicle. Bony reconstruction of segmental defects still represents a major challenge in musculo-skeletal surgery. Thereby, 3D-printed scaffolds or graft cages display a new treatment option for bone restoration. As missing vascularization sets the limits for the treatment of large-volume bone defects by 3D-printed scaffolds, in the present case, we firstly describe the reconstruction of an extensive radial shaft bone defect by using a graft cage with additional vascularization.
Collapse
Affiliation(s)
- Philipp Mommsen
- Department of Trauma Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Vincent März
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Nicco Krezdorn
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, 30625 Hannover, Germany
- Department of Plastic and Breast Surgery, Roskilde University Hospital, 4000 Roskilde, Denmark
| | - Gökmen Aktas
- Department of Trauma Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Stephan Sehmisch
- Department of Trauma Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Peter Maria Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Großner
- BellaSeno GmbH, 04103 Leipzig, Germany
- BellaSeno Pty Ltd., Brisbane, QLD 4220, Australia
| | - Tarek Omar Pacha
- Department of Trauma Surgery, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
16
|
Lv N, Zhou Z, Hou M, Hong L, Li H, Qian Z, Gao X, Liu M. Research progress of vascularization strategies of tissue-engineered bone. Front Bioeng Biotechnol 2024; 11:1291969. [PMID: 38312513 PMCID: PMC10834685 DOI: 10.3389/fbioe.2023.1291969] [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: 09/10/2023] [Accepted: 12/06/2023] [Indexed: 02/06/2024] Open
Abstract
The bone defect caused by fracture, bone tumor, infection, and other causes is not only a problematic point in clinical treatment but also one of the hot issues in current research. The development of bone tissue engineering provides a new way to repair bone defects. Many animal experimental and rising clinical application studies have shown their excellent application prospects. The construction of rapid vascularization of tissue-engineered bone is the main bottleneck and critical factor in repairing bone defects. The rapid establishment of vascular networks early after biomaterial implantation can provide sufficient nutrients and transport metabolites. If the slow formation of the local vascular network results in a lack of blood supply, the osteogenesis process will be delayed or even unable to form new bone. The researchers modified the scaffold material by changing the physical and chemical properties of the scaffold material, loading the growth factor sustained release system, and combining it with trace elements so that it can promote early angiogenesis in the process of induced bone regeneration, which is beneficial to the whole process of bone regeneration. This article reviews the local vascular microenvironment in the process of bone defect repair and the current methods of improving scaffold materials and promoting vascularization.
Collapse
Affiliation(s)
- Nanning Lv
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Zhangzhe Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mingzhuang Hou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lihui Hong
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Hongye Li
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Zhonglai Qian
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuzhu Gao
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| | - Mingming Liu
- Department of Orthopedic Surgery, The Second People’s Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
- Department of Orthopedic Surgery, The Affiliated Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu, China
| |
Collapse
|
17
|
Su X, Si X, Liu Y, Xiong N, Li S, Tang L, Shi Z, Cheng L, Zhang F. Comparison of different hydroxyapatite composites for bone tissue repair: In vitro and in vivo analyses. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1155-1161. [PMID: 39055877 PMCID: PMC11266744 DOI: 10.22038/ijbms.2024.78578.16995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 04/13/2024] [Indexed: 07/28/2024]
Abstract
Objectives The material used for bone tissue repair needs to be simultaneously osteoconductive, osteoinductive, and osteogenic. To overcome this problem, researchers combine hydroxyapatite (HA) with natural materials to improve properties. This paper compares the effects of angiogenesis and osteogenesis with different composites through in vivo experiments and characterization analysis. Materials and Methods Chitosan/nHA (CS/nHA) and sodium alginate/nHA (SA/nHA) microspheres were synthesized via reverse-phase emulsification crosslinking and analyzed using scanning electron microscopy (SEM), energy dispersion spectroscopy (EDS), and X-ray diffraction (XRD). Implanted into mouse thigh muscles, their angiogenic and osteogenic potentials were assessed after 8 and 12 weeks through various staining methods and immunohistochemistry. Results The mean vascular density (MVD) of CS/nHA, CaP/nHA, and SA/nHA groups was (134.92±35.30) n/mm2, (159.09±22.14) n/mm2, (160.31±42.23) n/mm2 at 12 weeks, respectively. The MVD of the CaP/nHA and SA/nHA groups were significantly higher than that of the CS/nHA group. The collagen volume fractions (CVF) were 34.13%, 51.53%, and 54.96% in the CS/nHA, CaP/nHA, and SA/nHA groups, respectively. In addition, the positive expression area ratios of OPN and CD31 in the CaP/nHA and SA/nHA groups were also significantly higher than those in the CS/nHA group. Conclusion The ability of SA/nHA composite microspheres in osteogenesis and angiogenesis is clearly superior to that of the CS/nHA group and is comparable to that of CaP/nHA, which has superior osteogenesis ability, indicating that SA/nHA composite microspheres have greater application prospects in bone tissue engineering.
Collapse
Affiliation(s)
- Xiaoyu Su
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
- These authors contributed eqully to this work
| | - Xiang Si
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
- These authors contributed eqully to this work
| | - Yuyang Liu
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| | - Nana Xiong
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| | - Siyuan Li
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| | - Lu Tang
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| | - Zheng Shi
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| | - Lijia Cheng
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| | - Fei Zhang
- 1 School of Basic Medical Sciences, Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
18
|
Jiang H, Li X, Chen T, Liu Y, Wang Q, Wang Z, Jia J. Bioprinted vascular tissue: Assessing functions from cellular, tissue to organ levels. Mater Today Bio 2023; 23:100846. [PMID: 37953757 PMCID: PMC10632537 DOI: 10.1016/j.mtbio.2023.100846] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
3D bioprinting technology is widely used to fabricate various tissue structures. However, the absence of vessels hampers the ability of bioprinted tissues to receive oxygen and nutrients as well as to remove wastes, leading to a significant reduction in their survival rate. Despite the advancements in bioinks and bioprinting technologies, bioprinted vascular structures continue to be unsuitable for transplantation compared to natural blood vessels. In addition, a complete assessment index system for evaluating the structure and function of bioprinted vessels in vitro has not yet been established. Therefore, in this review, we firstly highlight the significance of selecting suitable bioinks and bioprinting techniques as they two synergize with each other. Subsequently, focusing on both vascular-associated cells and vascular tissues, we provide a relatively thorough assessment of the functions of bioprinted vascular tissue based on the physiological functions that natural blood vessels possess. We end with a review of the applications of vascular models, such as vessel-on-a-chip, in simulating pathological processes and conducting drug screening at the organ level. We believe that the development of fully functional blood vessels will soon make great contributions to tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Haihong Jiang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xueyi Li
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Tianhong Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yang Liu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhimin Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Jia Jia
- School of Life Sciences, Shanghai University, Shanghai, China
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| |
Collapse
|
19
|
Patrocinio D, Galván-Chacón V, Gómez-Blanco JC, Miguel SP, Loureiro J, Ribeiro MP, Coutinho P, Pagador JB, Sanchez-Margallo FM. Biopolymers for Tissue Engineering: Crosslinking, Printing Techniques, and Applications. Gels 2023; 9:890. [PMID: 37998980 PMCID: PMC10670821 DOI: 10.3390/gels9110890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Currently, tissue engineering has been dedicated to the development of 3D structures through bioprinting techniques that aim to obtain personalized, dynamic, and complex hydrogel 3D structures. Among the different materials used for the fabrication of such structures, proteins and polysaccharides are the main biological compounds (biopolymers) selected for the bioink formulation. These biomaterials obtained from natural sources are commonly compatible with tissues and cells (biocompatibility), friendly with biological digestion processes (biodegradability), and provide specific macromolecular structural and mechanical properties (biomimicry). However, the rheological behaviors of these natural-based bioinks constitute the main challenge of the cell-laden printing process (bioprinting). For this reason, bioprinting usually requires chemical modifications and/or inter-macromolecular crosslinking. In this sense, a comprehensive analysis describing these biopolymers (natural proteins and polysaccharides)-based bioinks, their modifications, and their stimuli-responsive nature is performed. This manuscript is organized into three sections: (1) tissue engineering application, (2) crosslinking, and (3) bioprinting techniques, analyzing the current challenges and strengths of biopolymers in bioprinting. In conclusion, all hydrogels try to resemble extracellular matrix properties for bioprinted structures while maintaining good printability and stability during the printing process.
