51
|
Gaihre B, Potes MDA, Liu X, Tilton M, Camilleri E, Rezaei A, Serdiuk V, Park S, Lucien F, Terzic A, Lu L. Extrusion 3D-printing and characterization of poly(caprolactone fumarate) for bone regeneration applications. J Biomed Mater Res A 2024; 112:672-684. [PMID: 37971074 PMCID: PMC10948318 DOI: 10.1002/jbm.a.37646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Polycaprolactone fumarate (PCLF) is a cross-linkable PCL derivative extensively considered for tissue engineering applications. Although injection molding has been widely used to develop PCLF scaffolds, platforms developed using such technique lack precise control on architecture, design, and porosity required to ensure adequate cellular and tissue responses. In particular, the scaffolds should provide a suitable surface for cell attachment and proliferation, and facilitate cell-cell communication and nutrient flow. 3D printing technologies have led to new architype for biomaterial development with micro-architecture mimicking native tissue. Here, we developed a method for 3D printing of PCLF structures using the extrusion printing technique. The crosslinking property of PCLF enabled the unique post-processing of 3D printed scaffolds resulting in highly porous and flexible PCLF scaffolds with compressive properties imitating natural features of cancellous bone. Generated scaffolds supported excellent attachment and proliferation of mesenchymal stem cells (MSC). The high porosity of PCLF scaffolds facilitated vascularized membrane formation demonstrable with the stringency of the ex ovo chicken chorioallantoic membrane (CAM) implantation. Furthermore, upon implantation to rat calvarium defects, PCLF scaffolds enabled an exceptional new bone formation with a bone mineral density of newly formed bone mirroring native bone tissue. These studies suggest that the 3D-printed highly porous PCLF scaffolds may serve as a suitable biomaterial platform to significantly expand the utility of the PCLF biomaterial for bone tissue engineering applications.
Collapse
Affiliation(s)
- Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria D Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Emily Camilleri
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Asghar Rezaei
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Vitalii Serdiuk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Sungjo Park
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andre Terzic
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
52
|
Chrungoo S, Bharadwaj T, Verma D. Nanofibrous polyelectrolyte complex incorporated BSA-alginate composite bioink for 3D bioprinting of bone mimicking constructs. Int J Biol Macromol 2024; 266:131123. [PMID: 38537853 DOI: 10.1016/j.ijbiomac.2024.131123] [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: 09/26/2023] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Although several bioinks have been developed for 3D bioprinting applications, the lack of optimal printability, mechanical properties, and adequate cell response has limited their practical applicability. Therefore, this work reports the development of a composite bioink consisting of bovine serum albumin (BSA), alginate, and self-assembled nanofibrous polyelectrolyte complex aggregates of gelatin and chitosan (PEC-GC). The nanofibrous PEC-GC aggregates were prepared and incorporated into the bioink in varying concentrations (0 % to 3 %). The bioink samples were bioprinted and crosslinked post-printing by calcium chloride. The average nanofiber diameter of PEC-GC was 62 ± 15 nm. It was demonstrated that PEC-GC improves the printability and cellular adhesion of the developed bioink and modulates the swelling ratio, degradation rate, and mechanical properties of the fabricated scaffold. The in vitro results revealed that the bioink with 2 % PEC-GC had the best post-printing cell viability of the encapsulated MG63 osteosarcoma cells and well oragnized stress fibers, indicating enhanced cell adhesion. The cell viability was >90 %, as observed from the MTT assay. The composite bioink also showed osteogenic potential, as confirmed by the estimation of alkaline phosphatase activity and collagen synthesis assay. This study successfully fabricated a high-shape fidelity bioink with potential in bone tissue engineering.
Collapse
Affiliation(s)
- Shreya Chrungoo
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Tanmay Bharadwaj
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Devendra Verma
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
53
|
Nie R, Zhang QY, Feng ZY, Huang K, Zou CY, Fan MH, Zhang YQ, Zhang JY, Li-Ling J, Tan B, Xie HQ. Hydrogel-based immunoregulation of macrophages for tissue repair and regeneration. Int J Biol Macromol 2024; 268:131643. [PMID: 38643918 DOI: 10.1016/j.ijbiomac.2024.131643] [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: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
The rational design of hydrogel materials to modulate the immune microenvironment has emerged as a pivotal approach in expediting tissue repair and regeneration. Within the immune microenvironment, an array of immune cells exists, with macrophages gaining prominence in the field of tissue repair and regeneration due to their roles in cytokine regulation to promote regeneration, maintain tissue homeostasis, and facilitate repair. Macrophages can be categorized into two types: classically activated M1 (pro-inflammatory) and alternatively activated M2 (anti-inflammatory and pro-repair). By regulating the physical and chemical properties of hydrogels, the phenotypic transformation and cell behavior of macrophages can be effectively controlled, thereby promoting tissue regeneration and repair. A full understanding of the interaction between hydrogels and macrophages can provide new ideas and methods for future tissue engineering and clinical treatment. Therefore, this paper reviews the effects of hydrogel components, hardness, pore size, and surface morphology on cell behaviors such as macrophage proliferation, migration, and phenotypic polarization, and explores the application of hydrogels based on macrophage immune regulation in skin, bone, cartilage, and nerve tissue repair. Finally, the challenges and future prospects of macrophage-based immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zi-Yuan Feng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Kai Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Genetics, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Bo Tan
- Department of Orthopedic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, PR China.
| |
Collapse
|
54
|
Rai V, Munazzam SW, Wazir NU, Javaid I. Revolutionizing bone tumor management: cutting-edge breakthroughs in limb-saving treatments. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2024; 34:1741-1748. [PMID: 38461457 DOI: 10.1007/s00590-024-03876-z] [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: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024]
Abstract
Limb salvage surgery has revolutionized the approach to bone tumors in orthopedic oncology, steering away from historical amputations toward preserving limb function and enhancing patient quality of life. This transformative shift underscores the delicate balance between tumor eradication and optimal postoperative function. Primary and metastatic bone tumors present challenges in early detection, differentiation between benign and malignant tumors, preservation of function, and the risk of local recurrence. Conventional methods, including surgery, radiation therapy, chemotherapy, and targeted therapies, have evolved with a heightened focus on personalized medicine. A groundbreaking development in limb salvage surgery is the advent of 3D-printed patient-specific implants, which significantly enhance anatomical precision, stability, and fixation. These implants reduce soft tissue disruption and the associated risks, fostering improved osseointegration and correction of deformities for a more natural and functional postoperative outcome. Biological and molecular research has reshaped the understanding of bone tumors, guiding surgical interventions with advancements such as genomic profiling, targeted intraoperative imaging, precision targeting of molecular pathways, and immunotherapy tailored to individual tumor characteristics. In the realm of imaging technologies, MRI, CT scans, and intraoperative navigation systems have redefined preoperative planning, minimizing collateral damage and optimizing outcomes through accurate resections. Postoperative rehabilitation plays a crucial role in restoring function and improving the quality of life. Emphasizing early mobilization, effective pain management, and a multidisciplinary approach, rehabilitation addresses the physical, psychological, and social aspects of recovery. Looking ahead, future developments may encompass advanced biomaterials, smart implants, AI algorithms, robotics, and regenerative medicine. Challenges lie in standardization, cost-effectiveness, accessibility, long-term outcome assessment, mental health support, and fostering global collaboration. As research progresses, limb salvage surgery emerges not just as a preservation tool but as a transformative approach, restoring functionality, resilience, and hope in the recovery journey. This review summarizes the recent advances in limb salvage therapy for bone tumors over the past decade.
Collapse
Affiliation(s)
- Vikramaditya Rai
- Department of Orthopedics, Dr. Rajendra Prasad Government Medical College and Hospital, Himachal Pradesh, Tanda at Kangra, India.
| | | | | | - Irum Javaid
- Khyber Medical College, Hayatabad, Peshawar, Pakistan
| |
Collapse
|
55
|
Alonso-Fernández I, Haugen HJ, Nogueira LP, López-Álvarez M, González P, López-Peña M, González-Cantalapiedra A, Muñoz-Guzón F. Enhanced Bone Healing in Critical-Sized Rabbit Femoral Defects: Impact of Helical and Alternate Scaffold Architectures. Polymers (Basel) 2024; 16:1243. [PMID: 38732711 PMCID: PMC11085737 DOI: 10.3390/polym16091243] [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: 03/11/2024] [Revised: 04/20/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
This study investigates the effect of scaffold architecture on bone regeneration, focusing on 3D-printed polylactic acid-bioceramic calcium phosphate (PLA-bioCaP) composite scaffolds in rabbit femoral condyle critical defects. We explored two distinct scaffold designs to assess their influence on bone healing and scaffold performance. Structures with alternate (0°/90°) and helical (0°/45°/90°/135°/180°) laydown patterns were manufactured with a 3D printer using a fused deposition modeling technique. The scaffolds were meticulously characterized for pore size, strut thickness, porosity, pore accessibility, and mechanical properties. The in vivo efficacy of these scaffolds was evaluated using a femoral condyle critical defect model in eight skeletally mature New Zealand White rabbits. Then, the results were analyzed micro-tomographically, histologically, and histomorphometrically. Our findings indicate that both scaffold architectures are biocompatible and support bone formation. The helical scaffolds, characterized by larger pore sizes and higher porosity, demonstrated significantly greater bone regeneration than the alternate structures. However, their lower mechanical strength presented limitations for use in load-bearing sites.
Collapse
Affiliation(s)
- Iván Alonso-Fernández
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway; (H.J.H.); (L.P.N.)
| | - Liebert Parreiras Nogueira
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway; (H.J.H.); (L.P.N.)
| | - Miriam López-Álvarez
- Centro de Investigación en Tecnologías, Energía y Procesos Industriales (CINTECX), Universidade de Vigo, Grupo de Novos Materiais, 36310 Vigo, Spain; (M.L.-Á.); (P.G.)
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Pío González
- Centro de Investigación en Tecnologías, Energía y Procesos Industriales (CINTECX), Universidade de Vigo, Grupo de Novos Materiais, 36310 Vigo, Spain; (M.L.-Á.); (P.G.)
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Mónica López-Peña
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| | - Antonio González-Cantalapiedra
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| | - Fernando Muñoz-Guzón
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; (M.L.-P.); (A.G.-C.); (F.M.-G.)
| |
Collapse
|
56
|
Rani KGA, Al-Rawi AM, Al Qabbani A, AlKawas S, Mohammad MG, Samsudin AR. Response of human peripheral blood monocyte-derived macrophages (PBMM) to demineralized and decellularized bovine bone graft substitutes. PLoS One 2024; 19:e0300331. [PMID: 38635511 PMCID: PMC11025794 DOI: 10.1371/journal.pone.0300331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/26/2024] [Indexed: 04/20/2024] Open
Abstract
The performance of apparently biocompatible implanted bovine bone grafts may be compromised by unresolved chronic inflammation, and poor graft incorporation leading to implant failure. Monitoring the intensity and duration of the inflammatory response caused by implanted bone grafts is crucial. In this study, the ability of demineralized (DMB) and decellularized (DCC) bovine bone substitutes in initiating inflammatory responses to peripheral blood monocyte-derived macrophages (PBMMs) was investigated. The response of PBMMs to bone substitutes was evaluated by using both direct and indirect cell culture, reactive oxygen species (ROS) generation, apoptosis, immunophenotyping, and cytokine production. Analysis of DMB and DCC substitutes using scanning electron microscope (SEM) showed a roughened surface with a size ranging between 500 and 750 μm. PBMMs treated with DMB demonstrated cell aggregation and clumping mimicking lipopolysaccharide (LPS) treated PBMMs and a higher proliferation ability (166.93%) compared to control (100%) and DCC treatments (115.64%; p<0.001) at 24h. This was associated with a significantly increased production of intracellular ROS in PBMMs exposed to DMB substitutes than control (3158.5 vs 1715.5; p<0.001) and DCC treatment (2117.5). The bone substitute exposure also caused an increase in percentage apoptosis which was significantly (p<0.0001) higher in both DMB (27.85) and DCC (29.2) treatment than control (19.383). A significant increase in proinflammatory cytokine expression (TNF-α: 3.4 folds; p<0.05) was observed in DMB substitute-treated PBMMs compared to control. Notably, IL-1β mRNA was significantly higher in DMB (21.75 folds; p<0.0001) than control and DCC (5.01 folds). In contrast, DCC substitutes exhibited immunoregulatory effects on PBMMs, as indicated by the expression for CD86, CD206, and HLDR surface markers mimicking IL-4 treatments. In conclusion, DMB excites a higher immunological response compared to DCC suggesting decellularization process of tissues dampen down inflammatory reactions when exposed to PBMM.
