1
|
Bernardo MP, Foschini M, Santos ACC, Ueira Vieira C, Saito N, Mundim MEC, Lopes OF, Pasquini D. Fabrication of Antimicrobial Cellulose and Silver Niobate Aerogels for Enhanced Tissue Regeneration. ACS OMEGA 2025; 10:15493-15502. [PMID: 40290954 PMCID: PMC12019755 DOI: 10.1021/acsomega.5c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
Aging, trauma, infection, illness, and accidents can lead to the disruption of various human tissues, including skin, bone, and cartilage. Tissue engineering aims to promote the growth of cells and tissues within the human body, with scaffolds serving as vehicles to deliver a combination of mechanical and molecular signals to create new tissues for body reconstruction. Composite materials have gained significant attention as an attractive alternative for scaffolding due to their ability to enhance multiple material properties. For instance, cellulose nanofibers are known for their high specific surface area, flexibility, and elasticity. However, their limited bioactivity and slow degradation rates restrict their suitability for tissue engineering applications. In contrast, niobium-based materials, which are biocompatible and nontoxic, have been underexplored in this field. In this study, silver niobate is investigated for the first time as a component of a composite material designed to provide biological activity to an aerogel, thereby creating a multifunctional scaffold for tissue regeneration. Silver niobate nanoparticles were successfully synthesized and characterized by X-ray diffraction (XRD), Fourier transform infrared (FTIR), and scanning electron microscopy (SEM). The composite aerogels demonstrated improved thermal stability, hydrophilicity, bioactivity, and antimicrobial activity against Staphylococcus aureus. Additionally, the developed aerogels showed no cytotoxic effects on primary dermal fibroblast (HDFn) cells. These findings suggest that the silver niobate-based aerogel composite holds significant potential for applications in tissue regeneration, offering a promising avenue for the development of advanced biomaterials in regenerative medicine.
Collapse
Affiliation(s)
- Marcela Piassi Bernardo
- Institute
of Chemistry, Federal University of Uberlândia, Av João Naves de Ávila, Uberlândia, MG CEP 38400-902, Brazil
| | - Mauricio Foschini
- Physics
Institute, Federal University of Uberlandia, Av João Naves de Ávila, Uberlândia, MG CEP 38400-902, Brazil
| | - Ana Carolina Costa Santos
- Genetics
Laboratory, Institute of Biotechnology, Federal University of Uberlandia, Rua Ceará, Uberlândia, MG CEP: 38402-018, Brazil
| | - Carlos Ueira Vieira
- Genetics
Laboratory, Institute of Biotechnology, Federal University of Uberlandia, Rua Ceará, Uberlândia, MG CEP: 38402-018, Brazil
| | - Natieli Saito
- Nanobiotechnology
Prof. Dr. Luiz Ricardo Goulart Filho Laboratory, Institute of Biotechnology, Federal University of Uberlandia, Rua Ceará, Uberlândia, MG CEP: 38402-018, Brazil
| | - Maria Eduarda Costa Mundim
- Nanobiotechnology
Prof. Dr. Luiz Ricardo Goulart Filho Laboratory, Institute of Biotechnology, Federal University of Uberlandia, Rua Ceará, Uberlândia, MG CEP: 38402-018, Brazil
| | - Osmando Ferreira Lopes
- Institute
of Chemistry, Federal University of Uberlândia, Av João Naves de Ávila, Uberlândia, MG CEP 38400-902, Brazil
| | - Daniel Pasquini
- Institute
of Chemistry, Federal University of Uberlândia, Av João Naves de Ávila, Uberlândia, MG CEP 38400-902, Brazil
| |
Collapse
|
2
|
Ujjwal RR, Slaughter G. Advances in Bacterial Cellulose-Based Scaffolds for Tissue Engineering: Review. J Biomed Mater Res A 2025; 113:e37912. [PMID: 40233003 DOI: 10.1002/jbm.a.37912] [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: 01/23/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025]
Abstract