Collapse
Affiliation(s)
- David Patrocinio
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
| | - Victor Galván-Chacón
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
| | - J. Carlos Gómez-Blanco
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
| | - Sonia P. Miguel
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
- CICS-UBI, Health Science Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Jorge Loureiro
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
| | - Maximiano P. Ribeiro
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
- CICS-UBI, Health Science Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Paula Coutinho
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
- CICS-UBI, Health Science Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - J. Blas Pagador
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
- CIBER CV, Centro de Investigación Biomédica en Red—Enfermedades Cardiovasculares, 28029 Madrid, Spain;
| | - Francisco M. Sanchez-Margallo
- CIBER CV, Centro de Investigación Biomédica en Red—Enfermedades Cardiovasculares, 28029 Madrid, Spain;
- Scientific Direction, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain
- TERAV/ISCIII, Red Española de Terapias Avanzadas, Instituto de Salud Carlos III (RICORS, RD21/0017/0029), 28029 Madrid, Spain
| |
Collapse
|
20
|
Han X, Saiding Q, Cai X, Xiao Y, Wang P, Cai Z, Gong X, Gong W, Zhang X, Cui W. Intelligent Vascularized 3D/4D/5D/6D-Printed Tissue Scaffolds. NANO-MICRO LETTERS 2023; 15:239. [PMID: 37907770 PMCID: PMC10618155 DOI: 10.1007/s40820-023-01187-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023]
Abstract
Blood vessels are essential for nutrient and oxygen delivery and waste removal. Scaffold-repairing materials with functional vascular networks are widely used in bone tissue engineering. Additive manufacturing is a manufacturing technology that creates three-dimensional solids by stacking substances layer by layer, mainly including but not limited to 3D printing, but also 4D printing, 5D printing and 6D printing. It can be effectively combined with vascularization to meet the needs of vascularized tissue scaffolds by precisely tuning the mechanical structure and biological properties of smart vascular scaffolds. Herein, the development of neovascularization to vascularization to bone tissue engineering is systematically discussed in terms of the importance of vascularization to the tissue. Additionally, the research progress and future prospects of vascularized 3D printed scaffold materials are highlighted and presented in four categories: functional vascularized 3D printed scaffolds, cell-based vascularized 3D printed scaffolds, vascularized 3D printed scaffolds loaded with specific carriers and bionic vascularized 3D printed scaffolds. Finally, a brief review of vascularized additive manufacturing-tissue scaffolds in related tissues such as the vascular tissue engineering, cardiovascular system, skeletal muscle, soft tissue and a discussion of the challenges and development efforts leading to significant advances in intelligent vascularized tissue regeneration is presented.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, People's Republic of China
| | - Yi Xiao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China
| | - Xuan Gong
- University of Texas Southwestern Medical Center, Dallas, TX, 75390-9096, USA
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China.
| | - Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
21
|
Choi J, Lee EJ, Jang WB, Kwon SM. Development of Biocompatible 3D-Printed Artificial Blood Vessels through Multidimensional Approaches. J Funct Biomater 2023; 14:497. [PMID: 37888162 PMCID: PMC10607080 DOI: 10.3390/jfb14100497] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
Within the human body, the intricate network of blood vessels plays a pivotal role in transporting nutrients and oxygen and maintaining homeostasis. Bioprinting is an innovative technology with the potential to revolutionize this field by constructing complex multicellular structures. This technique offers the advantage of depositing individual cells, growth factors, and biochemical signals, thereby facilitating the growth of functional blood vessels. Despite the challenges in fabricating vascularized constructs, bioprinting has emerged as an advance in organ engineering. The continuous evolution of bioprinting technology and biomaterial knowledge provides an avenue to overcome the hurdles associated with vascularized tissue fabrication. This article provides an overview of the biofabrication process used to create vascular and vascularized constructs. It delves into the various techniques used in vascular engineering, including extrusion-, droplet-, and laser-based bioprinting methods. Integrating these techniques offers the prospect of crafting artificial blood vessels with remarkable precision and functionality. Therefore, the potential impact of bioprinting in vascular engineering is significant. With technological advances, it holds promise in revolutionizing organ transplantation, tissue engineering, and regenerative medicine. By mimicking the natural complexity of blood vessels, bioprinting brings us one step closer to engineering organs with functional vasculature, ushering in a new era of medical advancement.
Collapse
Affiliation(s)
- Jaewoo Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
22
|
He J, Sun Y, Gao Q, He C, Yao K, Wang T, Xie M, Yu K, Nie J, Chen Y, He Y. Gelatin Methacryloyl Hydrogel, from Standardization, Performance, to Biomedical Application. Adv Healthc Mater 2023; 12:e2300395. [PMID: 37115708 DOI: 10.1002/adhm.202300395] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/23/2023] [Indexed: 04/29/2023]
Abstract
Gelatin methacryloyl (GelMA), a photocurable hydrogel, is widely used in 3D culture, particularly in 3D bioprinting, due to its high biocompatibility, tunable physicochemical properties, and excellent formability. However, as the properties and performances of GelMA vary under different synthetic conditions, there is a lack of standardization, leading to conflicting results. In this study, a uniform standard is established to understand and enhance GelMA applications. First, the basic concept of GelMA and the density of the molecular network (DMN) are defined. Second, two properties, degrees of substitution and ratio of solid content, as the main measurable parameters determining the DMN are used. Third, the mechanisms and relationships between DMN and its performance in various applications in terms of porosity, viscosity, formability, mechanical strength, swelling, biodegradation, and cytocompatibility are theoretically explained. The main questions that are answered: what does performance mean, why is it important, how to optimize the basic parameters to improve the performance, and how to characterize it reasonably and accurately? Finally, it is hoped that this knowledge will eliminate the need for researchers to conduct tedious and repetitive pre-experiments, enable easy communication for achievements between groups under the same standard, and fully explore the potential of the GelMA hydrogel.
Collapse
Affiliation(s)
- Jing He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Engineering for Life Group (EFL), Suzhou, 215101, China
| | - Chanfan He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ke Yao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tongyao Wang
- State Key Laboratory of Catalysis, National Laboratory for Clean Energy, 2011-Collaborative Innovation Center of Chemistry for Energy Materials, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Kang Yu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Engineering for Life Group (EFL), Suzhou, 215101, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
23
|
Liu R, Wu S, Liu W, Wang L, Dong M, Niu W. microRNAs delivered by small extracellular vesicles in MSCs as an emerging tool for bone regeneration. Front Bioeng Biotechnol 2023; 11:1249860. [PMID: 37720323 PMCID: PMC10501734 DOI: 10.3389/fbioe.2023.1249860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Bone regeneration is a dynamic process that involves angiogenesis and the balance of osteogenesis and osteoclastogenesis. In bone tissue engineering, the transplantation of mesenchymal stem cells (MSCs) is a promising approach to restore bone homeostasis. MSCs, particularly their small extracellular vesicles (sEVs), exert therapeutic effects due to their paracrine capability. Increasing evidence indicates that microRNAs (miRNAs) delivered by sEVs from MSCs (MSCs-sEVs) can alter gene expression in recipient cells and enhance bone regeneration. As an ideal delivery vehicle of miRNAs, MSCs-sEVs combine the high bioavailability and stability of sEVs with osteogenic ability of miRNAs, which can effectively overcome the challenge of low delivery efficiency in miRNA therapy. In this review, we focus on the recent advancements in the use of miRNAs delivered by MSCs-sEVs for bone regeneration and disorders. Additionally, we summarize the changes in miRNA expression in osteogenic-related MSCs-sEVs under different microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian, China
| |
Collapse
|
24
|
Niu Y, Chen L, Wu T. Recent Advances in Bioengineering Bone Revascularization Based on Composite Materials Comprising Hydroxyapatite. Int J Mol Sci 2023; 24:12492. [PMID: 37569875 PMCID: PMC10419613 DOI: 10.3390/ijms241512492] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/18/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
The natural healing process of bone is impaired in the presence of tumors, trauma, or inflammation, necessitating external assistance for bone regeneration. The limitations of autologous/allogeneic bone grafting are still being discovered as research progresses. Bone tissue engineering (BTE) is now a crucial component of treating bone injuries and actively works to promote vascularization, a crucial stage in bone repair. A biomaterial with hydroxyapatite (HA), which resembles the mineral makeup of invertebrate bones and teeth, has demonstrated high osteoconductivity, bioactivity, and biocompatibility. However, due to its brittleness and porosity, which restrict its application, scientists have been prompted to explore ways to improve its properties by mixing it with other materials, modifying its structural composition, improving fabrication techniques and growth factor loading, and co-cultivating bone regrowth cells to stimulate vascularization. This review scrutinizes the latest five-year research on HA composite studies aimed at amplifying vascularization in bone regeneration.