Collapse
Affiliation(s)
- K. G. Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahmed M. Al-Rawi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ali Al Qabbani
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Sausan AlKawas
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammad G. Mohammad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - A. R. Samsudin
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
57
|
Zhou P, Liu T, Liu W, Sun L, Kang H, Liu K, Luo P, Wang Y, Luo L, Dai H. An Antibacterial Bionic Periosteum with Angiogenesis-Neurogenesis Coupling Effect for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38623938 DOI: 10.1021/acsami.4c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The periosteum, rich in neurovascular networks, bone progenitor cells, and stem cells, is vital for bone repair. Current artificial periosteal materials face challenges in mechanical strength, bacterial infection, and promoting osteogenic differentiation and angiogenesis. To address these issues, we adjusted the electrospinning ratio of poly-ε-caprolactone and chitosan and incorporated Zn doping whitlockite with polydopamine coating into a nanofiber membrane. After a series of characterizations, optimal results were achieved with a poly-ε-caprolactone: chitosan ratio of 8:1 and 5% nanoparticle content. In vitro cell experiments and in vivo calvarial defect models, the sustained release of Mg2+ and Ca2+ promoted vascularization and new bone formation, respectively, while the release of Zn2+ was conducive to antibacterial and cooperated with Mg2+ to promote neurovascularization. Consequently, this antibacterial bionic periosteum with an angiogenesis-neurogenesis coupling effect demonstrates a promising potential for bone repair applications.
Collapse
Affiliation(s)
- Peiqian Zhou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Tuozhou Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 511442, China
| | - Wenbin Liu
- Department of Orthopaedics, The Third Xiangya Hospital Central South University, 138 Tongzipo Road, Changsha, Hunan 410008, China
| | - Lingshun Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Haifei Kang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Kun Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Peiyuan Luo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Youfa Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Ling Luo
- Department of Orthopaedics, The Third Xiangya Hospital Central South University, 138 Tongzipo Road, Changsha, Hunan 410008, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
- National Energy Key Laboratory for New Hydrogen-Ammonia Energy Technologies, Foshan Xianhu Laboratory, Foshan 528200, China
| |
Collapse
|
58
|
Iravani S, Nazarzadeh Zare E, Makvandi P. Multifunctional MXene-Based Platforms for Soft and Bone Tissue Regeneration and Engineering. ACS Biomater Sci Eng 2024; 10:1892-1909. [PMID: 38466909 DOI: 10.1021/acsbiomaterials.3c01770] [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: 03/13/2024]
Abstract
MXenes and their composites hold great promise in the field of soft and bone tissue regeneration and engineering (TRE). However, there are challenges that need to be overcome, such as ensuring biocompatibility and controlling the morphologies of MXene-based scaffolds. The future prospects of MXenes in TRE include enhancing biocompatibility through surface modifications, developing multifunctional constructs, and conducting in vivo studies for clinical translation. The purpose of this perspective about MXenes and their composites in soft and bone TRE is to critically evaluate their potential applications and contributions in this field. This perspective aims to provide a comprehensive analysis of the challenges, advantages, limitations, and future prospects associated with the use of MXenes and their composites for soft and bone TRE. By examining the existing literature and research, the review seeks to consolidate the current knowledge and highlight the key findings and advancements in MXene-based TRE. It aims to contribute to the understanding of MXenes' role in promoting soft and bone TRE, addressing the challenges faced in terms of biocompatibility, morphology control, and tissue interactions.
Collapse
Affiliation(s)
- Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Avenue, Isfahan 81756-33551, Iran
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan 36716-45667, Iran
- Centre of Research Impact and Outreach, Chitkara University, Rajpura 140417, Punjab, India
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, Zhejiang, China
- Chitkara Centre for Research and Development, Chitkara University, Kalujhanda 174103, Himachal Pradesh, India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| |
Collapse
|
59
|
Makurat-Kasprolewicz B, Wekwejt M, Ronowska A, Gajowiec G, Grodzicka M, Dzionk S, Ossowska A. Influence of Ultrasound on the Characteristics of CaP Coatings Generated Via the Micro-arc Oxidation Process in Relation to Biomedical Engineering. ACS Biomater Sci Eng 2024; 10:2100-2115. [PMID: 38502729 PMCID: PMC11005015 DOI: 10.1021/acsbiomaterials.3c01433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/10/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
Over the past decade, bone tissue engineering has been at the core of attention because of an increasing number of implant surgeries. The purpose of this study was to obtain coatings on titanium (Ti) implants with improved properties in terms of biomedical applications and to investigate the effect of ultrasound (US) on these properties during the micro-arc oxidation (MAO) process. The influence of various process parameters, such as time and current density, as well as US mode, on the properties of such coatings was evaluated. Novel porous calcium-phosphate-based coatings were obtained on commercially pure Ti. Their microstructure, chemical composition, topography, wettability, nanomechanical properties, thickness, adhesion to the substrate, and corrosion resistance were analyzed. In addition, cytocompatibility evaluation was checked with the human osteoblasts. The properties of the coatings varied significantly, depending on applied process parameters. The US application during the MAO process contributes to the increase of coating thickness, porosity, roughness, and skewness, as well as augmented calcium incorporation. The most advantageous coating was obtained at a current of 136 mA, time 450 s, and unipolar rectangular US, as it exhibits high porosity, adequate wettability, and beneficial skewness, which enabled increased adhesion and proliferation of osteoblasts during in vitro studies. Finally, the conducted research demonstrated the influence of various UMAO process parameters, which allowed for the selection of appropriate Ti implant modification for specific biomedical utilization.
Collapse
Affiliation(s)
| | - Marcin Wekwejt
- Department
of Biomaterials Technology, Gdansk University
of Technology, 80-233 Gdańsk, Poland
| | - Anna Ronowska
- Department
of Laboratory Medicine, Medical University
of Gdańsk, 80-210 Gdańsk, Poland
| | - Grzegorz Gajowiec
- Department
of Materials Science and Technology, Gdansk
University of Technology, 80-233 Gdańsk, Poland
| | - Marlena Grodzicka
- Faculty
of Chemistry, Nicolaus Copernicus University
in Toruń, 87-100 Toruń, Poland
| | - Stefan Dzionk
- Department
of Manufacturing and Production Engineering, Gdansk University of Technology, 80-233 Gdańsk, Poland
| | - Agnieszka Ossowska
- Department
of Materials Science and Technology, Gdansk
University of Technology, 80-233 Gdańsk, Poland
| |
Collapse
|
60
|
Bauso LV, La Fauci V, Longo C, Calabrese G. Bone Tissue Engineering and Nanotechnology: A Promising Combination for Bone Regeneration. BIOLOGY 2024; 13:237. [PMID: 38666849 PMCID: PMC11048357 DOI: 10.3390/biology13040237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024]
Abstract
Large bone defects are the leading contributor to disability worldwide, affecting approximately 1.71 billion people. Conventional bone graft treatments show several disadvantages that negatively impact their therapeutic outcomes and limit their clinical practice. Therefore, much effort has been made to devise new and more effective approaches. In this context, bone tissue engineering (BTE), involving the use of biomaterials which are able to mimic the natural architecture of bone, has emerged as a key strategy for the regeneration of large defects. However, although different types of biomaterials for bone regeneration have been developed and investigated, to date, none of them has been able to completely fulfill the requirements of an ideal implantable material. In this context, in recent years, the field of nanotechnology and the application of nanomaterials to regenerative medicine have gained significant attention from researchers. Nanotechnology has revolutionized the BTE field due to the possibility of generating nanoengineered particles that are able to overcome the current limitations in regenerative strategies, including reduced cell proliferation and differentiation, the inadequate mechanical strength of biomaterials, and poor production of extrinsic factors which are necessary for efficient osteogenesis. In this review, we report on the latest in vitro and in vivo studies on the impact of nanotechnology in the field of BTE, focusing on the effects of nanoparticles on the properties of cells and the use of biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Luana Vittoria Bauso
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (V.L.F.); (C.L.)
| | | | | | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (V.L.F.); (C.L.)
| |
Collapse
|
61
|
Soleymani Eil Bakhtiari S, Karbasi S. Keratin-containing scaffolds for tissue engineering applications: a review. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:916-965. [PMID: 38349200 DOI: 10.1080/09205063.2024.2311450] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/24/2024] [Indexed: 04/13/2024]
Abstract
In tissue engineering and regenerative medicine applications, the utilization of bioactive materials has become a routine tool. The goal of tissue engineering is to create new organs and tissues by combining cell biology, materials science, reactor engineering, and clinical research. As part of the growth pattern for primary cells in an organ, backing material is frequently used as a supporting material. A porous three-dimensional (3D) scaffold can provide cells with optimal conditions for proliferating, migrating, differentiating, and functioning as a framework. Optimizing the scaffolds' structure and altering their surface may improve cell adhesion and proliferation. A keratin-based biomaterials platform has been developed as a result of discoveries made over the past century in the extraction, purification, and characterization of keratin proteins from hair and wool fibers. Biocompatibility, biodegradability, intrinsic biological activity, and cellular binding motifs make keratin an attractive biomaterial for tissue engineering scaffolds. Scaffolds for tissue engineering have been developed from extracted keratin proteins because of their capacity to self-assemble and polymerize into intricate 3D structures. In this review article, applications of keratin-based scaffolds in different tissues including bone, skin, nerve, and vascular are explained based on common methods of fabrication such as electrospinning, freeze-drying process, and sponge replication method.
Collapse
Affiliation(s)
- Sanaz Soleymani Eil Bakhtiari
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Saeed Karbasi
- Biomaterials and Tissue Engineering Department, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
62
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
63
|
Chen C, Li Z, Xu C, Kang M, Lee CS, Aghaloo T, Lee M. Self-Assembled Nanocomposite Hydrogels as Carriers for Demineralized Bone Matrix Particles and Enhanced Bone Repair. Adv Healthc Mater 2024; 13:e2303592. [PMID: 38275216 PMCID: PMC11023793 DOI: 10.1002/adhm.202303592] [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: 10/18/2023] [Revised: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Demineralized bone matrix (DBM) has been widely used as an allogeneic alternative to autologous bone graft for bone repair. However, more extensive use of DBM is limited due to its particulate nature after demineralization and rapid particle dispersion following irrigation, resulting in unpredictable osteoinductivity. Here, a new design of injectable hydrogel carriers for DBM that combine self-healing ability and osteogenic properties based on the self-assembly of guanidinylated hyaluronic acid and silica-rich nanoclays is reported. The nanoclays serve as reversible linkages to form a dynamic hydrogel network with the guanidine moieties on the polymer chains. Gelation kinetics and mechanical properties can be controlled by altering nanoclay content in the hydrogel. The resulting hydrogel exerts self-healing ability due to its dynamic crosslinks and well retains its overall performance with high DBM loading. The hydrogel exhibits great cytocompatibility and osteogenic effects mediated by the nanoclays. In vivo delivery of DBM using the nanocomposite hydrogel further demonstrates robust bone regeneration in a mouse calvarial defect model in comparison to DBM delivered with aqueous HA. This work suggests a promising hydrogel platform for many applications including therapeutic delivery and tissue engineering.
Collapse
Affiliation(s)
- Chen Chen
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Zhi Li
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Changlu Xu
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Minjee Kang
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Chung-Sung Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Min Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
64
|
Addepalli P, Sawangsri W, Ghani SAC. A scientometric analysis of bone cutting tools & methodologies: Mapping the research landscape. Injury 2024; 55:111458. [PMID: 38432100 DOI: 10.1016/j.injury.2024.111458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 02/15/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
This study undertakes a Scientometric analysis of bone-cutting tools, investigating a corpus of 735 papers from the Scopus database between 1941 and 2023. It employs bibliometric methodologies such as keyword coupling, co-citation, and co-authorship analysis to map the intellectual landscape and collaborative networks within this research domain. The analysis highlights a growing interest and significant advancements in bone-cutting tools, focusing on their design, the materials used, and the cutting processes involved. It identifies key research fronts and trends, such as the emphasis on surgical precision, material innovation, and the optimization of tool performance. Further, the study reveals a broad collaboration among researchers from various disciplines, including engineering, materials science, and medical sciences, reflecting the field's interdisciplinary nature. Despite the progress, the analysis points out several gaps, notably in tool design optimization and the impact of materials on bone health. This comprehensive review not only charts the evolution of bone-cutting tool research but also calls attention to areas requiring further investigation, aiming to inspire future studies that address these identified gaps and enhance surgical outcomes.