Bacterial cellulose (BC) has emerged as a highly versatile and promising biomaterial in tissue engineering, with potential applications across skin, bone, cartilage, and vascular regeneration. Its exceptional properties like high mechanical strength, superior biocompatibility, excellent moisture retention, and inherent ability to support cell adhesion and proliferation, make BC particularly effective for wound healing and skin regeneration. These attributes accelerate tissue repair and foster new tissue formation, highlighting its value in skin-related applications. Additionally, BC's capacity to support osteogenic differentiation, combined with its mechanical robustness, positions it as a strong candidate for bone tissue engineering, facilitating regeneration and repair. Recent advancements have emphasized the development of BC-based hybrid scaffolds to enhance tissue-specific functionalities, including vascularization and cartilage regeneration. These innovations aim to address the complex requirements of various tissue engineering applications. However, challenges remain, particularly regarding the scalability of BC production, cost-effectiveness, and the long-term stability of BC-based scaffolds. Such barriers continue to limit its broader clinical adoption. This review critically examines the synthesis methods, intrinsic properties, and recent innovations in the design of BC-based scaffolds, offering insights into their potential to revolutionize regenerative medicine. Furthermore, it addresses the key challenges and limitations that must be overcome to enable the clinical integration of BC. By addressing these limitations, BC could play a transformative role in advancing tissue engineering and regenerative therapies, bridging the gap between laboratory research and clinical application.
Collapse
Affiliation(s)
- Rewati Raman Ujjwal
- Center for Bioelectronics, Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, Virginia, USA
| | - Gymama Slaughter
- Center for Bioelectronics, Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, Virginia, USA
| |
Collapse
|
3
|
Dawood RM, Mahdee AF. Fabrication and characterization of 3D-printed polymeric-based scaffold coated with bioceramic and naringin for a potential use in dental pulp regeneration (in vitro study). Int Endod J 2025; 58:627-642. [PMID: 39815625 DOI: 10.1111/iej.14194] [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: 06/21/2024] [Revised: 10/12/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025]
Abstract
AIM 3D-printed scaffolds loaded with healing directed agents could be employed for better treatment outcome in regenerative dentistry. The aim of this study was to fabricate and characterize simple 3D-printed poly lactic acid (PLA) scaffolds coated with nanoHydroxyapatite (nHA), Naringin (NAR), or their combination, and testing their morphological, chemical, mechanical, antibacterial, biocompatible and bioactive properties. METHODOLOGY Two variants pore sizes, 300 and 700 μm, of 3D-printed PLA disc scaffolds measuring (10 × 1 mm) were fabricated. These scaffolds were dip-coated with nHA, NAR, or both (nHA/NAR). Field emission scanning electron microscopy (FeSEM), energy-dispersive X-ray spectroscopy (EDX), Fourier transforms infrared (FTIR), compressive and flexural strength testing was employed for optimizing pore size. Then, antibacterial activity against isolated Streptococcus mutans and Enterococcus faecalis, and cytotoxicity against normal human fibroblast were assessed. Additionally, appetite formation on scaffold surfaces was assessed after storage in simulated body fluid (SBF) for 14 days by further using FeSEM, EDX and XRD. RESULTS FeSEM showed uniform structure for 3D-printed scaffolds in both pore size designs, and a consistent surface coating with nHA and NAR, which were further confirmed by EDX and FTIR. However, mechanical testing revealed statistical significant higher compressive and flexural strengths (p < .000) for 300 μm pore size scaffolds. Statistical significant antibacterial activities (p < .05) were also found with PLA/NAR, and PLA/nHA /NAR scaffolds in comparison with neat. The MTT assay revealed biocompatibility of PLA, nHA and NAR, with the combinations of the latter two working synergistically. Lastly, the formation of a calcium-phosphate appetite layer was recognized on the surface of PLA/nHA, PLA/nHA/NAR scaffold after being stored in SBF. CONCLUSIONS 3D-printed, 300 μm pore size, PLA scaffold coated with a combination of nHA and NAR showed the best surface characteristics and improved mechanical, antibacterial and biocompatible properties for further investigation in regenerative studies.