Collapse
Affiliation(s)
- Yifan Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lei Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Tianfu Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
25
|
Caprio ND, Burdick JA. Engineered biomaterials to guide spheroid formation, function, and fabrication into 3D tissue constructs. Acta Biomater 2023; 165:4-18. [PMID: 36167240 PMCID: PMC10928646 DOI: 10.1016/j.actbio.2022.09.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/31/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022]
Abstract
Cellular spheroids are aggregates of cells that are being explored to address fundamental biological questions and as building blocks for engineered tissues. Spheroids possess distinct advantages over cellular monolayers or cell encapsulation in 3D natural and synthetic hydrogels, including direct cell-cell interactions and high cell densities, which better mimic aspects of many tissues. Despite these advantages, spheroid cultures often exhibit uncontrollable growth and may be too simplistic to mimic complex tissue structures. To address this, biomaterials are being leveraged to further expand the use of cellular spheroids for biomedical applications. In this review, we provide an overview of recent studies that utilize engineered biomaterials to guide spheroid formation and function, as well as their fabrication into tissues for use as tissue models and for therapeutic applications. First, we describe biomaterial strategies that allow the high-throughput fabrication of homogeneously-sized spheroids. Next, we summarize how engineered biomaterials are introduced into spheroid cultures either internally as microparticles or externally as hydrogel microenvironments to influence spheroid behavior (e.g., differentiation, fusion). Lastly, we discuss a variety of biofabrication strategies (e.g., 3D bioprinting, melt electrowriting) that have been used to develop macroscale tissue models and implantable constructs through the guided assembly of spheroids. Overall, the goal of this review is to provide a summary of how biomaterials are currently being engineered and leveraged to support spheroids in biomedical applications, as well as to provide a future outlook of the field. STATEMENT OF SIGNIFICANCE: Cellular spheroids are becoming increasingly used as in vitro tissue models or as 'building blocks' for tissue engineering and repair strategies. Engineered biomaterials and their processing through biofabrication approaches are being leveraged to structurally support and guide spheroid processes. This review summarizes current approaches where such biomaterials are being used to guide spheroid formation, function, and fabrication into tissue constructs. As the field is rapidly expanding, we also provide an outlook on future directions and how new engineered biomaterials can be implemented to further the development of biofabricated spheroid-based tissue constructs.
Collapse
Affiliation(s)
- Nikolas Di Caprio
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
26
|
Hu J, Shao J, Huang G, Zhang J, Pan S. In Vitro and In Vivo Applications of Magnesium-Enriched Biomaterials for Vascularized Osteogenesis in Bone Tissue Engineering: A Review of Literature. J Funct Biomater 2023; 14:326. [PMID: 37367290 DOI: 10.3390/jfb14060326] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Bone is a highly vascularized tissue, and the ability of magnesium (Mg) to promote osteogenesis and angiogenesis has been widely studied. The aim of bone tissue engineering is to repair bone tissue defects and restore its normal function. Various Mg-enriched materials that can promote angiogenesis and osteogenesis have been made. Here, we introduce several types of orthopedic clinical uses of Mg; recent advances in the study of metal materials releasing Mg ions (pure Mg, Mg alloy, coated Mg, Mg-rich composite, ceramic, and hydrogel) are reviewed. Most studies suggest that Mg can enhance vascularized osteogenesis in bone defect areas. Additionally, we summarized some research on the mechanisms related to vascularized osteogenesis. In addition, the experimental strategies for the research of Mg-enriched materials in the future are put forward, in which clarifying the specific mechanism of promoting angiogenesis is the crux.
Collapse
Affiliation(s)
- Jie Hu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiahui Shao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Gan Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jieyuan Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Shuting Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
27
|
Yang S, Hu H, Kung H, Zou R, Dai Y, Hu Y, Wang T, Lv T, Yu J, Li F. Organoids: The current status and biomedical applications. MedComm (Beijing) 2023; 4:e274. [PMID: 37215622 PMCID: PMC10192887 DOI: 10.1002/mco2.274] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Organoids are three-dimensional (3D) miniaturized versions of organs or tissues that are derived from cells with stem potential and can self-organize and differentiate into 3D cell masses, recapitulating the morphology and functions of their in vivo counterparts. Organoid culture is an emerging 3D culture technology, and organoids derived from various organs and tissues, such as the brain, lung, heart, liver, and kidney, have been generated. Compared with traditional bidimensional culture, organoid culture systems have the unique advantage of conserving parental gene expression and mutation characteristics, as well as long-term maintenance of the function and biological characteristics of the parental cells in vitro. All these features of organoids open up new opportunities for drug discovery, large-scale drug screening, and precision medicine. Another major application of organoids is disease modeling, and especially various hereditary diseases that are difficult to model in vitro have been modeled with organoids by combining genome editing technologies. Herein, we introduce the development and current advances in the organoid technology field. We focus on the applications of organoids in basic biology and clinical research, and also highlight their limitations and future perspectives. We hope that this review can provide a valuable reference for the developments and applications of organoids.
Collapse
Affiliation(s)
- Siqi Yang
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Haijie Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Hengchung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ruiqi Zou
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yushi Dai
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yafei Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Tiantian Wang
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tianrun Lv
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Jun Yu
- Departments of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Departments of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fuyu Li
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
28
|
Wong SK, Yee MMF, Chin KY, Ima-Nirwana S. A Review of the Application of Natural and Synthetic Scaffolds in Bone Regeneration. J Funct Biomater 2023; 14:jfb14050286. [PMID: 37233395 DOI: 10.3390/jfb14050286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023] Open
Abstract
The management of bone defects is complicated by the presence of clinical conditions, such as critical-sized defects created by high-energy trauma, tumour resection, infection, and skeletal abnormalities, whereby the bone regeneration capacity is compromised. A bone scaffold is a three-dimensional structure matrix serving as a template to be implanted into the defects to promote vascularisation, growth factor recruitment, osteogenesis, osteoconduction, and mechanical support. This review aims to summarise the types and applications of natural and synthetic scaffolds currently adopted in bone tissue engineering. The merits and caveats of natural and synthetic scaffolds will be discussed. A naturally derived bone scaffold offers a microenvironment closer to in vivo conditions after decellularisation and demineralisation, exhibiting excellent bioactivity, biocompatibility, and osteogenic properties. Meanwhile, an artificially produced bone scaffold allows for scalability and consistency with minimal risk of disease transmission. The combination of different materials to form scaffolds, along with bone cell seeding, biochemical cue incorporation, and bioactive molecule functionalisation, can provide additional or improved scaffold properties, allowing for a faster bone repair rate in bone injuries. This is the direction for future research in the field of bone growth and repair.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Michelle Min Fang Yee
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
29
|
Pan Y, Li Y, Dong W, Jiang B, Yu Y, Chen Y. Role of nano-hydrogels coated exosomes in bone tissue repair. Front Bioeng Biotechnol 2023; 11:1167012. [PMID: 37229488 PMCID: PMC10204869 DOI: 10.3389/fbioe.2023.1167012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
With the development of nanotechnology, nanomaterials are widely applied in different areas. Some nanomaterials are designed to be biocompatible and can be used in the medical field, playing an important role in disease treatment. Exosomes are nanoscale vesicles with a diameter of 30-200 nm. Studies have shown that exosomes have the effect of angiogenesis, tissue (skin, tendon, cartilage, et al.) repair and reconstruction. Nano-hydrogels are hydrogels with a diameter of 200 nm or less and can be used as the carrier to transport the exosomes into the body. Some orthopedic diseases, such as bone defects and bone infections, are difficult to handle. The emergence of nano-hydrogels coated exosomes may provide a new idea to solve these problems, improving the prognosis of patients. This review summarizes the function of nano-hydrogels coated exosomes in bone tissue repair, intending to illustrate the potential use and application of nano-hydrogels coated exosomes in bone disease.