Collapse
|
65
|
Ferraz MP. An Overview on the Big Players in Bone Tissue Engineering: Biomaterials, Scaffolds and Cells. Int J Mol Sci 2024; 25:3836. [PMID: 38612646 PMCID: PMC11012232 DOI: 10.3390/ijms25073836] [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: 02/20/2024] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Presently, millions worldwide suffer from degenerative and inflammatory bone and joint issues, comprising roughly half of chronic ailments in those over 50, leading to prolonged discomfort and physical limitations. These conditions become more prevalent with age and lifestyle factors, escalating due to the growing elderly populace. Addressing these challenges often entails surgical interventions utilizing implants or bone grafts, though these treatments may entail complications such as pain and tissue death at donor sites for grafts, along with immune rejection. To surmount these challenges, tissue engineering has emerged as a promising avenue for bone injury repair and reconstruction. It involves the use of different biomaterials and the development of three-dimensional porous matrices and scaffolds, alongside osteoprogenitor cells and growth factors to stimulate natural tissue regeneration. This review compiles methodologies that can be used to develop biomaterials that are important in bone tissue replacement and regeneration. Biomaterials for orthopedic implants, several scaffold types and production methods, as well as techniques to assess biomaterials' suitability for human use-both in laboratory settings and within living organisms-are discussed. Even though researchers have had some success, there is still room for improvements in their processing techniques, especially the ones that make scaffolds mechanically stronger without weakening their biological characteristics. Bone tissue engineering is therefore a promising area due to the rise in bone-related injuries.
Collapse
Affiliation(s)
- Maria Pia Ferraz
- Departamento de Engenharia Metalúrgica e de Materiais, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal;
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4099-002 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4099-002 Porto, Portugal
| |
Collapse
|
66
|
Park SS, Farwa U, Kim HD, Kim YS, Lee BT. Bone formation by Irisin-Poly vinyl alchol modified bioglass ceramic beads in the rabbit model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:23. [PMID: 38526676 DOI: 10.1007/s10856-024-06788-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/01/2024] [Indexed: 03/27/2024]
Abstract
In the aging society, slow bone regeneration poses a serious hindrance to the quality of life. To deal with this problem, in this study, we have combined irisin with the bioglass regular beads to enhance the bone regeneration process. For this purpose, highly porous bioglass was obtained as spherical beads by using sodium alginate. The bioglass was evaluated by various analytical techniques such as SEM, EDS, XRD, and pore size distribution. The results depicted that porous bioglass was prepared correctly and SEM analysis showed a highly porous bioglass was formulated. On this bioglass, irisin was loaded with the assistance of polyvinyl alcohol (PVA) in three concentrations (50 ng/ml, 100 ng/ml, and 150 ng/ml per 1 g of bioglass). SEM analysis showed that pores are covered with PVA. The irisin release profile showed a sustained release over the time period of 7 days. In vitro, biocompatibility evaluation by the MC3T3E1 cells showed that prepared bioglass and irisin loaded bioglass (BGI50, BGI100, and BG150) are highly biocompatible. Alizarin Red staining analysis showed that after 2 weeks BGI50 samples showed highest calcium nodule formation. In vivo in the rabbit femur model was conducted for 1 and 2 months. BGI150 samples showed highest BV/TV ratio of 37.1 after 2 months. The histological data showed new bone formation surrounding the beads and with beads loaded with irisin. Immunohistochemistry using markers OPN, RUNX, COL, and ALP supported the osteogenic properties of the irisin-loaded bioglass beads. The results indicated that irisin-loaded bioglass displayed remarkable bone regeneration.
Collapse
Affiliation(s)
- Seong-Su Park
- Department of regenerative medicine, College of Medicine, Soonchunhyang University Cheonan, Cheonan, Republic of Korea
| | - Ume Farwa
- Institute of tissue regeneration, Soonchunhyang University Cheonan, Cheonan, Republic of Korea
| | - Hai-Doo Kim
- Institute of tissue regeneration, Soonchunhyang University Cheonan, Cheonan, Republic of Korea
| | - Yong-Sik Kim
- Institute of tissue regeneration, Soonchunhyang University Cheonan, Cheonan, Republic of Korea
- Department of Microbiology, College of Medicine, Soonchunhyang University Cheonan, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of regenerative medicine, College of Medicine, Soonchunhyang University Cheonan, Cheonan, Republic of Korea.
- Institute of tissue regeneration, Soonchunhyang University Cheonan, Cheonan, Republic of Korea.
| |
Collapse
|
67
|
Bhatia A, Hanna J, Stuart T, Kasper KA, Clausen DM, Gutruf P. Wireless Battery-free and Fully Implantable Organ Interfaces. Chem Rev 2024; 124:2205-2280. [PMID: 38382030 DOI: 10.1021/acs.chemrev.3c00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Advances in soft materials, miniaturized electronics, sensors, stimulators, radios, and battery-free power supplies are resulting in a new generation of fully implantable organ interfaces that leverage volumetric reduction and soft mechanics by eliminating electrochemical power storage. This device class offers the ability to provide high-fidelity readouts of physiological processes, enables stimulation, and allows control over organs to realize new therapeutic and diagnostic paradigms. Driven by seamless integration with connected infrastructure, these devices enable personalized digital medicine. Key to advances are carefully designed material, electrophysical, electrochemical, and electromagnetic systems that form implantables with mechanical properties closely matched to the target organ to deliver functionality that supports high-fidelity sensors and stimulators. The elimination of electrochemical power supplies enables control over device operation, anywhere from acute, to lifetimes matching the target subject with physical dimensions that supports imperceptible operation. This review provides a comprehensive overview of the basic building blocks of battery-free organ interfaces and related topics such as implantation, delivery, sterilization, and user acceptance. State of the art examples categorized by organ system and an outlook of interconnection and advanced strategies for computation leveraging the consistent power influx to elevate functionality of this device class over current battery-powered strategies is highlighted.
Collapse
Affiliation(s)
- Aman Bhatia
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jessica Hanna
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Tucker Stuart
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Kevin Albert Kasper
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - David Marshall Clausen
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
| | - Philipp Gutruf
- Department of Biomedical Engineering, The University of Arizona, Tucson, Arizona 85721, United States
- Department of Electrical and Computer Engineering, The University of Arizona, Tucson, Arizona 85721, United States
- Bio5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
- Neuroscience Graduate Interdisciplinary Program (GIDP), The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
68
|
Kumar A, Brown RA, Roufaeil DB, Gupta A, Lipford EL, Muthusamy D, Zalzman A, Hertzano R, Lowe T, Stains JP, Zalzman M. DeepFreeze 3D-biofabrication for Bioengineering and Storage of Stem Cells in Thick and Large-Scale Human Tissue Analogs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306683. [PMID: 38183347 PMCID: PMC10953591 DOI: 10.1002/advs.202306683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 01/08/2024]
Abstract
3D bioprinting holds great promise for meeting the increasing need for transplantable tissues and organs. However, slow printing, interlayer mixing, and the extended exposure of cells to non-physiological conditions in thick structures still hinder clinical applications. Here the DeepFreeze-3D (DF-3D) procedure and bioink for creating multilayered human-scale tissue mimetics is presented for the first time. The bioink is tailored to support stem cell viability, throughout the rapid freeform DF-3D biofabrication process. While the printer nozzle is warmed to room temperature, each layer solidifies at contact with the stage (-80 °C), or the subsequent layers, ensuring precise separation. After thawing, the encapsulated stem cells remain viable without interlayer mixing or delamination. The composed cell-laden constructs can be cryogenically stored and thawed when needed. Moreover, it is shown that under inductive conditions the stem cells differentiate into bone-like cells and grow for months after thawing, to form large tissue-mimetics in the scale of centimeters. This is important, as this approach allows the generation and storage of tissue mimetics in the size and thickness of human tissues. Therefore, DF-3D biofabrication opens new avenues for generating off-the-shelf human tissue analogs. It further holds the potential for regenerative treatments and for studying tissue pathologies caused by disease, tumor, or trauma.
Collapse
Affiliation(s)
- Alok Kumar
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Cardiovascular Research CenterMassachusetts General Hospital (MGH)Harvard Medical SchoolBostonMA02114USA
| | - Robert A. Brown
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Daniel Benyamien Roufaeil
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Aditi Gupta
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Neurotology BranchNIDCD, NIHBethesdaMarylandUnited States
| | - Erika L. Lipford
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Divya Muthusamy
- Department of Oral and Maxillofacial SurgeryUniversity of Maryland School of DentistryBaltimoreMD21201USA
- Fischell Department of BioengineeringUniversity of Maryland A. James Clark School of EngineeringCollege ParkMD20742USA
| | - Amihai Zalzman
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Ronna Hertzano
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Neurotology BranchNIDCD, NIHBethesdaMarylandUnited States
| | - Tao Lowe
- Department of Oral and Maxillofacial SurgeryUniversity of Maryland School of DentistryBaltimoreMD21201USA
- Fischell Department of BioengineeringUniversity of Maryland A. James Clark School of EngineeringCollege ParkMD20742USA
| | - Joseph P. Stains
- Department of OrthopedicsUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Michal Zalzman
- Department of Biochemistry and Molecular BiologyDepartment of Otorhinolaryngology‐Head and Neck SurgeryMarlene and Stewart Greenbaum Cancer CenterThe Center for Stem Cell Biology and Regenerative MedicineUniversity of Maryland School of MedicineBaltimoreMD21201USA
| |
Collapse
|
69
|
Ranjbar FE, Ranjbar AE, Malekshahi ZV, Taghdiri-Nooshabadi Z, Faradonbeh DR, Youseflee P, Ghasemi S, Vatanparast M, Azim F, Nooshabadi VT. Bone tissue regeneration by 58S bioactive glass scaffolds containing exosome: an in vivo study. Cell Tissue Bank 2024; 25:389-400. [PMID: 38159136 DOI: 10.1007/s10561-023-10120-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024]
Abstract
Exosomes, the naturally secreted nanocarriers of cells, have recently been demonstrated to have therapeutic benefits in a variety of disease models where parent cells are not present. However, the use of exosomes in bone defect regeneration has been unusual, and little is documented about the underlying processes. In recent study we produced and characterized exosomes derived human endometrial mesenchymal stem stromal cells and 58S bioactive glass scaffolds; in following, in this research exosome loaded scaffolds synthetized and release of exosome, porosity and bioactivity of them were assessed. More over the effect of scaffolds on repair of critical-size bone defects in rat's calvaria was evaluated by histological examination and micro computed tomography (µ CT). The findings confirmed that constructed porous scaffolds consistently release exosomes; additionally, in vivo findings including Hematoxilin & Eosin staining, Immunohistochemistry, Masson's trichrome, histomorphometric analysis, and µ CT clarified that our implant has osteogenic properties. We discovered that Exo-treated scaffolds might promote osteogenesis especially compared to pure scaffolds, indicating that produced scaffolds containing exosomes could be a potential replacement in bone tissue engineering.
Collapse
Affiliation(s)
- Faezeh Esmaeili Ranjbar
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Afsaneh Esmaeili Ranjbar
- Emergency Department, Ali Ebn Abitaleb Hospital, Faculty of medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Davood Rabiei Faradonbeh
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouya Youseflee
- Medical student, Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sahar Ghasemi
- Medical student, Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahboubeh Vatanparast
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fazli Azim
- Isolation Hospital & Infections Treatment Center (IHITC), MNHSR&C, Islamabad, Pakistan
| | | |
Collapse
|
70
|
Maita KC, Avila FR, Torres-Guzman RA, Sarabia-Estrada R, Zubair AC, Quinones-Hinojosa A, Forte AJ. In Vitro Enhanced Osteogenic Potential of Human Mesenchymal Stem Cells Seeded in a Poly (Lactic- co-Glycolic) Acid Scaffold: A Systematic Review. Craniomaxillofac Trauma Reconstr 2024; 17:61-73. [PMID: 38371215 PMCID: PMC10874209 DOI: 10.1177/19433875231157454] [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] [Indexed: 02/20/2024] Open
Abstract
Study Design Human bone marrow stem cells (hBMSCs) and human adipose-derived stem cells (hADSCs) have demonstrated the capability to regenerate bone once they have differentiated into osteoblasts. Objective This systematic review aimed to evaluate the in vitro osteogenic differentiation potential of these cells when seeded in a poly (lactic-co-glycolic) acid (PLGA) scaffold. Methods A literature search of 4 databases following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines was conducted in January 2021 for studies evaluating the osteogenic differentiation potential of hBMSCs and hADSCs seeded in a PLGA scaffold. Only in vitro models were included. Studies in languages other than English were excluded. Results A total of 257 studies were identified after the removal of duplicates. Seven articles fulfilled our inclusion and exclusion criteria. Four of these reviews used hADSCs and three used hBMSCs in the scaffold. Upregulation in osteogenic gene expression was seen in all the cells seeded in a 3-dimensional scaffold compared with 2-dimensional films. High angiogenic gene expression was found in hADSCs. Addition of inorganic material to the scaffold material affected cell performance. Conclusions Viability, proliferation, and differentiation of cells strongly depend on the environment where they grow. There are several factors that can enhance the differentiation capacity of stem cells. A PLGA scaffold proved to be a biocompatible material capable of boosting the osteogenic differentiation potential and mineralization capacity in hBMSCs and hADSCs.