Collapse
Affiliation(s)
- Reem Mones Dawood
- Restorative and Aesthetic Dentistry Department, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Anas Falah Mahdee
- Restorative and Aesthetic Dentistry Department, College of Dentistry, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
4
|
Drake KA, Grubelich TA, Wong S, Deymier AC. A methodological comparison of synthesizing heavy metal substituted bioapatite. Methods 2025; 239:42-48. [PMID: 40147604 DOI: 10.1016/j.ymeth.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
This study evaluates two methods-maturation and direct precipitation-for synthesizing heavy metal substituted biomimetic hydroxyapatite (HA), focusing on their efficacy in mimicking human bone composition and crystallinity. Cobalt (Co) and chromium (Cr) substitutions were investigated due to their relevance to metal-on-metal implant degradation and the potential integration of these ions into bone mineral. The maturation method involves prolonged incubation, producing amorphous and bioresorbable apatites, while the direct precipitation (DP) method achieves rapid synthesis of highly crystalline apatites through controlled titration. Both approaches were characterized using X-ray diffraction (XRD), Raman spectroscopy, and Fourier Transform Infrared (FTIR) spectroscopy, confirming the apatitic nature of the samples and lattice strain induced by metal ion substitution. This study highlights the maturation method's adaptability for long-term biological interactions and the DP method's mechanical stability for load-bearing applications. Comparison of the structural and chemical properties of substituted HA from each method provides insights into optimizing synthesis techniques for diverse biomedical applications, such as bone tissue engineering and mitigating the effects of heavy metal ion release on bone health. These findings contribute to advancing hydroxyapatite-based biomaterials tailored for therapeutic and regenerative medicine needs.
Collapse
Affiliation(s)
- Kennedy A Drake
- Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Tyler A Grubelich
- Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Stephanie Wong
- Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Alix C Deymier
- Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
5
|
Wang J, Zhang Q, Wang H, Liu C, Jiang L, Liu W, Wu Y, Wang Y, Vivian, Yan H, Lin J, Sun X. A Sr@Ag-based spatiotemporal and step-release scaffold against chronic osteomyelitis, fabricated by coaxial 3D-printing. Biomaterials 2025; 314:122899. [PMID: 39437580 DOI: 10.1016/j.biomaterials.2024.122899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
Chronic osteomyelitis (OM) represents a severe and persistent infectious bone disease. Effective treatment requires controlled anti-inflammatory releases and bone regeneration across disease phases. A Sr@Ag-based scaffold was successfully printed, featuring micron-scale coaxial fibers containing Ag-doped hydroxyapatite (HA) in the outer layer of PLLA and Sr-doped HA in the inner layer of PLLA, facilitating the spatiotemporal and sequential release of Ag and Sr ions during OM treatment. Most antibacterial agent (Ag) was released during the first 20 days, followed by a slow-release plateau over the next 40 days in phosphate-buffered saline solution (PBS). Meanwhile, the pro-angiogenic agent (Sr) was released in minimal amounts during the initial 20 days, followed by a rapid and considerable release in the following 40 days. The coaxial design effectively inhibited the growth of Staphylococcus aureus and Escherichia coli while preserving the viability of bone cells. The ion-based scaffold exhibited broad-spectrum antibacterial effects and enhanced bone-regenerating gene expression in a complex air-bacteria environment. The Sr@Ag-based coaxial scaffold demonstrated effective antibacterial activity during the early stage and exhibited excellent non-toxic bone regeneration results during the middle and late stages in vivo. This work offered a promising treatment strategy through sequential anti-inflammatory and pro-osteogenic effects for infectious bone-defect diseases.