Collapse
Affiliation(s)
- Yuqi Pan
- Department of Joint Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yige Li
- Department of Rehabilitation, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjun Dong
- Department of Joint Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowei Jiang
- Department of Joint Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhao Yu
- Department of Joint Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunsu Chen
- Department of Joint Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Pham CHL, Zuo Y, Chen Y, Tran NM, Nguyen DT, Dang TT. Waffle-inspired hydrogel-based macrodevice for spatially controlled distribution of encapsulated therapeutic microtissues and pro-angiogenic endothelial cells. Bioeng Transl Med 2023; 8:e10495. [PMID: 37206238 PMCID: PMC10189477 DOI: 10.1002/btm2.10495] [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: 08/26/2022] [Revised: 12/26/2022] [Accepted: 01/13/2023] [Indexed: 03/17/2023] Open
Abstract
Macro-encapsulation systems for delivery of cellular therapeutics in diabetes treatment offer major advantages such as device retrievability and high cell packing density. However, microtissue aggregation and absence of vasculature have been implicated in the inadequate transfer of nutrients and oxygen to the transplanted cellular grafts. Herein, we develop a hydrogel-based macrodevice to encapsulate therapeutic microtissues positioned in homogeneous spatial distribution to mitigate their aggregation while concurrently supporting an organized intra-device network of vascular-inductive cells. Termed Waffle-inspired Interlocking Macro-encapsulation (WIM) device, this platform comprises two modules with complementary topography features that fit together in a lock-and-key configuration. The waffle-inspired grid-like micropattern of the "lock" component effectively entraps insulin-secreting microtissues in controlled locations while the interlocking design places them in a co-planar spatial arrangement with close proximity to vascular-inductive cells. The WIM device co-laden with INS-1E microtissues and human umbilical vascular endothelial cells (HUVECs) maintains desirable cellular viability in vitro with the encapsulated microtissues retaining their glucose-responsive insulin secretion while embedded HUVECs express pro-angiogenic markers. Furthermore, a subcutaneously implanted alginate-coated WIM device encapsulating primary rat islets achieves blood glucose control for 2 weeks in chemically induced diabetic mice. Overall, this macrodevice design lays foundation for a cell delivery platform, which has the potential to facilitate nutrients and oxygen transport to therapeutic grafts and thereby might lead to improved disease management outcome.
Collapse
Affiliation(s)
- Chi H. L. Pham
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Yicong Zuo
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Yang Chen
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Nam M. Tran
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Dang T. Nguyen
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Tram T. Dang
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| |
Collapse
|
31
|
Molino BZ, O'Connell C, Kageyama T, Yan L, Wu Y, Kawamura I, Maruo S, Fukuda J. Gelatin acrylamide with improved UV crosslinking and mechanical properties for 3D biofabrication. J Biosci Bioeng 2023:S1389-1723(23)00109-3. [PMID: 37121831 DOI: 10.1016/j.jbiosc.2023.03.014] [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: 03/30/2022] [Revised: 03/02/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023]
Abstract
Photocrosslinkable gelatin has attracted increasing interest in the field of biofabrication, with the most studied and widely used photocrosslinkable gelatin being gelatin methacrylate (GelMa). However, the 3D fabrication of GelMa has presented several limitations and challenges, primarily due to its slow crosslinking speed. It is generally known that acryl-based functional groups have faster reaction kinetics than methacryl-base groups. However, gelatin acrylamide (GelAc) has not been widely investigated, largely due to its increased complexity of synthesis relative to GelMA. In this study, we developed a novel synthesis method for GelAc. By varying the reaction ratio of reagents, GelAc with a degree of substitution from 20% to 95% was produced. The UV crosslinking properties of GelAc was studied, demonstrating significantly faster crosslinking kinetics than GelMa, especially at lower concentrations and low photoinitiator concentrations. The swelling ratio and mechanical properties of the crosslinked GelAc hydrogel were also characterized, and biocompatibility experiments conducted via both surface seeding and hydrogel encapsulation of cells, with good cell viability observed. The application of GelAc for 3D biofabrication was demonstrated by 3D printing. GelAc can be a useful material for the fabrication of 3D conduits for tissue engineering applications.
Collapse
Affiliation(s)
- Binbin Zhang Molino
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Cathal O'Connell
- School of Engineering, Discipline of Electrical and Biomedical Engineering, RMIT University, 124 La Trobe St, Melbourne VIC 3000, Australia; BioFab3D, Aikenhead Center for Medical Discovery, St Vincent's Hospital Melbourne, 29 Regent St, Fitzroy VIC 3065, Australia
| | - Tatsuto Kageyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Lei Yan
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Yumeng Wu
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Izuru Kawamura
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Shoji Maruo
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan.
| |
Collapse
|
32
|
Li P, Zhang M, Chen Z, Tian B, Kang X. Tissue-Engineered Injectable Gelatin-Methacryloyl Hydrogel-Based Adjunctive Therapy for Intervertebral Disc Degeneration. ACS OMEGA 2023; 8:13509-13518. [PMID: 37091429 PMCID: PMC10116505 DOI: 10.1021/acsomega.3c00211] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/14/2023] [Indexed: 05/03/2023]
Abstract
Gelatin-methacryloyl (GelMA) hydrogels are photosensitive with good biocompatibility and adjustable mechanical properties. The GelMA hydrogel composite system is a prospective therapeutic material based on a tissue engineering platform for treating intervertebral disc (IVD) degeneration (IVDD). The potential application value of the GelMA hydrogel composite system in the treatment of IVDD mainly includes three aspects: first, optimization of the current clinical treatment methods, including conservative treatment and surgical treatment; second, regeneration of IVD cells to reverse or repair IVDD; and finally, IVDD instead of injury plays a biomechanical role. In this paper, we summarized and analyzed the preparation of GelMA hydrogels and their excellent biological characteristics as carriers and comprehensively demonstrated the research status and prospects of GelMA hydrogel composite systems in IVDD treatment. In addition, the challenges facing the application of GelMA hydrogel composite systems and the progress of research on new hydrogels modified by GelMA hydrogels are presented. Hopefully, this study will provide theoretical guidance for the future application of GelMA hydrogel composite systems in IVDD.
Collapse
Affiliation(s)
- Peng Li
- Department
of Hand Surgery, Honghui Hospital, Xi’an
Jiao Tong University, Shaanxi 710054, P.R. China
| | - Ming Zhang
- Department
of General Practice, Honghui Hospital, Xi’an
Jiao Tong University, Shaanxi 710054, P.R. China
| | - Zhengyu Chen
- Department
of Spine Surgery, Xianyang First People’s
Hospital, Shaanxi, 712000, P.R. China
| | - Bin Tian
- Department
of Sports Medicine, Honghui Hospital, Xi’an
Jiao Tong University, Shaanxi 710054, P.R. China
| | - Xin Kang
- Department
of Sports Medicine, Honghui Hospital, Xi’an
Jiao Tong University, Shaanxi 710054, P.R. China
- E-mail:
| |
Collapse
|
33
|
Liu J, Yang L, Liu K, Gao F. Hydrogel scaffolds in bone regeneration: Their promising roles in angiogenesis. Front Pharmacol 2023; 14:1050954. [PMID: 36860296 PMCID: PMC9968752 DOI: 10.3389/fphar.2023.1050954] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Bone tissue engineering (BTE) has become a hopeful potential treatment strategy for large bone defects, including bone tumors, trauma, and extensive fractures, where the self-healing property of bone cannot repair the defect. Bone tissue engineering is composed of three main elements: progenitor/stem cells, scaffold, and growth factors/biochemical cues. Among the various biomaterial scaffolds, hydrogels are broadly used in bone tissue engineering owing to their biocompatibility, controllable mechanical characteristics, osteoconductive, and osteoinductive properties. During bone tissue engineering, angiogenesis plays a central role in the failure or success of bone reconstruction via discarding wastes and providing oxygen, minerals, nutrients, and growth factors to the injured microenvironment. This review presents an overview of bone tissue engineering and its requirements, hydrogel structure and characterization, the applications of hydrogels in bone regeneration, and the promising roles of hydrogels in bone angiogenesis during bone tissue engineering.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Lili Yang
- Department of Spinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kexin Liu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Gao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China,*Correspondence: Feng Gao,
| |
Collapse
|
34
|
Ghahri T, Salehi Z, Aghajanpour S, Eslaminejad MB, Kalantari N, Akrami M, Dinarvand R, Jang HL, Esfandyari-Manesh M. Development of osteon-like scaffold-cell construct by quadruple coaxial extrusion-based 3D bioprinting of nanocomposite hydrogel. BIOMATERIALS ADVANCES 2023; 145:213254. [PMID: 36584583 DOI: 10.1016/j.bioadv.2022.213254] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Despite advances in bone tissue engineering, fabricating a scaffold which can be used as an implant for large bone defects remains challenge. One of the great importance in fabricating a biomimetic bone implant is considering the possibility of the integration of the structure and function of implants with hierarchical structure of bone. Herein, we propose a method to mimic the structural unit of compact bone, osteon, with spatial pattern of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) in the adjacent layers that mimic Haversian canal and lamella, respectively. To this end, coaxial extrusion-based bioprinting technique via a customized quadruple-layer core-shell nozzle was employed. 3D implant scaffold-cell construct was fabricated by using polyethylene glycol as a hollowing agent in the first layer, gelatin methacryloyl (GelMA) and alginate blended hydrogel encapsulating HUVEC cells with vascular endothelial growth factor nanoparticles in the second layer (vasculogenic layer) to mimic vascular vessel, and GelMA and alginate blended hydrogel containing hMSCs cells in the outer osteogenic layer to imitate lamella. Two types of bone minerals, whitlockite and hydroxyapatite, were incorporated in osteogenic layer to induce osteoblastic differentiation and enhance mechanical properties (the young's modules of nanocomposite increased from 35 kPa to 80 kPa). In-vitro evaluations demonstrated high cell viability (94 % within 10 days) and proliferation. Furthermore, ALP enzyme activity increased considerably within 2 weeks and mineralized extra cellular matrix considerably produced within 3 weeks. Also, a significant increase in osteogenic markers was observed indicating the presence of differentiated osteoblast cells. Therefore, the work indicates the potential of single step 3D bioprinting process to fabricate biomimetic osteons to use as bone grafts for regeneration.