Collapse
Affiliation(s)
- Karla C. Maita
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | - Abba C. Zubair
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | | | | |
Collapse
|
71
|
Barpour N, Ghorbani M, Baradaran B, Jodari-Mohammadpour Z, Nejati-Koshki K, Abdollahpour-Alitappeh M, Dabbaghi R, Gharibi T. Development of an injectable chitosan-based hydrogel containing nano-hydroxy-apatite and alendronate for MSC-based therapy. Int J Biol Macromol 2024; 261:129737. [PMID: 38286373 DOI: 10.1016/j.ijbiomac.2024.129737] [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: 05/14/2023] [Revised: 01/06/2024] [Accepted: 01/23/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND The combination of cells and biomaterials has become a powerful approach to regenerative medicine in recent years. Understanding the in-vitro interactions between cells and biomaterials is crucial for the success of regenerative medicine. AIM In this study, we developed an AD-pectin/chitosan/nano-crystalline cellulose scaffold with nano-hydroxy-apatite (n-HAP) and alendronate (ALN). The second step was to evaluate its effect on the immunomodulatory properties and biological behaviors of seeded adipose-derived mesenchymal stem cells (ADSCs) for bone tissue repair. MATERIAL AND METHOD After preparing and evaluating the characterization tests of the new combined n-HAP scaffold, we established different culture conditions to evaluate ADSC growth on this scaffold with or without ALN. The main assays were MTT assay, RT-PCR, and ELISA. RESULTS Our data regarding characterization tests (including SEM, TGA, FTIR, gelation time, swelling ratio, rheology and degradation tests) of ALN-loaded n-HAP scaffold showed the proper stability and good mechanical status of the scaffold. ADSC proliferation and viability increased in the presence of the scaffold compared with other conditions. Moreover, our data demonstrated increased gene expression and protein levels of anti-inflammatory TGF-β, HGF, and IDO cytokines in the presence of the ALN-loaded n-HAP scaffold, indicating the increased immunosuppressive activity of ADSCs in vitro. CONCLUSION This study demonstrates the promising abilities of the ALN-loaded n-HAP scaffold to increase the proliferation, viability, and immunomodulatory capacity of ADSCs, elucidating new aspects of cell-material interactions that can be used for bone tissue regeneration/repair, and paving the path of future research in developing new approaches for MSC- based therapy.
Collapse
Affiliation(s)
- Nesa Barpour
- Department of Genetics, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Ghorbani
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Kazem Nejati-Koshki
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Rozhin Dabbaghi
- Department of Genetics, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
72
|
Valamvanos TF, Dereka X, Katifelis H, Gazouli M, Lagopati N. Recent Advances in Scaffolds for Guided Bone Regeneration. Biomimetics (Basel) 2024; 9:153. [PMID: 38534838 DOI: 10.3390/biomimetics9030153] [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: 02/04/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
The rehabilitation of alveolar bone defects of moderate to severe size is often challenging. Currently, the therapeutic approaches used include, among others, the guided bone regeneration technique combined with various bone grafts. Although these techniques are widely applied, several limitations and complications have been reported such as morbidity, suboptimal graft/membrane resorption rate, low structural integrity, and dimensional stability. Thus, the development of biomimetic scaffolds with tailor-made characteristics that can modulate cell and tissue interaction may be a promising tool. This article presents a critical consideration in scaffold's design and development while also providing information on various fabrication methods of these nanosystems. Their utilization as delivery systems will also be mentioned.
Collapse
Affiliation(s)
- Theodoros-Filippos Valamvanos
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Xanthippi Dereka
- Department of Periodontology, School of Dentistry, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Hector Katifelis
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- School of Science and Technology, Hellenic Open University, 26335 Patra, Greece
| | - Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Greece Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
73
|
He F, Wang L, Umrath F, Naros A, Reinert S, Alexander D. Three-Dimensionally Cultured Jaw Periosteal Cells Attenuate Macrophage Activation of CD4 + T Cells and Inhibit Osteoclastogenesis. Int J Mol Sci 2024; 25:2355. [PMID: 38397031 PMCID: PMC10889513 DOI: 10.3390/ijms25042355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The implementation of a successful therapeutic approach that includes tissue-engineered grafts requires detailed analyses of graft-immune cell interactions in order to predict possible immune reactions after implantation. The phenotypic plasticity of macrophages plays a central role in immune cell chemotaxis, inflammatory regulation and bone regeneration. The present study addresses effects emanating from JPC-seeded β-TCP constructs (3DJPCs) co-cultivated with THP-1 derived M1/M2 macrophages within a horizontal co-culture system. After five days of co-culture, macrophage phenotype and chemokine secretion were analyzed by flow cytometry, quantitative PCR and proteome arrays. The results showed that pro-inflammatory factors in M1 macrophages were inhibited by 3DJPCs, while anti-inflammatory factors were activated, possibly affected by the multiple chemokines secreted by 3D-cultured JPCs. In addition, osteoclast markers of polarized macrophages were inhibited by osteogenically induced 3DJPCs. Functional assays revealed a significantly lower percentage of proliferating CD4+ T cells in the groups treated with secretomes from M1/M2 macrophages previously co-cultured with 3DJPCs compared to controls without secretomes. Quantifications of pit area resorption assays showed evidence that supernatants from 3DJPCs co-cultured with M1/M2 macrophages were able to completely suppress osteoclast maturation, compared to the control group without secretomes. These findings demonstrate the ability of 3D cultured JPCs to modulate macrophage plasticity.
Collapse
Affiliation(s)
- Fang He
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.H.); (L.W.); (F.U.); (A.N.); (S.R.)
| | - Liuran Wang
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.H.); (L.W.); (F.U.); (A.N.); (S.R.)
| | - Felix Umrath
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.H.); (L.W.); (F.U.); (A.N.); (S.R.)
- Clinic for Orthopaedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany
| | - Andreas Naros
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.H.); (L.W.); (F.U.); (A.N.); (S.R.)
| | - Siegmar Reinert
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.H.); (L.W.); (F.U.); (A.N.); (S.R.)
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.H.); (L.W.); (F.U.); (A.N.); (S.R.)
| |
Collapse
|
74
|
Kumari S, Mondal P, Tyeb S, Chatterjee K. Visible light-based 3D bioprinted composite scaffolds of κ-carrageenan for bone tissue engineering applications. J Mater Chem B 2024; 12:1926-1936. [PMID: 38314524 DOI: 10.1039/d3tb02179c] [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: 02/06/2024]
Abstract
Three-dimensional (3D) printing of bone scaffolds using digital light processing (DLP) bioprinting technology empowers the treatment of patients suffering from bone disorders and defects through the fabrication of cell-laden patient-specific scaffolds. Here, we demonstrate the visible-light-induced photo-crosslinking of methacrylate-κ-carrageenan (MA-κ-CA) mixed with bioactive silica nanoparticles (BSNPs) to fabricate 3D composite hydrogels using digital light processing (DLP) printing. The 3D printing of complex bone structures, such as the gyroid, was demonstrated with high precision and resolution. DLP-printed 3D composite hydrogels of MA-κ-CA-BSNP were prepared and systematically assessed for their macroporous structure, swelling, and degradation characteristics. The viscosity, rheological, and mechanical properties were also investigated for the influence of nanoparticle incorporation in the MA-κ-CA hydrogels. The in vitro study performed with MC3T3-E1 pre-osteoblast-laden scaffolds of MA-κ-CA-BSNP revealed high cell viability, no cytotoxicity, and proliferation over 21 days with markedly enhanced osteogenic differentiation compared to neat polymeric scaffolds. Furthermore, no inflammation was observed in the 21-day study involving the in vivo examination of DLP-printed 3D composite scaffolds in a Wistar rat model. Overall, the observed results for the DLP-printed 3D composite scaffolds of MA-κ-CA and BSNP demonstrate their biocompatibility and suitability for bone tissue engineering.
Collapse
Affiliation(s)
- Sushma Kumari
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India.
| | - Pritiranjan Mondal
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India.
| | - Suhela Tyeb
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India.
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India.
| |
Collapse
|
75
|
de Leeuw AM, Graf R, Lim PJ, Zhang J, Schädli GN, Peterhans S, Rohrbach M, Giunta C, Rüger M, Rubert M, Müller R. Physiological cell bioprinting density in human bone-derived cell-laden scaffolds enhances matrix mineralization rate and stiffness under dynamic loading. Front Bioeng Biotechnol 2024; 12:1310289. [PMID: 38419730 PMCID: PMC10900528 DOI: 10.3389/fbioe.2024.1310289] [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: 10/09/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Human organotypic bone models are an emerging technology that replicate bone physiology and mechanobiology for comprehensive in vitro experimentation over prolonged periods of time. Recently, we introduced a mineralized bone model based on 3D bioprinted cell-laden alginate-gelatin-graphene oxide hydrogels cultured under dynamic loading using commercially available human mesenchymal stem cells. In the present study, we created cell-laden scaffolds from primary human osteoblasts isolated from surgical waste material and investigated the effects of a previously reported optimal cell printing density (5 × 106 cells/mL bioink) vs. a higher physiological cell density (10 × 106 cells/mL bioink). We studied mineral formation, scaffold stiffness, and cell morphology over a 10-week period to determine culture conditions for primary human bone cells in this microenvironment. For analysis, the human bone-derived cell-laden scaffolds underwent multiscale assessment at specific timepoints. High cell viability was observed in both groups after bioprinting (>90%) and after 2 weeks of daily mechanical loading (>85%). Bioprinting at a higher cell density resulted in faster mineral formation rates, higher mineral densities and remarkably a 10-fold increase in stiffness compared to a modest 2-fold increase in the lower printing density group. In addition, physiological cell bioprinting densities positively impacted cell spreading and formation of dendritic interconnections. We conclude that our methodology of processing patient-specific human bone cells, subsequent biofabrication and dynamic culturing reliably affords mineralized cell-laden scaffolds. In the future, in vitro systems based on patient-derived cells could be applied to study the individual phenotype of bone disorders such as osteogenesis imperfecta and aid clinical decision making.
Collapse
Affiliation(s)
| | - Reto Graf
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Pei Jin Lim
- Connective Tissue Unit, Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jianhua Zhang
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | | - Marianne Rohrbach
- Connective Tissue Unit, Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cecilia Giunta
- Connective Tissue Unit, Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Rüger
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- Department of Pediatric Orthopaedics and Traumatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Marina Rubert
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
76
|
Boscaro D, Sikorski P. Spheroids as a 3D in vitro model to study bone and bone mineralization. BIOMATERIALS ADVANCES 2024; 157:213727. [PMID: 38101067 DOI: 10.1016/j.bioadv.2023.213727] [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: 10/10/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Traumas, fractures, and diseases can severely influence bone tissue. Insight into bone mineralization is essential for the development of therapies and new strategies to enhance bone regeneration. 3D cell culture systems, in particular cellular spheroids, have gained a lot of interest as they can recapitulate crucial aspects of the in vivo tissue microenvironment, such as the extensive cell-cell and cell-extracellular matrix (ECM) interactions found in tissue. The potential of combining spheroids and various classes of biomaterials opens also new opportunities for research within bone tissue engineering. Characterizing cellular organization, ECM structure, and ECM mineralization is a fundamental step for understanding the biological processes involved in bone tissue formation in a spheroid-based model system. Still, many experimental techniques used in this field of research are optimized for use with monolayer cell cultures. There is thus a need to develop new and improving existing experimental techniques, for applications in 3D cell culture systems. In this review, bone composition and spheroids properties are described. This is followed by an insight into the techniques that are currently used in bone spheroids research and how these can be used to study bone mineralization. We discuss the application of staining techniques used with optical and confocal fluorescence microscopy, molecular biology techniques, second harmonic imaging microscopy, Raman spectroscopy and microscopy, as well as electron microscopy-based techniques, to evaluate osteogenic differentiation, collagen production and mineral deposition. Challenges in the applications of these methods in bone regeneration and bone tissue engineering are described. STATEMENT OF SIGNIFICANCE: 3D cell cultures have gained a lot of interest in the last decades as a possible technique that can be used to recreate in vitro in vivo biological process. The importance of 3D environment during bone mineralization led scientists to use this cell culture to study this biological process, to obtain a better understanding of the events involved. New and improved techniques are also required for a proper analysis of this cell model and the process under investigation. This review summarizes the state of the art of the techniques used to study bone mineralization and how 3D cell cultures, in particular spheroids, are tested and analysed to obtain better resolved results related to this complex biological process.