Collapse
Affiliation(s)
- Jingyun Wang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Qin Zhang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China; Department of Orthopaedics, First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Hetong Wang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Chunlin Liu
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; School of Mechanical Engineering, Chengdu University, Chengdu, Sichuan, 610000, People's Republic of China.
| | - Le Jiang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wanting Liu
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yixian Wu
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Yifan Wang
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Vivian
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Hao Yan
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China
| | - Jun Lin
- Department of Orthopaedics, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China; Department of Orthopaedics, First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215000, People's Republic of China.
| | - Xiaodan Sun
- Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China; Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China.
| |
Collapse
|
6
|
Cota Quintero JL, Ramos-Payán R, Romero-Quintana JG, Ayala-Ham A, Bermúdez M, Aguilar-Medina EM. Hydrogel-Based Scaffolds: Advancing Bone Regeneration Through Tissue Engineering. Gels 2025; 11:175. [PMID: 40136878 PMCID: PMC11942283 DOI: 10.3390/gels11030175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Bone tissue engineering has emerged as a promising approach to addressing the limitations of traditional bone grafts for repairing bone defects. This regenerative medicine strategy leverages biomaterials, growth factors, and cells to create a favorable environment for bone regeneration, mimicking the body's natural healing process. Among the various biomaterials explored, hydrogels (HGs), a class of three-dimensional, hydrophilic polymer networks, have gained significant attention as scaffolds for bone tissue engineering. Thus, this review aimed to investigate the potential of natural and synthetic HGs, and the molecules used for its functionalization, for enhanced bone tissue engineering applications. HGs offer several advantages such as scaffolds, including biocompatibility, biodegradability, tunable mechanical properties, and the ability to encapsulate and deliver bioactive molecules. These properties make them ideal candidates for supporting cell attachment, proliferation, and differentiation, ultimately guiding the formation of new bone tissue. The design and optimization of HG-based scaffolds involve adapting their composition, structure, and mechanical properties to meet the specific requirements of bone regeneration. Current research focuses on incorporating bioactive molecules, such as growth factors and cytokines, into HG scaffolds to further enhance their osteoinductive and osteoconductive properties. Additionally, strategies to improve the mechanical strength and degradation kinetics of HGs are being explored to ensure long-term stability and support for new bone formation. The development of advanced HG-based scaffolds holds great potential for revolutionizing bone tissue engineering and providing effective treatment options for patients with bone defects.
Collapse
Affiliation(s)
- Juan Luis Cota Quintero
- Faculty of Biology, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico;
| | - Rosalío Ramos-Payán
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico; (R.R.-P.); (J.G.R.-Q.)
| | - José Geovanni Romero-Quintana
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico; (R.R.-P.); (J.G.R.-Q.)
| | - Alfredo Ayala-Ham
- Faculty of Odontology, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico;
| | - Mercedes Bermúdez
- Faculty of Odontology, Autonomous University of Chihuahua, Circuito Universitario Campus I, Chihuahua 31000, Chihuahua, Mexico;
| | - Elsa Maribel Aguilar-Medina
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico; (R.R.-P.); (J.G.R.-Q.)
| |
Collapse
|
7
|
Ghabour O, Taha N, Aboul Gheit S, Mohy El Din M. Evaluation of magnesium-based scaffolds fabricated using a modified sintering technique and two types of space holding agents (in vitro study). BDJ Open 2025; 11:20. [PMID: 39979289 PMCID: PMC11842699 DOI: 10.1038/s41405-025-00299-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/15/2024] [Accepted: 12/31/2024] [Indexed: 02/22/2025] Open
Abstract
OBJECTIVE The aim of this work was to study the mechanical, degradation behavior and bioactivity of porous magnesium-based scaffolds alloyed with zinc and hydroxyapatite, fabricated using two different types of space holding agents and a modified powder metallurgy route. METHODS Powder particles of magnesium, zinc, hydroxyapatite (HA) and spacers were mixed, then mixtures were divided into 6 groups: IA (urea/0%HA), IB (urea/5%HA), IC (urea/7.5%HA), IIA (ammonium bicarbonate/0%HA), IIB (ammonium bicarbonate/5%HA) and IIC (ammonium bicarbonate/7.5%HA). A modified powder metallurgy route was used to fabricate the composites. Porosity analysis and microstructural characterization using Scanning Electron Microscope (SEM), Energy Dispersive X-ray Analysis (EDX), and X-ray Diffraction Analysis (XRD) were done. Evaluation of mechanical properties, in-vitro degradation rate in simulated body fluid (SBF) and in-vitro bioactivity using SEM and XRD were done. Data were statistically analyzed using two-way and three-way repeated ANOVA tests. RESULTS All scaffolds were found to be highly porous. Significant differences were observed regarding mechanical properties, degradation rate and concentration of magnesium released during degradation (P < 0.0001). The results showed that group IIB had the lowest strength and fastest corrosion rate, while IB had the highest strength and elastic modulus and the slowest corrosion rate among all groups. Bioactivity evaluation revealed extensive formation of calcium phosphate crystals and precipitations covering the scaffolds' surfaces. CONCLUSION This study showed that using up to 5% HA as a reinforcing element with moderate compaction pressure and urea as a space holding agent can result in the fabrication of magnesium scaffolds suitable for orthopedic applications.