Collapse
Affiliation(s)
- Tahmineh Ghahri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Biotechnology Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Zeinab Salehi
- Department of Biotechnology Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Sareh Aghajanpour
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Kalantari
- Department of Stem Cells and Developmental Biology, Royan institute, Tehran, Iran
| | - Mohammad Akrami
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Institute of Biomaterials, University of Tehran & Tehran University of Medical Sciences (IBUTUMS), Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Leicester School of Pharmacy, De Montfort University, Leicester, UK.
| | - Hae Lin Jang
- Center for Engineered Therapeutics, Department of Medicine and Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
35
|
Alarçin E, İzbudak B, Yüce Erarslan E, Domingo S, Tutar R, Titi K, Kocaaga B, Guner FS, Bal-Öztürk A. Optimization of methacrylated gelatin /layered double hydroxides nanocomposite cell-laden hydrogel bioinks with high printability for 3D extrusion bioprinting. J Biomed Mater Res A 2023; 111:209-223. [PMID: 36213938 DOI: 10.1002/jbm.a.37450] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022]
Abstract
Layered double hydroxides (LDHs) offer unique source of inspiration for design of bone mimetic biomaterials due to their superior mechanical properties, drug delivery capability and regulation cellular behaviors, particularly by divalent metal cations in their structure. Three-dimensional (3D) bioprinting of LDHs holds great promise as a novel strategy thanks to highly tunable physiochemical properties and shear-thinning ability of LDHs, which allow shape fidelity after deposition. Herein, we introduce a straightforward strategy for extrusion bioprinting of cell laden nanocomposite hydrogel bioink of gelatin methacryloyl (GelMA) biopolymer and LDHs nanoparticles. First, we synthesized LDHs by co-precipitation process and systematically examined the effect of LDHs addition on printing parameters such as printing pressure, extrusion rate, printing speed, and finally bioink printability in creating grid-like constructs. The developed hydrogel bioinks provided precise control over extrudability, extrusion uniformity, and structural integrity after deposition. Based on the printability and rheological analysis, the printability could be altered by controlling the concentration of LDHs, and printability was found to be ideal with the addition of 3 wt % LDHs. The addition of LDHs resulted in remarkably enhanced compressive strength from 652 kPa (G-LDH0) to 1168 kPa (G-LDH3). It was shown that the printed nanocomposite hydrogel scaffolds were able to support encapsulated osteoblast survival, spreading, and proliferation in the absence of any osteoinductive factors taking advantage of LDHs. In addition, cells encapsulated in G-LDH3 had a larger cell spreading area and higher cell aspect ratio than those encapsulated in G-LDH0. Altogether, the results demonstrated that the developed GelMA/LDHs nanocomposite hydrogel bioink revealed a high potential for extrusion bioprinting with high structural fidelity to fabricate implantable 3D hydrogel constructs for repair of bone defects.
Collapse
Affiliation(s)
- Emine Alarçin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Burçin İzbudak
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey
| | - Elif Yüce Erarslan
- Chemical Engineering Department, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sherif Domingo
- Chemical Engineering Department, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kariman Titi
- Department of Chemistry, Faculty of Science and Technology, Hebron University, Hebron, West Bank, Palestine
| | - Banu Kocaaga
- Department of Chemical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - F Seniha Guner
- Department of Chemical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ayça Bal-Öztürk
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey.,Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Istanbul, Turkey.,3D Bioprinting Design&Prototyping R&D Center, Istinye University, Istanbul, Turkey
| |
Collapse
|
36
|
Rougier G, Maistriaux L, Fievé L, Xhema D, Evrard R, Manon J, Olszewski R, Szmytka F, Thurieau N, Boisson J, Kadlub N, Gianello P, Behets C, Lengelé B. Decellularized vascularized bone grafts: A preliminary in vitro porcine model for bioengineered transplantable bone shafts. Front Bioeng Biotechnol 2023; 10:1003861. [PMID: 36743653 PMCID: PMC9890275 DOI: 10.3389/fbioe.2022.1003861] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/09/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction: Durable reconstruction of critical size bone defects is still a surgical challenge despite the availability of numerous autologous and substitute bone options. In this paper, we have investigated the possibility of creating a living bone allograft, using the perfusion/decellularization/recellularization (PDR) technique, which was applied to an original model of vascularized porcine bone graft. Materials and Methods: 11 porcine bone forelimbs, including radius and ulna, were harvested along with their vasculature including the interosseous artery and then decellularized using a sequential detergent perfusion protocol. Cellular clearance, vasculature, extracellular matrix (ECM), and preservation of biomechanical properties were evaluated. The cytocompatibility and in vitro osteoinductive potential of acellular extracellular matrix were studied by static seeding of NIH-3T3 cells and porcine adipose mesenchymal stem cells (pAMSC), respectively. Results: The vascularized bone grafts were successfully decellularized, with an excellent preservation of the 3D morphology and ECM microarchitecture. Measurements of DNA and ECM components revealed complete cellular clearance and preservation of ECM's major proteins. Bone mineral density (BMD) acquisitions revealed a slight, yet non-significant, decrease after decellularization, while biomechanical testing was unmodified. Cone beam computed tomography (CBCT) acquisitions after vascular injection of barium sulphate confirmed the preservation of the vascular network throughout the whole graft. The non-toxicity of the scaffold was proven by the very low amount of residual sodium dodecyl sulfate (SDS) in the ECM and confirmed by the high live/dead ratio of fibroblasts seeded on periosteum and bone ECM-grafts after 3, 7, and 16 days of culture. Moreover, cell proliferation tests showed a significant multiplication of seeded cell populations at the same endpoints. Lastly, the differentiation study using pAMSC confirmed the ECM graft's potential to promote osteogenic differentiation. An osteoid-like deposition occurred when pAMSC were cultured on bone ECM in both proliferative and osteogenic differentiation media. Conclusion: Fully decellularized bone grafts can be obtained by perfusion decellularization, thereby preserving ECM architecture and their vascular network, while promoting cell growth and differentiation. These vascularized decellularized bone shaft allografts thus present a true potential for future in vivo reimplantation. Therefore, they may offer new perspectives for repairing large bone defects and for bone tissue engineering.