Collapse
Affiliation(s)
- Diamante Boscaro
- Department of Physics, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, Trondheim 7034, Norway.
| | - Pawel Sikorski
- Department of Physics, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, Trondheim 7034, Norway.
| |
Collapse
|
77
|
Lee H, Kengla C, Kim HS, Kim I, Cho J, Renteria E, Shin K, Atala A, Yoo JJ, Lee SJ. Enhancing Craniofacial Bone Reconstruction with Clinically Applicable 3D Bioprinted Constructs. Adv Healthc Mater 2024; 13:e2302508. [PMID: 37906084 PMCID: PMC11250468 DOI: 10.1002/adhm.202302508] [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: 08/02/2023] [Revised: 10/18/2023] [Indexed: 11/02/2023]
Abstract
Medical imaging and 3D bioprinting can be used to create patient-specific bone scaffolds with complex shapes and controlled inner architectures. This study investigated the effectiveness of a biomimetic approach to scaffold design by employing geometric control. The biomimetic scaffold with a dense external layer showed improved bone regeneration compared to the control scaffold. New bone filled the defected region in the biomimetic scaffolds, while the control scaffolds only presented new bone at the boundary. Histological examination also shows effective bone regeneration in the biomimetic scaffolds, while fibrotic tissue ingrowth is observed in the control scaffolds. These findings suggest that the biomimetic bone scaffold, designed to minimize competition for fibrotic tissue formation in the bony defect, can enhance bone regeneration. This study underscores the notion that patient-specific anatomy can be accurately translated into a 3D bioprinting strategy through medical imaging, leading to the fabrication of constructs with significant clinical relevance.
Collapse
Affiliation(s)
- Hyeongjin Lee
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- Present address:
Department of Biotechnology and BioinformaticsKorea UniversitySejong30019Republic of Korea
| | - Carlos Kengla
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia TechWinston‐SalemNC27157USA
| | - Han Su Kim
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- Department of Otorhinolaryngology‐Head and Neck SurgeryCollege of MedicineEwha Womans UniversitySeoul07804Republic of Korea
| | - Ickhee Kim
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Jae‐Gu Cho
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- Department of Otorhinolaryngology‐Head and Neck SurgeryCollege of MedicineKorea UniversitySeoul02708Republic of Korea
| | - Eric Renteria
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
| | - Kyungsup Shin
- Department of OrthodonticsUniversity of Iowa College of DentistryIowa CityIA52242USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia TechWinston‐SalemNC27157USA
| | - James J. Yoo
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia TechWinston‐SalemNC27157USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative MedicineWake Forest University School of MedicineWinston‐SalemNC27157USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia TechWinston‐SalemNC27157USA
| |
Collapse
|
78
|
Zhang YD, Ma AB, Sun L, Chen JD, Hong G, Wu HK. Nanoclay-Modified Hyaluronic Acid Microspheres for Bone Induction by Sustained rhBMP-2 Delivery. Macromol Biosci 2024; 24:e2300245. [PMID: 37572308 DOI: 10.1002/mabi.202300245] [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: 05/30/2023] [Revised: 07/12/2023] [Indexed: 08/14/2023]
Abstract
Microspheres (MSs) are ideal candidates as biological scaffolds loading with growth factors or cells for bone tissue engineering to repair irregular alveolar bone defects by minimally invasive injection. However, the high initial burst release of growth factor and low cell attachment limit the application of microspheres. The modification of microspheres often needs expensive experiments facility or complex chemical reactions, which is difficult to achieve and may bring other problems. In this study, a sol-grade nanoclay, laponite XLS is used to modify the surface of MSs to enhance its affinity to either positively or negatively charged proteins and cells without changing the interior structure of the MSs. Recombinant human bone morphogenetic protein-2 (rhBMP-2) is used as a representation of growth factor to check the osteoinduction ability of laponite XLS-modified MSs. By modification, the protein sustained release, cell loading, and osteoinduction ability of MSs are improved. Modified by 1% laponite XLS, the MSs can not only promote osteogenic differentiation of MC3T3-E1 cells by themselves, but also enhance the effect of the rhBMP-2 below the effective dose. Collectively, the study provides an easy and viable method to modify the biological behavior of microspheres for bone tissue regeneration.
Collapse
Affiliation(s)
- Yi-Ding Zhang
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Section 3, South Renmin Road, Chengdu, Sichuan, 610041, P. R. China
| | - Ao-Bo Ma
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Lu Sun
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Jun-Duo Chen
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Guang Hong
- Division for Globalization Initiative, Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
- Department of Prosthodontics, Faculty of Dental Medicine, Airlangga University, Surabaya, 60115, Indonesia
| | - Hong-Kun Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Section 3, South Renmin Road, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
79
|
Liu H, Li K, Guo B, Yuan Y, Ruan Z, Long H, Zhu J, Zhu Y, Chen C. Engineering an injectable gellan gum-based hydrogel with osteogenesis and angiogenesis for bone regeneration. Tissue Cell 2024; 86:102279. [PMID: 38007880 DOI: 10.1016/j.tice.2023.102279] [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: 07/05/2023] [Revised: 11/10/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
Injectable hydrogels are currently a topic of great interest in bone tissue engineering, which could fill irregular bone defects in a short time and avoid traditional major surgery. Herein, we developed an injectable gellan gum (GG)-based hydrogel for bone defect repair by blending nano-hydroxyapatite (nHA) and magnesium sulfate (MgSO4). In order to acquire an injectable GG-based hydrogel with superior osteogenesis, nHA were blended into GG solution with an optimized proportion. For the aim of endowing this hydrogel capable of angiogenesis, MgSO4 was also incorporated. Physicochemical evaluation revealed that GG-based hydrogel containing 5% nHA (w/v) and 2.5 mM MgSO4 (GG/5%nHA/MgSO4) had appropriate sol-gel transition time, showed a porosity-like structure, and could release magnesium ions for at least 14 days. Rheological studies showed that the GG/5%nHA/MgSO4 hydrogel had a stable structure and repeatable self-healing properties. In-vitro results determined that GG/5%nHA/MgSO4 hydrogel presented superior ability on stimulating bone marrow mesenchymal stem cells (BMSCs) to differentiate into osteogenic linage and human umbilical vein endothelial cells (HUVECs) to generate vascularization. In-vivo, GG/5%nHA/MgSO4 hydrogel was evaluated via a rat cranial defect model, as shown by better new bone formation and more neovascularization invasion. Therefore, the study demonstrated that the new injectable hydrogel, is a favorable bioactive GG-based hydrogel, and provides potential strategies for robust therapeutic interventions to improve the repair of bone defect.
Collapse
Affiliation(s)
- Hongbin Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Kaihu Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410000, Hunan, China
| | - Bin Guo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Yuhao Yuan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Zhe Ruan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Haitao Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Jianxi Zhu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Yong Zhu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China.
| | - Can Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China.
| |
Collapse
|
80
|
Manohar SS, Das C, Kakati V. Bone Tissue Engineering Scaffolds: Materials and Methods. 3D PRINTING AND ADDITIVE MANUFACTURING 2024; 11:347-362. [PMID: 38389691 PMCID: PMC10880649 DOI: 10.1089/3dp.2022.0216] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The wide development in biomedical, regenerative medicine, and surgical techniques has ensured that new technologies are developed to improve patient-specific treatment and care. Tissue engineering is a special field in biomedical engineering that works toward cell development using scaffolds. Bone tissue engineering is a separate branch of tissue engineering, in which the construction of bone, functionalities of bone, and bone tissue regeneration are studied in detail to repair or regenerate new functional bone tissues. In India alone, people suffering from bone diseases are extensive in numbers. Almost 15% to 20% of the population suffers from osteoporosis. Bone scaffolds are proving to be an excellent solution for osseous abnormalities or defect treatment. Scaffolds are three dimensional (3D) and mostly porous structures created to enhance new tissue growth. Bone scaffolds are specially designed to promote osteoinductive cell growth, expansion, and migration on their surface. This review article aims to provide an overview of possible bone scaffolding materials in practice, different 3D techniques to fabricate these scaffolds, and effective bone scaffold characteristics targeted by researchers to fabricate tissue-engineered bone scaffolds.
Collapse
Affiliation(s)
- Shreeprasad S. Manohar
- Mechanical Engineering Department, Assam Don Bosco University, Guwahati, India
- Mechanical Department, DBIT, Mumbai, India
| | - Chinmoy Das
- Department of Orthopaedics, Tezpur Medical College and Hospital, Tezpur, India
| | - Vikramjit Kakati
- Mechanical Engineering Department, Assam Don Bosco University, Guwahati, India
| |
Collapse
|
81
|
Narde J, Ganapathy D, Pandurangan KK. Evaluation of the Success of Autogenous Block Grafting in Atrophic Maxillary and Mandibular Ridges Prior to and After Implant Placement. Cureus 2024; 16:e53829. [PMID: 38465098 PMCID: PMC10924432 DOI: 10.7759/cureus.53829] [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: 12/30/2023] [Accepted: 02/03/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Dental implantology's success relies on adequate bone volume and quality, necessitating bone augmentation for implant placement. Primary lateral bone augmentation, utilizing autogenous block grafts, addresses horizontal bone loss. OBJECTIVE This study aims to evaluate the efficacy of autogenous block grafting, specifically ramus and fibula blocks, in addressing severe atrophic ridges before and after implant placement. METHODS Twenty-one patients underwent block grafting, predominantly using the ramus technique (80/20 ratio). CBCT measurements assessed horizontal grafting outcomes. Implant success and bone volume changes were analyzed. RESULTS Post-grafting, bone width increased from 1.8-3.1 mm to 4.5-6 mm, exceeding critical thresholds. Implant success reached 95%, indicating the grafting techniques' effectiveness. CONCLUSION Autogenous block grafting, especially with ramus and fibula blocks, transforms severe atrophic ridges, enabling successful implant integration. Long-term follow-up is essential for a comprehensive evaluation. CLINICAL RELEVANCE This study provides crucial insights into autogenous block grafting's transformative impact on challenging cases, guiding future applications in reconstructive dentistry.
Collapse
Affiliation(s)
- Joshua Narde
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Kiran Kumar Pandurangan
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
82
|
Kadi F, Dini G, Poursamar SA, Ejeian F. Fabrication and characterization of 3D-printed composite scaffolds of coral-derived hydroxyapatite nanoparticles/polycaprolactone/gelatin carrying doxorubicin for bone tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:7. [PMID: 38285297 PMCID: PMC10824813 DOI: 10.1007/s10856-024-06779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
In this study, nanocomposite scaffolds of hydroxyapatite (HA)/polycaprolactone (PCL)/gelatin (Gel) with varying amounts of HA (42-52 wt. %), PCL (42-52 wt. %), and Gel (6 wt. %) were 3D printed. Subsequently, a scaffold with optimal mechanical properties was utilized as a carrier for doxorubicin (DOX) in the treatment of bone cancer. For this purpose, HA nanoparticles were first synthesized by the hydrothermal conversion of Acropora coral and characterized by using different techniques. Also, a compression test was performed to investigate the mechanical properties of the fabricated scaffolds. The mineralization of the optimal scaffold was determined by immersing it in simulated body fluid (SBF) solution for 28 days, and the biocompatibility was investigated by seeding MG-63 osteoblast-like cells on it after 1-7 days. The obtained results showed that the average size of the synthesized HA particles was about 80 nm. The compressive modulus and strength of the scaffold with 47 wt. % HA was reported to be 0.29 GPa and 9.9 MPa, respectively, which was in the range of trabecular bones. In addition, the scaffold surface was entirely coated with an apatite layer after 28 days of soaking in SBF. Also, the efficiency and loading percentage of DOX were obtained as 30.8 and 1.6%, respectively. The drug release behavior was stable for 14 days. Cytotoxicity and adhesion evaluations showed that the fabricated scaffold had no negative effects on the viability of MG-63 cells and led to their proliferation during the investigated period. From these results, it can be concluded that the HA/PCL/Gel scaffold prepared in this study, in addition to its drug release capability, has good bioactivity, mechanical properties, and biocompatibility, and can be considered a suitable option for bone tumor treatment.