Collapse
Affiliation(s)
- Omnia Ghabour
- Master's Student, Department of Dental Biomaterials, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.
- Teaching Assistant of Dental Biomaterials, Department of Dental Biomaterials, College of Dentistry, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El-Alamein, Egypt.
| | - Nahla Taha
- Modeling and Simulation Department, Advanced Technology and New Materials Research Institute, City of Scientific Research and Technological Applications, New Borg Al-Arab City, Alexandria, Egypt
| | - Salma Aboul Gheit
- Department of Dental Biomaterials Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Mona Mohy El Din
- Department of Dental Biomaterials Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| |
Collapse
|
8
|
Kim YH, Kim HS, Hong IS. Advances in biomaterials-based tissue engineering for regeneration of female reproductive tissues. Biofabrication 2025; 17:022001. [PMID: 39854843 DOI: 10.1088/1758-5090/adae38] [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: 06/24/2024] [Accepted: 01/24/2025] [Indexed: 01/27/2025]
Abstract
The anatomical components of the female reproductive system-comprising the ovaries, uterus, cervix, vagina, and fallopian tubes-interact intricately to provide the structural and hormonal support essential for reproduction. However, this system is susceptible to various detrimental factors, both congenital and acquired, that can impair fertility and adversely affect quality of life. Recent advances in bioengineering have led to the development of sophisticated three-dimensional models that mimic the complex architecture and functionality of reproductive organs. These models, incorporating diverse cell types and tissue layers, are crucial for understanding physiological processes within the reproductive tract. They offer insights into decidualization, ovulation, folliculogenesis, and the progression of reproductive cancers, thereby enhancing personalized medical treatments and addressing female infertility. This review highlights the pivotal role of tissue engineering in diagnosing and treating female infertility, emphasizing the importance of considering factors like biocompatibility, biomaterial selection, and mechanical properties in the design of bioengineered systems. The challenge of replicating the functionally specialized and structurally complex organs, such as the uterus and ovary, underscores the need for reliable techniques that improve morphological and functional restoration. Despite substantial progress, the goal of creating a fully artificial female reproductive system is still a challenge. Nonetheless, the recent fabrication of artificial ovaries, uteruses, cervixes, and vaginas marks significant advancements toward this aim. Looking forward, the challenges in bioengineering are expected to spur further innovations in both basic and applied sciences, potentially hastening the clinical adoption of these technologies.