Collapse
Affiliation(s)
- Guillaume Rougier
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Department of Oncological and Cervicofacial Reconstructive Surgery, Otorhinolaryngology, Maxillofacial Surgery—Institut Curie, Paris, France
| | - Louis Maistriaux
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,*Correspondence: Louis Maistriaux,
| | - Lies Fievé
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Daela Xhema
- Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Robin Evrard
- Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Neuromusculoskeletal Lab (NMSK)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Julie Manon
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Neuromusculoskeletal Lab (NMSK)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Raphael Olszewski
- Neuromusculoskeletal Lab (NMSK)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Department of Maxillofacial Surgery and Stomatology—Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Fabien Szmytka
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France
| | - Nicolas Thurieau
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France
| | - Jean Boisson
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France
| | - Natacha Kadlub
- IMSIA, ENSTA Paris, Institut Polytechnique de Paris, Palaiseau, France,Department of Maxillofacial and Reconstructive Surgery—Necker Enfants Malades, Paris, France
| | - Pierre Gianello
- Pole of Experimental Surgery and Transplantation (CHEX)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Catherine Behets
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium
| | - Benoît Lengelé
- Pole of Morphology (MORF)—Institute of Experimental and Clinical Research (IREC)—UCLouvain, Brussels, Belgium,Department of Plastic and Reconstructive Surgery—Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
37
|
Decellularized vascularized bone grafts as therapeutic solution for bone reconstruction: A mechanical evaluation. PLoS One 2023; 18:e0280193. [PMID: 36638107 PMCID: PMC9838862 DOI: 10.1371/journal.pone.0280193] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Large bone defects are challenging for surgeons. Available reimplanted bone substitutes can't properly restore optimal function along and long term osteointegration of the bone graft. Bone substitute based on the perfusion-decellularization technique seem to be interesting in order to overcome these limitations. We present here an evaluation of the biomechanics of the bones thus obtained. MATERIAL AND METHODS Two decellularization protocols were chosen for this study. One using Sodium Dodecyl Sulfate (SDS) (D1) and one using NaOH and H2O2 (D2). The decellularization was performed on porcine forearms. We then carried out compression, three-point bending, indentation and screw pull-out tests on each sample. Once these tests were completed, we compared the results obtained between the different decellularization protocols and with samples left native. RESULTS The difference in the means was similar between the tests performed on bones decellularized with the SDS protocol and native bones for pull-out test: +1.4% (CI95% [-10.5%- 12.4%]) of mean differences when comparing Native vs D1, compression -14.9% (CI95% [-42.7%- 12.5%]), 3-point bending -5.7% (CI95% [-22.5%- 11.1%]) and indentation -10.8% (CI95% [-19.5%- 4.6%]). Bones decellularized with the NaOH protocol showed different results from those obtained with the SDS protocol or native bones during the pull-out screw +40.7% (CI95% [24.3%- 57%]) for Native vs D2 protocol and 3-point bending tests +39.2% (CI95% [13.7%- 64.6%]) for Native vs D2 protocol. The other tests, compression and indentation, gave similar results for all our samples. CONCLUSION Vascularized decellularized grafts seem to be an interesting means for bone reconstruction. Our study shows that the decellularization method affects the mechanical results of our specimens. Some methods seem to limit these alterations and could be used in the future for bone decellularization.
Collapse
|
38
|
Xu Z, Wu L, Tang Y, Xi K, Tang J, Xu Y, Xu J, Lu J, Guo K, Gu Y, Chen L. Spatiotemporal Regulation of the Bone Immune Microenvironment via Dam-Like Biphasic Bionic Periosteum for Bone Regeneration. Adv Healthc Mater 2023; 12:e2201661. [PMID: 36189833 PMCID: PMC11469314 DOI: 10.1002/adhm.202201661] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/24/2022] [Indexed: 02/03/2023]
Abstract
The bone immune microenvironment (BIM) regulates bone regeneration and affects the prognosis of fractures. However, there is currently no effective strategy that can precisely modulate macrophage polarization to improve BIM for bone regeneration. Herein, a hybridized biphasic bionic periosteum, inspired by the BIM and functional structure of the natural periosteum, is presented. The gel phase is composed of genipin-crosslinked carboxymethyl chitosan and collagen self-assembled hybrid hydrogels, which act as the "dam" to intercept IL-4 released during the initial burst from the bionic periosteum fiber phase, thus maintaining the moderate inflammatory response of M1 macrophages for mesenchymal stem cell recruitment and vascular sprouting at the acute fracture. With the degradation of the gel phase, released IL-4 cooperates with collagen to promote the polarization towards M2 macrophages, which reconfigure the local microenvironment by secreting PDGF-BB and BMP-2 to improve vascular maturation and osteogenesis twofold. In rat cranial defect models, the controlled regulation of the BIM is validated with the temporal transition of the inflammatory/anti-inflammatory process to achieve faster and better bone defect repair. This strategy provides a drug delivery system that constructs a coordinated BIM, so as to break through the predicament of the contradiction between immune response and bone tissue regeneration.
Collapse
Affiliation(s)
- Zonghan Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Liang Wu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Yu Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Kun Xi
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jincheng Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Yichang Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jingzhi Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jian Lu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Kaijin Guo
- Department of Orthopedicsthe Affiliated Hospital of Xuzhou Medical University99 Huaihai West RoadXuzhouJiangsu221000P. R. China
| | - Yong Gu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Liang Chen
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| |
Collapse
|
39
|
de Souza TV, Pastena Giorno L, Malmonge SM, Santos AR. Bioprinting: From Technique to Application in Tissue Engineering and Regenerative Medicine. Curr Mol Med 2023; 23:934-951. [PMID: 36017861 DOI: 10.2174/1566524023666220822152448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/22/2022] [Accepted: 05/30/2022] [Indexed: 11/22/2022]
Abstract
Among the different approaches present in regenerative medicine and tissue engineering, the one that has attracted the most interest in recent years is the possibility of printing functional biological tissues. Bioprinting is a technique that has been applied to create cellularized three-dimensional structures that mimic biological tissues and thus allow their replacement. Hydrogels are interesting materials for this type of technique. Hydrogels based on natural polymers are known due to their biocompatible properties, in addition to being attractive biomaterials for cell encapsulation. They provide a threedimensional aqueous environment with biologically relevant chemical and physical signals, mimicking the natural environment of the extracellular matrix (ECM). Bioinks are ink formulations that allow the printing of living cells. The controlled deposition of biomaterials by bioinks needs to maintain cell viability and offer specific biochemical and physical stimuli capable of guiding cell migration, proliferation, and differentiation. In this work, we analyze the theoretical and practical issues of bioprinting, citing currently used methods, their advantages, and limitations. We present some important molecules that have been used to compose bioinks, as well as the cellular responses that have been observed in different tissues. Finally, we indicate future perspectives of the method.
Collapse
Affiliation(s)
- Thaís Vieira de Souza
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Luciana Pastena Giorno
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Sonia Maria Malmonge
- Centro de Engenharia, Modelagem e Ciências Sociais Aplicadas, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Arnaldo R Santos
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| |
Collapse
|
40
|
Peng Z, Pang H, Wu H, Peng X, Tan Q, Lin S, Wei B. CCL2 promotes proliferation, migration and angiogenesis through the MAPK/ERK1/2/MMP9, PI3K/AKT, Wnt/β‑catenin signaling pathways in HUVECs. Exp Ther Med 2022; 25:77. [PMID: 36684650 PMCID: PMC9842938 DOI: 10.3892/etm.2022.11776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/28/2022] [Indexed: 12/28/2022] Open
Abstract
Severe bone trauma can lead to poor or delayed bone healing and nonunion. Bone regeneration is based on the interaction between osteogenesis and angiogenesis. Angiogenesis serves a unique role in the repair and remodeling of bone defects. Monocyte chemoattractant protein-1, also known as CC motif ligand 2 (CCL2), is a member of the CC motif chemokine family and was the first human chemokine to be revealed to be an effective chemokine of monocytes. However, its underlying mechanism in angiogenesis of bone defect repair remains to be elucidated. Therefore, the present study investigated the detailed mechanism by which CCL2 promoted angiogenesis in bone defects based on cell and animal model experiments. In the present study, CCL2 promoted proliferation, migration and tube formation in human umbilical vein endothelial cells (HUVECs) in a concentration-dependent manner. Western blot analysis revealed that treatment of HUVECs with CCL2 upregulated the protein expression levels of rho-associated coiled-coil-containing protein kinase (Rock)1, Rock2, N-cadherin, c-Myc and VEGFR2. Furthermore, CCL2 promoted the expression of MAPK/ERK1/2/MMP9, PI3K/AKT and Wnt/β-catenin signaling pathway-related proteins, which also demonstrated that CCL2 promoted these functions in HUVECs. Immunohistochemical staining of Sprague Dawley rat femurs following bone defects revealed that VEGF expression was positive in the newly formed bone area in each group, while the expression area of VEGF in the CCL2 addition group was markedly increased. Therefore, CCL2 is a potential therapeutic approach for bone defect repair and reconstruction through the mechanism of angiogenesis-osteogenesis coupling.