Collapse
Affiliation(s)
- Fatima Kadi
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan, 81746-73441, Iran
| | - Ghasem Dini
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan, 81746-73441, Iran.
| | - S Ali Poursamar
- Department of Biomaterials, Nanotechnology, and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, 81593-58686, Iran
| |
Collapse
|
83
|
Gabetti S, Masante B, Schiavi A, Scatena E, Zenobi E, Israel S, Sanginario A, Del Gaudio C, Audenino A, Morbiducci U, Massai D. Adaptable test bench for ASTM-compliant permeability measurement of porous scaffolds for tissue engineering. Sci Rep 2024; 14:1722. [PMID: 38242930 PMCID: PMC10799031 DOI: 10.1038/s41598-024-52159-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/15/2024] [Indexed: 01/21/2024] Open
Abstract
Intrinsic permeability describes the ability of a porous medium to be penetrated by a fluid. Considering porous scaffolds for tissue engineering (TE) applications, this macroscopic variable can strongly influence the transport of oxygen and nutrients, the cell seeding process, and the transmission of fluid forces to the cells, playing a crucial role in determining scaffold efficacy. Thus, accurately measuring the permeability of porous scaffolds could represent an essential step in their optimization process. In literature, several methods have been proposed to characterize scaffold permeability. Most of the currently adopted approaches to assess permeability limit their applicability to specific scaffold structures, hampering protocols standardization, and ultimately leading to incomparable results among different laboratories. The content of novelty of this study is in the proposal of an adaptable test bench and in defining a specific testing protocol, compliant with the ASTM International F2952-22 guidelines, for reliable and repeatable measurements of the intrinsic permeability of TE porous scaffolds. The developed permeability test bench (PTB) exploits the pump-based method, and it is composed of a modular permeability chamber integrated within a closed-loop hydraulic circuit, which includes a peristaltic pump and pressure sensors, recirculating demineralized water. A specific testing protocol was defined for characterizing the pressure drop associated with the scaffold under test, while minimizing the effects of uncertainty sources. To assess the operational capabilities and performance of the proposed test bench, permeability measurements were conducted on PLA scaffolds with regular (PS) and random (RS) micro-architecture and on commercial bovine bone matrix-derived scaffolds (CS) for bone TE. To validate the proposed approach, the scaffolds were as well characterized using an alternative test bench (ATB) based on acoustic measurements, implementing a blind randomized testing procedure. The consistency of the permeability values measured using both the test benches demonstrated the reliability of the proposed approach. A further validation of the PTB's measurement reliability was provided by the agreement between the measured permeability values of the PS scaffolds and the theory-based predicted permeability value. Once validated the proposed PTB, the performed measurements allowed the investigation of the scaffolds' transport properties. Samples with the same structure (guaranteed by the fused-deposition modeling technique) were characterized by similar permeability values, and CS and RS scaffolds showed permeability values in agreement with the values reported in the literature for bovine trabecular bone. In conclusion, the developed PTB and the proposed testing protocol allow the characterization of the intrinsic permeability of porous scaffolds of different types and dimensions under controlled flow regimes, representing a powerful tool in view of providing a reliable and repeatable framework for characterizing and optimizing scaffolds for TE applications.
Collapse
Affiliation(s)
- Stefano Gabetti
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Corso Duca Degli Abruzzi, 24, 10129, Turin, Italy
- Centro 3R, Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Beatrice Masante
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Corso Duca Degli Abruzzi, 24, 10129, Turin, Italy
- Centro 3R, Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
- Department of Surgical Sciences, CIR-Dental School, University of Turin, Turin, Italy
| | - Alessandro Schiavi
- Applied Metrology and Engineering Division, INRiM-National Institute of Metrological Research, Turin, Italy
| | | | | | - Simone Israel
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Corso Duca Degli Abruzzi, 24, 10129, Turin, Italy
- Centro 3R, Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Alessandro Sanginario
- Department of Electronics and Telecommunications, Politecnico di Torino, Turin, Italy
| | | | - Alberto Audenino
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Corso Duca Degli Abruzzi, 24, 10129, Turin, Italy
- Centro 3R, Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Corso Duca Degli Abruzzi, 24, 10129, Turin, Italy
- Centro 3R, Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Diana Massai
- Department of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Corso Duca Degli Abruzzi, 24, 10129, Turin, Italy.
- Centro 3R, Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy.
| |
Collapse
|
84
|
Mauro N, Calabrese G, Sciortino A, Rizzo MG, Messina F, Giammona G, Cavallaro G. Microporous Fluorescent Poly(D,L-lactide) Acid-Carbon Nanodot Scaffolds for Bone Tissue Engineering Applications. MATERIALS (BASEL, SWITZERLAND) 2024; 17:449. [PMID: 38255617 PMCID: PMC10820564 DOI: 10.3390/ma17020449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024]
Abstract
In this study, we introduce novel microporous poly(D,L-lactide) acid-carbon nanodot (PLA-CD) nanocomposite scaffolds tailored for potential applications in image-guided bone regeneration. Our primary objective was to investigate concentration-dependent structural variations and their relevance to cell growth, crucial aspects in bone regeneration. The methods employed included comprehensive characterization techniques such as DSC/TGA, FTIR, rheological, and degradation assessments, providing insights into the scaffolds' thermoplastic behavior, microstructure, and stability over time. Notably, the PLA-CD scaffolds exhibited distinct self-fluorescence, which persisted after 21 days of incubation, allowing detailed visualization in various multicolor modalities. Biocompatibility assessments were conducted by analyzing human adipose-derived stem cell (hADSC) growth on PLA-CD scaffolds, with results substantiated through cell viability and morphological analyses. hADSCs reached a cell viability of 125% and penetrated throughout the scaffold after 21 days of incubation. These findings underscore the scaffolds' potential in bone regeneration and fluorescence imaging. The multifunctional nature of the PLA-CD nanocomposite, integrating diagnostic capabilities with tunable properties, positions it as a promising candidate for advancing bone tissue engineering. Our study not only highlights key aspects of the investigation but also underscores the scaffolds' specific application in bone regeneration, providing a foundation for further research and optimization in this critical biomedical field.
Collapse
Affiliation(s)
- Nicolò Mauro
- Department of “Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche” (STEBICEF), Università Degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy; (G.G.); (G.C.)
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98168 Messina, Italy; (G.C.); (M.G.R.)
| | - Alice Sciortino
- Department of Chimica e Fisica “E. Segrè”, Università Degli Studi di Palermo, Via Archirafi 36, 90123 Palermo, Italy; (A.S.); (F.M.)
| | - Maria G. Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98168 Messina, Italy; (G.C.); (M.G.R.)
| | - Fabrizio Messina
- Department of Chimica e Fisica “E. Segrè”, Università Degli Studi di Palermo, Via Archirafi 36, 90123 Palermo, Italy; (A.S.); (F.M.)
| | - Gaetano Giammona
- Department of “Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche” (STEBICEF), Università Degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy; (G.G.); (G.C.)
| | - Gennara Cavallaro
- Department of “Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche” (STEBICEF), Università Degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy; (G.G.); (G.C.)
| |
Collapse
|
85
|
Luo Y, Kim J. Achieving the ideal balance between biological and mechanical requirements in composite bone scaffolds through a voxel-based approach. Comput Methods Biomech Biomed Engin 2024:1-14. [PMID: 38231253 DOI: 10.1080/10255842.2024.2304709] [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: 11/04/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024]
Abstract
Achieving successful bone regeneration necessitates the design of scaffolds that meet diverse biological and mechanical requirements, often leading to conflicts in the design parameters. A key conflict arises between scaffold porosity and stiffness. Increasing porosity facilitates cell infiltration and nutrient exchange, promoting bone regeneration. However, higher porosity compromises scaffold stiffness, which is crucial for providing structural support in the defective region. Furthermore, appropriate scaffold stiffness is crucial for preventing stress shielding. Conventional geometry-based design methods utilizing single-phase materials have limited flexibility in resolving such conflicts. To address this challenge, we propose a voxel-based method for designing composite scaffolds composed of hydroxyapatite (HA) and polylactic acid (PLA). Our strategy involves first satisfying primary biological requirements by selecting appropriate porosity, pore shape, and size. Subsequently, scaffold stiffness requirements are met by selecting suitable phase materials and tuning their contents. The study demonstrates that the voxel-based approach effectively balances both biological and mechanical requirements in scaffold design. This method addresses the limitations of traditional designs by achieving an optimal balance between porosity and stiffness, which is crucial for scaffold performance in biomedical applications. Moreover, the scaffolds designed using this method can be manufactured using voxel-based 3D printing technology, which is emerging in the field. Future advancements in voxel-based 3D printing technology will further enhance the feasibility and practicality of this approach for bone tissue engineering applications.
Collapse
Affiliation(s)
- Yunhua Luo
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Canada
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, Canada
| | - Jonghyun Kim
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
86
|
Kuperkar K, Atanase LI, Bahadur A, Crivei IC, Bahadur P. Degradable Polymeric Bio(nano)materials and Their Biomedical Applications: A Comprehensive Overview and Recent Updates. Polymers (Basel) 2024; 16:206. [PMID: 38257005 PMCID: PMC10818796 DOI: 10.3390/polym16020206] [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: 12/06/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Degradable polymers (both biomacromolecules and several synthetic polymers) for biomedical applications have been promising very much in the recent past due to their low cost, biocompatibility, flexibility, and minimal side effects. Here, we present an overview with updated information on natural and synthetic degradable polymers where a brief account on different polysaccharides, proteins, and synthetic polymers viz. polyesters/polyamino acids/polyanhydrides/polyphosphazenes/polyurethanes relevant to biomedical applications has been provided. The various approaches for the transformation of these polymers by physical/chemical means viz. cross-linking, as polyblends, nanocomposites/hybrid composites, interpenetrating complexes, interpolymer/polyion complexes, functionalization, polymer conjugates, and block and graft copolymers, are described. The degradation mechanism, drug loading profiles, and toxicological aspects of polymeric nanoparticles formed are also defined. Biomedical applications of these degradable polymer-based biomaterials in and as wound dressing/healing, biosensors, drug delivery systems, tissue engineering, and regenerative medicine, etc., are highlighted. In addition, the use of such nano systems to solve current drug delivery problems is briefly reviewed.
Collapse
Affiliation(s)
- Ketan Kuperkar
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology (SVNIT), Ichchhanath, Piplod, Surat 395007, Gujarat, India;
| | - Leonard Ionut Atanase
- Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania
- Academy of Romanian Scientists, 050045 Bucharest, Romania
| | - Anita Bahadur
- Department of Zoology, Sir PT Sarvajanik College of Science, Surat 395001, Gujarat, India;
| | - Ioana Cristina Crivei
- Department of Public Health, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” University of Life Sciences, 700449 Iasi, Romania;
| | - Pratap Bahadur
- Department of Chemistry, Veer Narmad South Gujarat University (VNSGU), Udhana-Magdalla Road, Surat 395007, Gujarat, India;
| |
Collapse
|
87
|
Çobandede Z, Çulha M. Ultrasound stimulated piezoelectric barium titanate and boron nitride nanotubes in nonconductive poly- ε-caprolactone nanofibrous scaffold for bone tissue engineering. NANOTECHNOLOGY 2024; 35:135101. [PMID: 38081081 DOI: 10.1088/1361-6528/ad1446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/10/2023] [Indexed: 01/10/2024]
Abstract
Nanomaterials can provide unique solutions for the problems experienced in tissue engineering by improving a scaffold's physico-bio-chemical properties. With its piezoelectric property, bone is an active tissue with easy adaptation and remodeling through complicated mechanisms of electromechanical operations. Although poly(ε-caprolactone) (PCL) is an excellent polymer for bone tissue engineering, it is lack of conductivity. In this study, piezoelectric barium titanates (BaTiO3) and boron nitride nanotubes (BNNTs) are used as ultrasound (US) stimulated piezoelectric components in PCL to mimic piezoelectric nature of bone tissue. Electric-responsive Human Osteoblast cells on the scaffolds were stimulated by applying low-frequency US during cell growth. Biocompatibility, cell adhesion, alkaline phosphatase activities and mineralization of osteoblast cells on piezo-composite scaffolds were investigated. BaTiO3or BNNTs as reinforcement agents improved physical and mechanical properties of PCL scaffolds.In vitrostudies show that the use of BaTiO3or BNNTs as additives in non-conductive scaffolds significantly induces and increases the osteogenic activities even without US stimulation. Although BaTiO3is one of the best piezoelectric materials, the improvement is more dramatic in the case of BNNTs with the increased mineralization, and excellent chemical and mechanical properties.
Collapse
Affiliation(s)
- Zehra Çobandede
- Department of Genetics and Bioenginering, Yeditepe University, Atasehir, Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, 34956, Turkey
| | - Mustafa Çulha
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, 34956, Turkey
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA, United States of America
| |
Collapse
|
88
|
Suneetha M, Kim H, Han SS. Bone-like apatite formation in biocompatible phosphate-crosslinked bacterial cellulose-based hydrogels for bone tissue engineering applications. Int J Biol Macromol 2024; 256:128364. [PMID: 38000603 DOI: 10.1016/j.ijbiomac.2023.128364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
Addressing major bone injuries is a challenge in bone regeneration, necessitating innovative 3D hydrogel-based therapeutic approaches to enhance scaffold properties for better bioactivity. Bacterial cellulose (BC) is an excellent scaffold for bone tissue engineering due to its biocompatibility, high porosity, substantial surface area, and remarkable mechanical strength. However, its practical application is limited due to a lack of inherent osteogenic activity and biomineralization ability. In this study, we synthesized bone-like apatite in biocompatible BC hydrogel by introducing phosphate groups. Hydrogels were prepared using fibrous BC, acrylamide (AM), and bis [2-methacryloyloxy] ethyl phosphate (BMEP) as a crosslinker through free radical polymerization (P-BC-PAM). P-BC-PAM hydrogels exhibited outstanding compressive mechanical properties, highly interconnected porous structures, good swelling, and biodegradable properties. BMEP content significantly influenced the physicochemical and biological properties of the hydrogels. Increasing BMEP content enhanced the fibrous structure, porosity from 85.1 % to 89.5 %, and compressive mechanical strength. The optimized hydrogel (2.0P-BC-PAM) displayed maximum compressive stress, toughness, and elastic modulus at 75 % strain: 221 ± 0.08 kPa, 24,674.2 ± 978 kPa, and 11 ± 0.47 kPa, respectively. P-BC-PAM hydrogels underwent biomineralization in simulated body fluid (SBF) for 14 days, forming bone-like apatite with a Ca/P ratio of 1.75, similar to hydroxyapatite. Confirmed by Fourier-transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), and field-emission scanning electron microscopy (FE-SEM), this suggests their potential as scaffolds for bone tissue engineering. MC3T3-E1 osteoblast cells effectively attached and proliferated on P-BC-PAM. In summary, this study contributes insights into developing phosphate-functionalized BC-based hydrogels with potential applications in bone tissue engineering.