Collapse
Affiliation(s)
- Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| |
Collapse
|
9
|
Thangavel M, Elsen S R. Evaluation and optimization of physical, mechanical, and biological characteristics of 3D printed Whitlockite/calcium silicate composite scaffold for bone tissue regeneration using response surface methodology. Biomed Mater 2025; 20:025017. [PMID: 39842082 DOI: 10.1088/1748-605x/adad27] [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: 08/29/2024] [Accepted: 01/22/2025] [Indexed: 01/24/2025]
Abstract
Calcium phosphate-based bioscaffolds are used for bone tissue regeneration because of their physical and chemical resemblance to human bone. Calcium, phosphate, sodium, potassium, magnesium, and silicon are important components of human bone. The successful biomimicking of human bone characteristics involves incorporating all the human bone elements into the scaffold material. In this work, Mg-Whitlockite (WH) and Calcium Silicate (CS) were selected as matrix and reinforcement respectively, because of their desirable elemental composition and regenerative properties. The magnesium in WH increases mineralization in bone, and the silicon ions in CS support vascularization. The Mg-WH was synthesized using the wet chemical method, and powder characterization tests were performed. Response surface methodology (RSM) is used to design the experiments with a combination of material compositions, infill ratios (IFs), and sintering temperatures (STs). The WH/CS bioceramic composite is 3D printed in three different compositions: 100/0, 75/25, and 50/50 wt%, with IFs of 50%, 75%, and 100%. The physical and mechanical characterization study of printed samples is conducted and the result is optimized using RSM. ANOVA (Analysis of Variance) is used to establish the relationship between input parameters and responses. The optimized input parameters were the WH/CS composition of 50/50 wt%, IF of 50%, and ST of 1150 °C, which bring out the best possible combination of physical and mechanical characteristics. The RSM optimized response was a density of 2.27 g cm-3, porosity of 36.74%, wettability of 45.79%, shrinkage of 25.13%, compressive strength of 12 MPa, and compressive modulus of 208.49 MPa with 92% desirability. The biological characterization studies were conducted for the scaffold samples prepared with optimized input parameters. The biological studies confirmed the capabilities of the WH/CS composite scaffolds in bone regenerative applications.
Collapse
Affiliation(s)
- Mahendran Thangavel
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Renold Elsen S
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
10
|
Li N, Wang M, Luo H, Tse SD, Gao Y, Zhu Z, Guo H, He L, Zhu C, Yin K, Sun L, Guo J, Hong H. Processing and properties of graphene-reinforced polylactic acid nanocomposites for bioelectronic and tissue regenerative functions. BIOMATERIALS ADVANCES 2025; 167:214113. [PMID: 39549370 DOI: 10.1016/j.bioadv.2024.214113] [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: 07/04/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
An in-situ polymer-solution-processing approach enables the efficient production of uniform graphene-reinforced polylactic acid (G-PLA) nanocomposites with notable physical and biomedical properties. The approach effectively enhances the interfacial bonding between graphene and PLA by creating graphene dangling bonds and defects during exfoliation. As a result, an 182 % increase in Young's modulus and an 85 % increase in tensile strength can be achieved in G-PLA. Only 0.5 wt% graphene addition can reduce the contact angle of the composite from 75.3 to 70.4 and reduce its oxygen permeability by 23 %. The improved hydrophilicity, hermeticity, and mechanical properties make G-PLA an excellent encapsulation material for implantable bioelectronics. Moreover, the composite surface attributes and cell behaviors at the material-tissue interface are investigated histologically through the culture of stem cells on as-synthesized G-PLA. G-PLA composites can significantly boost cell proliferation and regulate cell differentiation towards vascular endothelium, offering tissue regeneration at the surface of implants to recover the injured tissues. The degradation rate of G-PLA nanocomposite can also be regulated since the graphene slows down the autocatalytic chain splitting induced by the terminal carboxylic acid groups of PLA. Therefore, such G-PLA nanocomposites with physical and biomedical properties regulated by graphene loading enable the development of next-generation implantable electronic systems providing both sensing and tissue engineering functions for complicated applications such as implanted sensors monitoring the healing of fractured bones or intracranial pressure.
Collapse
Affiliation(s)
- Nan Li
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Mengjia Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, China
| | - Haoyu Luo
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Stephen D Tse
- Department of Mechanical and Aerospace Engineering, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Yun Gao
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, China
| | - Zhen Zhu
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Hongxuan Guo
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Longbing He
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Chao Zhu
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Kuibo Yin
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Litao Sun
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Jie Guo
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, China
| | - Hua Hong
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China.