Collapse
Affiliation(s)
- Zhonghua Peng
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - He Pang
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hang Wu
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xin Peng
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Qichao Tan
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Sien Lin
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China,Correspondence to: Dr Sien Lin or Dr Bo Wei, Department of Orthopedics Center, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Xiashan, Zhanjiang, Guangdong 524001, P.R. China
| | - Bo Wei
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China,Correspondence to: Dr Sien Lin or Dr Bo Wei, Department of Orthopedics Center, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Xiashan, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
41
|
Yokoi T, Watanabe M, Goto T, Meng S, Sekino T, Shimabukuro M, Kawashita M. Synthesis of Octacalcium Phosphate Containing Glutarate Ions with a High Incorporation Fraction. MATERIALS (BASEL, SWITZERLAND) 2022; 16:64. [PMID: 36614402 PMCID: PMC9821757 DOI: 10.3390/ma16010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
Octacalcium phosphate (OCP) has received considerable attention in the field of ceramic biomaterials as an advanced functional material. It exhibits a layered structure composed of apatitic and hydrated layers and can incorporate various dicarboxylate ions into the hydrated layer. Saturated dicarboxylic acids (HOOC(CH2)nCOOH) with an odd number of methylene groups (-CH2-) exhibit lower incorporation fractions than those with an even number of methylene groups, possibly owing to a compositional dependence on the synthetic method. In this study, calcium carbonate, phosphoric acid, and various amounts of glutaric acid were used to produce glutarate-ion-incorporated OCP by a wet chemical method, which is different from the conventional synthetic strategy. While utilising 1-20 mmol of glutaric acid during synthesis did not produce the desired product, using 25 mmol of glutaric acid resulted in the formation of single-phase glutarate-ion-incorporated OCP with a Ca/P molar ratio of 1.57 and a 90% incorporation fraction of glutarate ions. This glutarate-ion-incorporation fraction is significantly higher than that reported in the previous studies (35%). Thus, the synthetic procedure proposed herein was able to produce single-phase OCP containing glutarate ions with a high incorporation fraction. Our findings can contribute to development of novel functional ceramic biomaterials in the future.
Collapse
Affiliation(s)
- Taishi Yokoi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Masahiro Watanabe
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Tomoyo Goto
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mohogaoka, Ibaraki, Osaka 567-0047, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, 1-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sikun Meng
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Tohru Sekino
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mohogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Masaya Shimabukuro
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Masakazu Kawashita
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| |
Collapse
|
42
|
Aaryasree K, Yagnik A, Chordiya PK, Choudhury K, Kumar P. Nature-Inspired Vascularised Materials and Devices for Biomedical Engineering. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
43
|
Shen M, Wang L, Gao Y, Feng L, Xu C, Li S, Wang X, Wu Y, Guo Y, Pei G. 3D bioprinting of in situ vascularized tissue engineered bone for repairing large segmental bone defects. Mater Today Bio 2022; 16:100382. [PMID: 36033373 PMCID: PMC9403505 DOI: 10.1016/j.mtbio.2022.100382] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 11/18/2022] Open
Abstract
Large bone defects remain an unsolved clinical challenge because of the lack of effective vascularization in newly formed bone tissue. 3D bioprinting is a fabrication technology with the potential to create vascularized bone grafts with biological activity for repairing bone defects. In this study, vascular endothelial cells laden with thermosensitive bio-ink were bioprinted in situ on the inner surfaces of interconnected tubular channels of bone mesenchymal stem cell-laden 3D-bioprinted scaffolds. Endothelial cells exhibited a more uniform distribution and greater seeding efficiency throughout the channels. In vitro, the in situ bioprinted endothelial cells can form a vascular network through proliferation and migration. The in situ vascularized tissue-engineered bone also resulted in a coupling effect between angiogenesis and osteogenesis. Moreover, RNA sequencing analysis revealed that the expression of genes related to osteogenesis and angiogenesis is upregulated in biological processes. The in vivo 3D-bioprinted in situ vascularized scaffolds exhibited excellent performance in promoting new bone formation in rat calvarial critical-sized defect models. Consequently, in situ vascularized tissue-engineered bones constructed using 3D bioprinting technology have a potential of being used as bone grafts for repairing large bone defects, with a possible clinical application in the future. 3D bioprinting was used to fabricate in situ vascularized tissue engineered bone. In situ bioprinted endothelial cells exhibited uniform distribution and greater seeding efficiency. 3D-bioprinted scaffold produced coupling between angiogenesis and osteogenesis.
Collapse
Key Words
- 3D bioprinted BMSCs-laden GelMA hydrogel scaffold, (GB)
- 3D bioprinting
- 3D dual-extrusion bioprinted BMSCs-laden GelMA hydrogel and RAOECs-laden 3P hydrogel scaffold, (GB-3PR)
- 3D dual-extrusion bioprinted GelMA hydrogel and RAOECs-laden 3P hydrogel scaffold, (G-3PR)
- 3D printed GelMA hydrogel scaffold, (G)
- 4′,6-diamidino-2-phenylindole, (DAPI)
- Alizarin red S, (ARS)
- Alkaline phosphatase, (ALP)
- Dulbecco's modified Eagle's medium, (DMEM)
- Dulbecco's phosphate-buffered saline, (DPBS)
- Fourier-transform infrared, (FTIR)
- In situ vascularization
- Large segmental bone defects
- PLA-PEG-PLA, (3P)
- RNA sequencing Analysis
- Tissue engineering
- analysis of variance, (ANOVA)
- bone mesenchymal stem cells, (BMSCs)
- bone mineral density, (BMD)
- bone volume to tissue volume, (BV/TV)
- complementary DNA, (cDNA)
- differentially expressed genes, (DEGs)
- endothelial cells, (ECs)
- ethylenediamine tetraacetic acid, (EDTA)
- extracellular matrix, (ECM)
- fetal bovine serum, (FBS)
- gelatin methacryloyl, (GelMA)
- gene ontology, (GO)
- glyceraldehyde-3-phosphate dehydrogenase, (GAPDH)
- green fluorescent protein, (GFP)
- hematoxylin and eosin, (H&E)
- lithium phenyl-2,4,6-trimethylbenzoylphosphinate, (LAP)
- micro-computed tomography, (micro-CT)
- nuclear magnetic resonance, (NMR)
- optical density, (OD)
- paraformaldehyde, (PFA)
- phosphate-buffered saline, (PBS)
- polyethylene glycol, (PEG)
- polylactic acid, (PLA)
- polyvinylidene fluoride, (PVDF)
- radioimmunoprecipitation assay, (RIPA)
- rat aortic endothelial cells, (RAOECs)
- real-time polymerase chain reaction, (RT-PCR)
- standard deviation, (SD)
- tissue-engineered bone, (TEB)
- tris buffered saline with Tween-20, (TBST)
Collapse
Affiliation(s)
- Mingkui Shen
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lulu Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yi Gao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Li Feng
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chuangye Xu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sijing Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xiaohu Wang
- Department of Orthopedics, Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Yulan Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yao Guo
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
- Corresponding author.
| | - Guoxian Pei
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
- Corresponding author.
| |
Collapse
|
44
|
Kang X, Zhang XB, Gao XD, Hao DJ, Li T, Xu ZW. Bioprinting for bone tissue engineering. Front Bioeng Biotechnol 2022; 10:1036375. [PMID: 36507261 PMCID: PMC9732272 DOI: 10.3389/fbioe.2022.1036375] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Abstract
The shape transformation characteristics of four-dimensional (4D)-printed bone structures can meet the individual bone regeneration needs, while their structure can be programmed to cross-link or reassemble by stimulating responsive materials. At the same time, it can be used to design vascularized bone structures that help establish a bionic microenvironment, thus influencing cellular behavior and enhancing stem cell differentiation in the postprinting phase. These developments significantly improve conventional three-dimensional (3D)-printed bone structures with enhanced functional adaptability, providing theoretical support to fabricate bone structures to adapt to defective areas dynamically. The printing inks used are stimulus-responsive materials that enable spatiotemporal distribution, maintenance of bioactivity and cellular release for bone, vascular and neural tissue regeneration. This paper discusses the limitations of current bone defect therapies, 4D printing materials used to stimulate bone tissue engineering (e.g., hydrogels), the printing process, the printing classification and their value for clinical applications. We focus on summarizing the technical challenges faced to provide novel therapeutic implications for bone defect repair.
Collapse
Affiliation(s)
- Xin Kang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong University, Xian, Shaanxi, China
| | - Xiao-Bo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong University, Xian, Shaanxi, China
| | - Xi-Dan Gao
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Ding-Jun Hao
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong University, Xian, Shaanxi, China
| | - Tao Li
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong University, Xian, Shaanxi, China
| | - Zheng-Wei Xu
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong University, Xian, Shaanxi, China,*Correspondence: Zheng-Wei Xu,
| |
Collapse
|
45
|
Ma J, Zhao S, Li Y, Hu J, Zhang L, Zhou X, Yan L. Stereoscopic projection lithography based 3D printing with high precision for advanced tissue engineering application. Front Bioeng Biotechnol 2022; 10:1074157. [DOI: 10.3389/fbioe.2022.1074157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/07/2022] [Indexed: 11/20/2022] Open
Abstract
The emergence of tissue engineering technology provides an option for the treatment of early organ and tissue lesions by combination of biomimetic scaffolds and stem cells. Stereoscopic projection lithography is utilized broadly in varied application areas due to its high-precision, resolution, and efficiency features. It can be used to fabricate and manufacture complex scaffolds with hierarchical construct, which are highly suitable for advanced tissue engineering application. In current work, gelatin methacrylate (GelMA) was synthesized and fabricated to bioactive scaffold because of its excellent biocompatibility and biodegradability by using stereoscopic projection lithography based 3D printer (YC-M3D-10). The scaffold displayed multilayered micro structures that supported stem cell growth and promoted cell proliferation. The results demonstrated that the cells proliferated significantly on the printed GelMA scaffold after 6 days. Moreover, GelMA scaffolds can promote cell proliferation and show great prospects in future tissue engineering applications.