Collapse
Affiliation(s)
- Maduru Suneetha
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Hyeonjin Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
89
|
Senthil R. Bone implant substitutes from synthetic polymer and reduced graphene oxide: Current perspective. Int J Artif Organs 2024; 47:57-66. [PMID: 38087802 DOI: 10.1177/03913988231216572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2024]
Abstract
In the present work, bone implant materials (BIM) were produced, in sheet form which comprises epoxy resin (synthetic polymer) (ER), calcium carbonate (CaCO3), and reduced graphene oxide (R-GO), by open mold method, for the possibility uses in bone tissue engineering. The developed BIM was analyzed for its physico-chemical, mechanical, bioactivity test, antimicrobial study, and biocompatibility. The BIM had excellent mechanical properties such as tensile strength (194.44 + 0.21 MPa), flexural strength (278.76 + 0.41 MPa), and water absorption (02.61 + 0.24%). A pore size distribution study using the HR-SEM has proved the 180 and 255 μm average pore was observed in the BIM structure. The Bioactivity test of BIM was examined after being immersed in a simulated body fluids (SBF) solution. The result of BIM formed an excellent deposition of bone tube apatite crystals. High-resolution scanning electron microscopy (HR-SEM) morphology of the bone tube apatite crystals revealed the diameter size in the range from 100 ± 159 to 210 ± 188 nm. BIM has excellent antimicrobial characteristics against E. coli (8.75 + 0.06 mm) and S. aureus (9.82 + 0.08 mm). The biocompatibility of the study MTT (3-(4, 5-dimethyl) thiazol-2-yl-2, 5-dimethyl tetrazolium bromide) assay using the MG-63 (human osteoblast cell line) has proven to be the 78% viable cell presence in BIM. After receiving the necessary approval, the scaffold with the required strength and biocompatibility could be tested as a bone implant material in large animals.
Collapse
Affiliation(s)
- Rethinam Senthil
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
90
|
da Silva RBP, Biguetti CC, Munerato MS, Siqueira RL, Zanotto ED, Kudo GHA, Simionato GB, Bacelar ACZ, Ortiz RC, Ferreira-Junior JS, Rangel-Junior IG, Matsumoto MA. Effects of glass-ceramic produced by the sol-gel route in macrophages recruitment and polarization into bone tissue regeneration. J Biomed Mater Res B Appl Biomater 2024; 112:e35340. [PMID: 37929804 DOI: 10.1002/jbm.b.35340] [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: 05/26/2023] [Revised: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 11/07/2023]
Abstract
Effective bone substitute biomaterials remain an important challenge in patients with large bone defects. Glass ceramics produced by different synthesis routes may result in changes in the material physicochemical properties and consequently affect the success or failure of the bone healing response. To investigate the differences in the orchestration of the inflammatory and healing process in bone grafting and repair using different glass-ceramic routes production. Thirty male Wistar rats underwent surgical unilateral parietal defects filled with silicate glass-ceramic produced by distinct routes: BS - particulate glass-ceramic produced via the fusion/solidification route, and BG - particulate glass-ceramic produced via the sol-gel route. After 7, 14, and 21 days from biomaterial grafting, parietal bones were removed to be analyzed under H&E and Massons' Trichome staining, and immunohistochemistry for CD206, iNOS, and TGF-β. Our findings demonstrated that the density of lymphocytes and plasma cells was significantly higher in the BS group at 45, and 7 days compared to the BG group, respectively. Furthermore, a significant increase of foreign body giant cells (FBGCs) in the BG group at day 7, compared to BS was found, demonstrating early efficient recruitment of FBGCs against sol-gel-derived glass-ceramic particulate (BS group). According to macrophage profiles, CD206+ macrophages enhanced at the final periods of both groups, being significantly higher at 45 days of BS compared to the BG group. On the other hand, the density of transformation growth factor beta (TGF-β) positive cells on 21 days were the highest in BG, and the lowest in the BS group, demonstrating a differential synergy among groups. Noteworthy, TGF-β+ cells were significantly higher at 21 days of BG compared to the BS group. Glass-ceramic biomaterials can act differently in the biological process of bone remodeling due to their route production, being the sol-gel route more efficient to activate M2 macrophages and specific FBGCs compared to the traditional route. Altogether, these features lead to a better understanding of the effectiveness of inflammatory response for biomaterial degradation and provide new insights for further preclinical and clinical studies involved in bone healing.
Collapse
Affiliation(s)
| | - Claudia Cristina Biguetti
- Regenerative Medicine Laboratory, School of Podiatric Medicine, The University of Texas Rio Grande Valley - UTRGV, Harlingen, Texas, USA
| | | | - Renato Luis Siqueira
- Department of Material Engineering, São Carlos Federal University, São Paulo, Brazil
| | - Edgard Dutra Zanotto
- Department of Material Engineering, São Carlos Federal University, São Paulo, Brazil
| | | | - Gustavo Baroni Simionato
- Department of Basic Sciences, São Paulo State University (Unesp), School of Dentistry, Araçatuba, Brazil
| | - Ana Carolina Zucon Bacelar
- Department of Basic Sciences, São Paulo State University (Unesp), School of Dentistry, Araçatuba, Brazil
| | - Rafael Carneiro Ortiz
- Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo, Bauru, Brazil
| | | | - Idelmo Garcia Rangel-Junior
- Department of Diagnosis and Surgery, São Paulo State University (Unesp), School of Dentistry, Araçatuba, Brazil
| | - Mariza Akemi Matsumoto
- Department of Basic Sciences, São Paulo State University (Unesp), School of Dentistry, Araçatuba, Brazil
| |
Collapse
|
91
|
Fan L, Chen S, Yang M, Liu Y, Liu J. Metallic Materials for Bone Repair. Adv Healthc Mater 2024; 13:e2302132. [PMID: 37883735 DOI: 10.1002/adhm.202302132] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/16/2023] [Indexed: 10/28/2023]
Abstract
Repair of large bone defects caused by trauma or disease poses significant clinical challenges. Extensive research has focused on metallic materials for bone repair because of their favorable mechanical properties, biocompatibility, and manufacturing processes. Traditional metallic materials, such as stainless steel and titanium alloys, are widely used in clinics. Biodegradable metallic materials, such as iron, magnesium, and zinc alloys, are promising candidates for bone repair because of their ability to degrade over time. Emerging metallic materials, such as porous tantalum and bismuth alloys, have gained attention as bone implants owing to their bone affinity and multifunctionality. However, these metallic materials encounter many practical difficulties that require urgent improvement. This article systematically reviews and analyzes the metallic materials used for bone repair, providing a comprehensive overview of their morphology, mechanical properties, biocompatibility, and in vivo implantation. Furthermore, the strategies and efforts made to address the short-comings of metallic materials are summarized. Finally, the perspectives for the development of metallic materials to guide future research and advancements in clinical practice are identified.
Collapse
Affiliation(s)
- Linlin Fan
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Sen Chen
- Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Minghui Yang
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yajun Liu
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Spine Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Jing Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
92
|
Calvert ND, Proulx S, Rodriguez-Navarro A, Ahmed T, Lehoux EA, Hincke MT, Catelas I. Development of hydrogel-based composite scaffolds containing eggshell particles for bone regeneration applications. J Biomed Mater Res B Appl Biomater 2024; 112:e35296. [PMID: 37702399 DOI: 10.1002/jbm.b.35296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/23/2023] [Accepted: 06/10/2023] [Indexed: 09/14/2023]
Abstract
This study describes the development and characterization of novel composite scaffolds, made of an alginate-chitosan hydrogel matrix containing eggshell (ES) particles, for bone tissue engineering applications. Scaffolds with ES particles, either untreated or treated with phosphoric acid to create a nanotextured particle surface, were compared to scaffolds without particles. Results indicate that the nanotexturing process exposed occluded ES proteins orthologous to those in human bone extracellular matrix. Scaffolds with ES or nanotextured ES (NTES) particles had a higher porosity (81 ± 4% and 89 ± 5%, respectively) than scaffolds without particles (59 ± 5%) (p = .002 and p < .001, respectively). Scaffolds with NTES particles had a larger median pore size (113 μm [interquartile range [IQ]: 88-140 μm]) than scaffolds with ES particles (94 μm [IQ: 75-112 μm]) and scaffolds without particles (99 μm [IQ: 74-135 μm]) (p < .001 and p = .011, respectively). The compressive modulus of the scaffolds with ES or NTES particles remained low (3.69 ± 0.70 and 3.14 ± 0.62 kPa, respectively), but these scaffolds were more resistant to deformation following maximum compression than those without particles. Finally, scaffolds with ES or NTES particles allowed better retention of human mesenchymal stem cells during seeding (53 ± 12% and 57 ± 8%, respectively, vs. 17 ± 5% for scaffolds without particles; p < .001 in both cases), as well as higher cell viability up to 21 days of culture (67 ± 17% and 61 ± 11%, respectively, vs. 15 ± 7% for scaffolds without particles; p < .001 in both cases). In addition, alkaline phosphatase (ALP) activity increased up to 558 ± 164% on day 21 in the scaffolds with ES particles, and up to 567 ± 217% on day 14 in the scaffolds with NTES particles (p = .006 and p = .002, respectively, relative to day 0). Overall, this study shows that the physicochemical properties of the alginate-chitosan hydrogel scaffolds with ES or NTES particles are similar to those of cancellous bone. In addition, scaffolds with particles supported early osteogenic differentiation and therefore represent a promising new bone substitute, especially for non-load bearing applications.
Collapse
Affiliation(s)
- Nicholas D Calvert
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Scott Proulx
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Tamer Ahmed
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Eric A Lehoux
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Ontario, Canada
| | - Maxwell T Hincke
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Innovation in Medical Education, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Isabelle Catelas
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
93
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
94
|
Yao H, Fu Q, Zhang Y, Wan Y, Min Q. Strong, elastic and degradation-tolerated hydrogels composed of chitosan, silk fibroin and bioglass nanoparticles with factor-bestowed activity for bone tissue engineering. Int J Biol Macromol 2023; 253:126619. [PMID: 37657578 DOI: 10.1016/j.ijbiomac.2023.126619] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/06/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Polymer hydrogels intended for use in bone repair need to be strong, elastic, and capable of enduring degradation. However, many natural polymer hydrogels lack these essential properties and thus, are unsuitable for bone repair applications. Here, a new type of multi-network hydrogel with improved mechanical and degradation-resistant properties has been developed for use in bone repair. The hydrogel is composed of thiolated chitosan (TCH), silk fibroin (SF), and thiolated bioglass (TBG) nanoparticles (NPs). The multi-networks are built through sulfhydryl self-crosslinking, diepoxide crosslinker-involved linkages of amino or hydroxyl groups, and enzyme-mediated phenol hydroxyl crosslinking. Additionally, mesoporous TBG NPs serve as a vehicle for loading stromal cell-derived factor-1 (SDF-1) to provide the gel with cell-recruiting activity. The formulated TCH/SF/TBG hydrogels exhibit remarkably enhanced strength, elasticity, and improved degradation tolerance compared to some gels made from only TCH or SF. Furthermore, TCH/SF/TBG gels can support the growth of seeded cells and the deposition of matrix components. Some TCH/SF/TBG gels also demonstrate the ability to release SDF-1 in an approximately linear manner for a few weeks while retaining the chemotactic properties of the released SDF-1. Overall, the multi-network hydrogel has the potential as an in situ forming material for cell-recruiting bone repair and regeneration.