Collapse
|
46
|
Zhang X, Hu Y, Hao D, Li T, Jia Y, Hu W, Xu Z. New strategies for the treatment of intervertebral disc degeneration: cell, exosome, gene, and tissue engineering. Am J Transl Res 2022; 14:8031-8048. [PMID: 36505274 PMCID: PMC9730054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
Abstract
Low back pain (LBP) caused by intervertebral disc (IVD) generation (IVDD) has always been an important problem that cannot be ignored. Traditional therapies have many deep-rooted and intractable complications that promote their treatment mode transfer to new therapies. This article mainly summarizes the shortcomings of traditional treatment methods and analyzes the research status and future development direction of IVDD treatment. We outlined the most promising IVDD therapies, including cell, exosome, gene, and tissue engineering therapy, especially tissue engineering therapy, which runs through the whole process of other therapies. In addition, the article focuses on the cellular, animal, and preclinical challenges faced by each therapeutic approach, as well as their respective advantages and disadvantages, to provide better ideas for relieving the IVDD patients' pain through new treatment methods.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong UniversityXi’an 710054, Shaanxi, China
| | - Yicun Hu
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong UniversityXi’an 710054, Shaanxi, China
| | - Tao Li
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong UniversityXi’an 710054, Shaanxi, China
| | - Yuhan Jia
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong UniversityXi’an 710054, Shaanxi, China
| | - Wei Hu
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong UniversityXi’an 710054, Shaanxi, China
| | - Zhengwei Xu
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiao Tong UniversityXi’an 710054, Shaanxi, China
| |
Collapse
|
47
|
ECM-Mimicking Hydrogels Loaded with Bone Mesenchymal Stem Cell-Derived Exosomes for the Treatment of Cartilage Defects. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3450672. [PMID: 36387356 PMCID: PMC9649317 DOI: 10.1155/2022/3450672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022]
Abstract
It is well-established that treating articular cartilage injuries is clinically challenging since they lack blood arteries, nerves, and lymphoid tissue. Recent studies have revealed that bone marrow stem cell-derived exosomes (BMSCs-Exos) exert significant chondroprotective effects through paracrine secretions, and hydrogel-based materials can synergize the exosomes through sustained release. Therefore, this research aims to synthesize an ECM (extracellular matrix)-mimicking gelatin methacryloyl (GelMA) hydrogel modified by gelatin combined with BMSCs-derived exosomes to repair cartilage damage. We first isolated and characterized exosomes from BMSCs supernatant and then loaded the exosomes into GelMA hydrogel to investigate cartilage repair effects in in vitro and in vivo experiments. The outcomes showed that the GelMA hydrogel has good biocompatibility with a 3D (three-dimensional) porous structure, exhibiting good carrier characteristics for exosomes. Furthermore, BMSCs-Exos had a significant effect on promoting chondrocyte ECM production and chondrocyte proliferation, and the GelMA hydrogel could enhance this effect through a sustained-release effect. Similarly, in vivo experiments showed that GelMA-Exos promoted cartilage regeneration in rat joint defects and the synthesis of related cartilage matrix proteins.
Collapse
|
48
|
Wu Y, Li M, Su H, Chen H, Zhu Y. Up-to-date progress in bioprinting of bone tissue. Int J Bioprint 2022; 9:628. [PMID: 36636136 PMCID: PMC9830997 DOI: 10.18063/ijb.v9i1.628] [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: 04/15/2022] [Accepted: 07/20/2022] [Indexed: 11/05/2022] Open
Abstract
The major apparatuses used for three-dimensional (3D) bioprinting include extrusion-based, droplet-based, and laser-based bioprinting. Numerous studies have been proposed to fabricate bioactive 3D bone tissues using different bioprinting techniques. In addition to the development of bioinks and assessment of their printability for corresponding bioprinting processes, in vitro and in vivo success of the bioprinted constructs, such as their mechanical properties, cell viability, differentiation capability, immune responses, and osseointegration, have been explored. In this review, several major considerations, challenges, and potential strategies for bone bioprinting have been deliberated, including bioprinting apparatus, biomaterials, structure design of vascularized bone constructs, cell source, differentiation factors, mechanical properties and reinforcement, hypoxic environment, and dynamic culture. In addition, up-to-date progress in bone bioprinting is summarized in detail, which uncovers the immense potential of bioprinting in re-establishing the 3D dynamic microenvironment of the native bone. This review aims to assist the researchers to gain insights into the reconstruction of clinically relevant bone tissues with appropriate mechanical properties and precisely regulated biological behaviors.
Collapse
Affiliation(s)
- Yang Wu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, China,State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, China,Corresponding author: Yang Wu ()
| | - Ming Li
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, China
| | - Hao Su
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, China
| | - Huaying Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, China
| | - Yonggang Zhu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, China
| |
Collapse
|
49
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
50
|
Kageyama T, Akieda H, Sonoyama Y, Sato K, Yoshikawa H, Isono H, Hirota M, Kitajima H, Chun YS, Maruo S, Fukuda J. Bone Beads Enveloped with Vascular Endothelial Cells for Bone Regenerative Medicine. Acta Biomater 2022:S1742-7061(22)00520-7. [PMID: 36030051 DOI: 10.1016/j.actbio.2022.08.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/27/2022]
Abstract
The transplantation of pre-vascularized bone grafts is a promising strategy to improve the efficacy of engraftment and bone regeneration. We propose a hydrogel microbead-based approach for preparing vascularized and high-density tissue grafts. Mesenchymal stem cell-encapsulated collagen microgels (2 µL), termed bone beads, were prepared through spontaneous constriction, which improved the density of the mesenchymal stem cells and collagen molecules by more than 15-fold from the initial day of culture. Constriction was attributed to cell-attractive forces and involved better osteogenic differentiation of mesenchymal stem cells than that of spheroids. This approach was scalable, and ∼2,000 bone beads were prepared semi-automatically using a liquid dispenser and spinner flask. The mechanical stimuli in the spinner flask further improved the osteogenic differentiation of the mesenchymal stem cells in the bone beads compared with that in static culture. Vascular endothelial cells readily attach to and cover the surface of bone beads. The in vitro assembly of the endothelial cell-enveloped bone beads resulted in microchannel formation in the interspaces between the bone beads. Significant effects of endothelialization on in vivo bone regeneration were shown in rats with cranial bone defects. The use of endothelialized bone beads may be a scalable and robust approach for treating large bone defects. STATEMENT OF SIGNIFICANCE: A unique aspect of this study is that the hMSC-encapsulated collagen microgels were prepared through spontaneous constriction, leading to the enrichment of collagen and cell density. This constriction resulted in favorable microenvironments for the osteogenic differentiation of hMSCs, which is superior to conventional spheroid culture. The microgel beads were then enveloped with vascular endothelial cells and assembled to fabricate a tissue graft with vasculature in the interspaces among the beads. The significant effects of endothelialization on in vivo bone regeneration were clearly demonstrated in rats with cranial bone defects. We believe that microgel beads covered with vascular endothelial cells provide a promising approach for engineering better tissue grafts for bone-regenerative medicine.
Collapse
Affiliation(s)
- Tatsuto Kageyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa, 213-0012, JAPAN
| | - Hikaru Akieda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Yukie Sonoyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Ken Sato
- Department of Chemistry, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama City, Saitama 338-8570, JAPAN
| | - Hiroshi Yoshikawa
- Department of Chemistry, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama City, Saitama 338-8570, JAPAN
| | - Hitoshi Isono
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fuku-ura, Kanazawa-ku Yokohama, Kanagawa 236-0004, JAPAN
| | - Makoto Hirota
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Ura-fune, Minami-ku Yokohama, Kanagawa 232-0024, JAPAN
| | - Hiroaki Kitajima
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Ura-fune, Minami-ku Yokohama, Kanagawa 232-0024, JAPAN
| | - Yang-Sook Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 110-799, KOREA
| | - Shoji Maruo
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa, 213-0012, JAPAN.
| |
Collapse
|