Collapse
Affiliation(s)
- Hui Yao
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, PR China
| | - Qiaoqin Fu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Yuchen Zhang
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, PR China
| | - Ying Wan
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China.
| | - Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, PR China.
| |
Collapse
|
95
|
Bharathi R, Harini G, Sankaranarayanan A, Shanmugavadivu A, Vairamani M, Selvamurugan N. Nuciferine-loaded chitosan hydrogel-integrated 3D-printed polylactic acid scaffolds for bone tissue engineering: A combinatorial approach. Int J Biol Macromol 2023; 253:127492. [PMID: 37858655 DOI: 10.1016/j.ijbiomac.2023.127492] [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: 08/18/2023] [Revised: 10/07/2023] [Accepted: 10/15/2023] [Indexed: 10/21/2023]
Abstract
Critical-sized bone defects resulting from severe trauma and open fractures cannot spontaneously heal and require surgical intervention. Limitations of traditional bone grafting include immune rejection and demand-over-supply issues leading to the development of novel tissue-engineered scaffolds. Nuciferine (NF), a plant-derived alkaloid, has excellent therapeutic properties, but its osteogenic potential is yet to be reported. Furthermore, the bioavailability of NF is obstructed due to its hydrophobicity, requiring an efficient drug delivery system, such as chitosan (CS) hydrogel. We designed and fabricated polylactic acid (PLA) scaffolds via 3D printing and integrated them with NF-containing CS hydrogel to obtain the porous biocomposite scaffolds (PLA/CS-NF). The fabricated scaffolds were subjected to in vitro physicochemical characterization, cytotoxicity assays, and osteogenic evaluation studies. Scanning electron microscopic studies revealed uniform pore size distribution on PLA/CS-NF scaffolds. An in vitro drug release study showed a sustained and prolonged release of NF. The cyto-friendly nature of NF in PLA/CS-NF scaffolds towards mouse mesenchymal stem cells (mMSCs) was observed. Also, cellular and molecular level studies signified the osteogenic potential of NF in PLA/CS-NF scaffolds on mMSCs. These results indicate that the PLA/CS-NF scaffolds could promote new bone formation and have potential applications in bone tissue engineering.
Collapse
Affiliation(s)
- Ramanathan Bharathi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ganesh Harini
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Aravind Sankaranarayanan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Mariappanadar Vairamani
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India..
| |
Collapse
|
96
|
Bektas C, Mao Y. Hydrogel Microparticles for Bone Regeneration. Gels 2023; 10:28. [PMID: 38247752 PMCID: PMC10815488 DOI: 10.3390/gels10010028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Hydrogel microparticles (HMPs) stand out as promising entities in the realm of bone tissue regeneration, primarily due to their versatile capabilities in delivering cells and bioactive molecules/drugs. Their significance is underscored by distinct attributes such as injectability, biodegradability, high porosity, and mechanical tunability. These characteristics play a pivotal role in fostering vasculature formation, facilitating mineral deposition, and contributing to the overall regeneration of bone tissue. Fabricated through diverse techniques (batch emulsion, microfluidics, lithography, and electrohydrodynamic spraying), HMPs exhibit multifunctionality, serving as vehicles for drug and cell delivery, providing structural scaffolding, and functioning as bioinks for advanced 3D-printing applications. Distinguishing themselves from other scaffolds like bulk hydrogels, cryogels, foams, meshes, and fibers, HMPs provide a higher surface-area-to-volume ratio, promoting improved interactions with the surrounding tissues and facilitating the efficient delivery of cells and bioactive molecules. Notably, their minimally invasive injectability and modular properties, offering various designs and configurations, contribute to their attractiveness for biomedical applications. This comprehensive review aims to delve into the progressive advancements in HMPs, specifically for bone regeneration. The exploration encompasses synthesis and functionalization techniques, providing an understanding of their diverse applications, as documented in the existing literature. The overarching goal is to shed light on the advantages and potential of HMPs within the field of engineering bone tissue.
Collapse
Affiliation(s)
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA;
| |
Collapse
|
97
|
Baines DK, Platania V, Tavernaraki NN, Parati M, Wright K, Radecka I, Chatzinikolaidou M, Douglas TEL. The Enrichment of Whey Protein Isolate Hydrogels with Poly-γ-Glutamic Acid Promotes the Proliferation and Osteogenic Differentiation of Preosteoblasts. Gels 2023; 10:18. [PMID: 38247741 PMCID: PMC10815088 DOI: 10.3390/gels10010018] [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: 11/23/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Osseous disease accounts for over half of chronic pathologies, but there is a limited supply of autografts, the gold standard; hence, there is a demand for new synthetic biomaterials. Herein, we present the use of a promising, new dairy-derived biomaterial: whey protein isolate (WPI) in the form of hydrogels, modified with the addition of different concentrations of the biotechnologically produced protein-like polymeric substance poly-γ-glutamic acid (γ-PGA) as a potential scaffold for tissue regeneration. Raman spectroscopic analysis demonstrated the successful creation of WPI-γ-PGA hydrogels. A cytotoxicity assessment using preosteoblastic cells demonstrated that the hydrogels were noncytotoxic and supported cell proliferation from day 3 to 14. All γ-PGA-containing scaffold compositions strongly promoted cell attachment and the formation of dense interconnected cell layers. Cell viability was significantly increased on γ-PGA-containing scaffolds on day 14 compared to WPI control scaffolds. Significantly, the cells showed markers of osteogenic differentiation; they synthesised increasing amounts of collagen over time, and cells showed significantly enhanced alkaline phosphatase activity at day 7 and higher levels of calcium for matrix mineralization at days 14 and 21 on the γ-PGA-containing scaffolds. These results demonstrated the potential of WPI-γ-PGA hydrogels as scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Daniel K. Baines
- Faculty of Science and Technology, School of Engineering, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
- Faculty of Health and medicine, Division of Biomedical and Life Sciences, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| | - Varvara Platania
- Department of Materials Science and Technology, University of Crete, GR-70013 Heraklion, Greece; (V.P.); (N.N.T.); (M.C.)
| | - Nikoleta N. Tavernaraki
- Department of Materials Science and Technology, University of Crete, GR-70013 Heraklion, Greece; (V.P.); (N.N.T.); (M.C.)
| | - Mattia Parati
- Faculty of Science and Engineering, School of Life Sciences, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.P.); (I.R.)
| | - Karen Wright
- Faculty of Health and medicine, Division of Biomedical and Life Sciences, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| | - Iza Radecka
- Faculty of Science and Engineering, School of Life Sciences, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.P.); (I.R.)
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, GR-70013 Heraklion, Greece; (V.P.); (N.N.T.); (M.C.)
- Institute of Electronic Structure and Laser, Foundation for Research and Technology Hellas, GR-70013 Heraklion, Greece
| | - Timothy E. L. Douglas
- Faculty of Science and Technology, School of Engineering, Lancaster University, Gillow Avenue, Lancaster LA1 4YW, UK;
| |
Collapse
|
98
|
Prabhakaran V, Melchels FP, Murray LM, Paxton JZ. Engineering three-dimensional bone macro-tissues by guided fusion of cell spheroids. Front Endocrinol (Lausanne) 2023; 14:1308604. [PMID: 38169965 PMCID: PMC10758461 DOI: 10.3389/fendo.2023.1308604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction Bioassembly techniques for the application of scaffold-free tissue engineering approaches have evolved in recent years toward producing larger tissue equivalents that structurally and functionally mimic native tissues. This study aims to upscale a 3-dimensional bone in-vitro model through bioassembly of differentiated rat osteoblast (dROb) spheroids with the potential to develop and mature into a bone macrotissue. Methods dROb spheroids in control and mineralization media at different seeding densities (1 × 104, 5 × 104, and 1 × 105 cells) were assessed for cell proliferation and viability by trypan blue staining, for necrotic core by hematoxylin and eosin staining, and for extracellular calcium by Alizarin red and Von Kossa staining. Then, a novel approach was developed to bioassemble dROb spheroids in pillar array supports using a customized bioassembly system. Pillar array supports were custom-designed and printed using Formlabs Clear Resin® by Formlabs Form2 printer. These supports were used as temporary frameworks for spheroid bioassembly until fusion occurred. Supports were then removed to allow scaffold-free growth and maturation of fused spheroids. Morphological and molecular analyses were performed to understand their structural and functional aspects. Results Spheroids of all seeding densities proliferated till day 14, and mineralization began with the cessation of proliferation. Necrotic core size increased over time with increased spheroid size. After the bioassembly of spheroids, the morphological assessment revealed the fusion of spheroids over time into a single macrotissue of more than 2.5 mm in size with mineral formation. Molecular assessment at different time points revealed osteogenic maturation based on the presence of osteocalcin, downregulation of Runx2 (p < 0.001), and upregulated alkaline phosphatase (p < 0.01). Discussion With the novel bioassembly approach used here, 3D bone macrotissues were successfully fabricated which mimicked physiological osteogenesis both morphologically and molecularly. This biofabrication approach has potential applications in bone tissue engineering, contributing to research related to osteoporosis and other recurrent bone ailments.
Collapse
Affiliation(s)
- Vinothini Prabhakaran
- Anatomy@Edinburgh, Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Ferry P.W. Melchels
- School of Engineering and Physical Sciences, Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom
- Future Industries Institute, University of South Australia, Adelaide, SA, Australia
| | - Lyndsay M. Murray
- Anatomy@Edinburgh, Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer Z. Paxton
- Anatomy@Edinburgh, Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
99
|
Taher Mohamed SA, Emin N. Effects of using collagen and aloe vera grafted fibroin scaffolds on osteogenic differentiation of rat bone marrow mesenchymal stem cells in SBF-enriched cell culture medium. Biomed Mater 2023; 19:015011. [PMID: 38055984 DOI: 10.1088/1748-605x/ad12e2] [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: 08/18/2023] [Accepted: 12/06/2023] [Indexed: 12/08/2023]
Abstract
In the study, collagen and aloe vera were grafted onto silk fibroin with two different methods, and 3D-microporous scaffolds (1F5C4A1 and 2F5C4A1) were formed by lyophilization. Three osteogenic cultures were started by seeding rat bone marrow mesenchymal stem cells (MSCs) and pre-induced MSC (osteoblast (OB)) on biopolymeric scaffolds. The osteogenic medium was enriched with 10% (v/v) simulated body fluid (SBF) to promote mineralization and osteogenic differentiation in one of the MSC cultures and the OB culture. X-ray diffraction (XRD), scanning electron microscopy (SEM), scanning electron microscopy- energy dispersive spectrum (SEM-EDS) analyses on cellular samples and histochemical (alizarin red, safranin-O, alcian blue) and immunohistochemical (anti-collagen-1, anti-osteocalcin, anti-osteopontin) staining showed that bone-like mineralization was occurred by both chemically and cellular activity. In addition, pre-osteogenic induction of MSCs in 2D-cultured was found to promote osteogenesis more rapidly when started 3D-cultured. These results indicated that enrichment of the cell culture medium with SBF is sufficient forin vitromineralization rather than using high concentrations of SBF. The findings showed that OB cells on the 2F5C4A1 scaffold obtained the best osteogenic activity. Still, other culture media with 10% SBF content could be used for bone tissue engineering under osteogenic induction.
Collapse
Affiliation(s)
- Salma A Taher Mohamed
- Material Science and Engineering Department, Institute of Science and Technology, Kastamonu University, Kastamonu, Turkey
| | - Nuray Emin
- Material Science and Engineering Department, Institute of Science and Technology, Kastamonu University, Kastamonu, Turkey
- Biomedical Engineering Department, Engineering and Architecture Faculty, Kastamonu University, Kastamonu, Turkey
| |
Collapse
|
100
|
Albaqami M, Aguida B, Pourmostafa A, Ahmad M, Kishore V. Photobiomodulation effects of blue light on osteogenesis are induced by reactive oxygen species. Lasers Med Sci 2023; 39:5. [PMID: 38091111 DOI: 10.1007/s10103-023-03951-7] [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: 09/11/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
Blue light-mediated photobiomodulation (PBM) is a promising approach to promote osteogenesis. However, the underlying mechanisms of PBM in osteogenesis are poorly understood. In this study, a human osteosarcoma cell line (i.e., Saos-2 cells) was subjected to intermittent blue light exposure (2500 µM/m2/s, 70 mW/cm2, 4.2 J/cm2, once every 48 h) and the effects on Saos-2 cell viability, metabolic activity, differentiation, and mineralization were investigated. In addition, this study addressed a possible role of blue light induced cellular oxidative stress as a mechanism for enhanced osteoblast differentiation and mineralization. Results showed that Saos-2 cell viability and metabolic activity were maintained upon blue light exposure compared to unilluminated controls, indicating no negative effects. To the contrary, blue light exposure significantly increased (p < 0.05) alkaline phosphatase activity and Saos-2 cell mediated mineralization. High-performance liquid chromatography (HPLC) assay was used for measurement of reactive oxygen species (ROS) activity and showed a significant increase (p < 0.05) in superoxide (O2•-) and hydrogen peroxide (H2O2) formed after blue light exposure. Together, these results suggest that the beneficial effects of blue light-mediated PBM on osteogenesis may be induced by controlled release of ROS.
Collapse
Affiliation(s)
- Maria Albaqami
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 W. University Blvd, Melbourne, FL, 32901, USA
| | - Blanche Aguida
- UMR8256, CNRS, IBPS, Sorbonne, Université, Paris, France
| | - Ayda Pourmostafa
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 W. University Blvd, Melbourne, FL, 32901, USA
| | - Margaret Ahmad
- UMR8256, CNRS, IBPS, Sorbonne, Université, Paris, France
| | - Vipuil Kishore
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, 150 W. University Blvd, Melbourne, FL, 32901, USA.
| |
Collapse
|