| |
Collapse
|
11
|
Xiao L, Shimamura N, Kamio T, Ide R, Mochizuki M, Nakahara T. Polycarbonate-Acrylonitrile Butadiene Styrene Three Dimensional Printing Material Exhibits Biocompatibility and Enhances Osteogenesis and Gingival Tissue Formation with Human Cells. Cells 2025; 14:167. [PMID: 39936959 PMCID: PMC11816493 DOI: 10.3390/cells14030167] [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: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Three dimensional (3D) printing materials are widely used in dental applications, but their biocompatibility and interactions with human cells require evaluation. This study aimed to identify materials meeting biocompatibility, mechanical strength, and tissue-forming requirements for safe dental applications. We assessed the cytotoxicity of resins and thermoplastic filaments in human HaCaT keratinocytes, gingival fibroblasts (hGFs), and stem cells from human exfoliated deciduous teeth (SHED) using PrestoBlue assays. Three resins, including two types of surgical guide resins, exhibited strong cytotoxicity after 4-72 h, while 2 h exposure to an FDA-approved surgical guide resin did not affect SHED cell viability. In contrast, six thermoplastic filaments showed no significant cytotoxicity even after 72 h. Among these, polycarbonate-acrylonitrile butadiene styrene (PC-ABS) demonstrated excellent toughness, heat resistance, and surface quality at a low cost. SHED cells cultured on PC-ABS dishes and micro bone structures showed strong proliferation and osteogenic potential. Culture inserts made of PC-ABS also supported the growth of HaCaT keratinocytes and the hGFs formed gingival tissue, which was superior to that formed on commercially available PET inserts. In conclusion, PC-ABS is a promising 3D printing material for dental applications due to its biocompatibility, ability to promote osteogenesis, and support for gingival tissue formation, with no observed cytotoxicity.
Collapse
Affiliation(s)
- Li Xiao
- Department of Physiology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan;
| | - Naohiro Shimamura
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan;
| | - Takashi Kamio
- Department of Oral and Maxillofacial Radiology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan;
| | - Ryoji Ide
- Department of Physiology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan;
| | - Mai Mochizuki
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan; (M.M.); (T.N.)
- Department of Life Science Dentistry, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan; (M.M.); (T.N.)
| |
Collapse
|
12
|
Zhu S, Sun H, Mu T, Richel A. Research Progress in 3D Printed Biobased and Biodegradable Polyester/Ceramic Composite Materials: Applications and Challenges in Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2791-2813. [PMID: 39760202 DOI: 10.1021/acsami.4c15719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Transplantation of bone implants is currently recognized as one of the most effective means of treating bone defects. Biobased and biodegradable polyester composites combine the good mechanical and degradable properties of polyester, thereby providing an alternative for bone implant materials. Bone tissue engineering (BTE) accelerates bone defect repair by simulating the bone microenvironment. Composite scaffolds support bone formation and further accelerate the process of bone repair. The introduction of 3D printing technology enables the preparation of scaffolds to be more precise, reproducible, and flexible, which is a very promising development. This review presents the physical properties of BTE scaffolds and summarizes the strategies adopted by domestic and international scholars to improve the properties of scaffolds based on biobased and biodegradable polyester/ceramic composites in recent years. In addition, future development prospects in the field and the challenges of expanding production in clinical applications are presented.
Collapse
Affiliation(s)
- Shunshun Zhu
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences; Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
- Laboratory of Biomass and Green Technologies, Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés, 2, 5030 Gembloux, Belgium
| | - Hongnan Sun
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences; Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
| | - Taihua Mu
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences; Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
| | - Aurore Richel
- Laboratory of Biomass and Green Technologies, Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés, 2, 5030 Gembloux, Belgium
| |
Collapse
|
13
|
Han Q, Zhao D, Wang X, Shang M, Zhou W, Li Q, Song H. Composite barrier membrane for bone regeneration: advancing biomaterial strategies in defect repair. RSC Adv 2025; 15:1290-1299. [PMID: 39816171 PMCID: PMC11733738 DOI: 10.1039/d4ra07623k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/01/2025] [Indexed: 01/18/2025] Open
Abstract
Bone defects represent a significant challenge in clinical practice, driving the need for innovative solutions that effectively support bone regeneration. Barrier membranes, due to playing a critical role in creating an environment conducive to bone regeneration by preventing the infiltration of non-osteogenic tissues, are widely applied to bone repair. However, inadequate spatial stability and osteogenesis-promoting ability often limit current barrier membranes. In response to these challenges, we have developed an advanced gelatin methacrylate/hydroxyapatite/hydroxyapatite membrane (GelMA/HAp/HAM) composite biomaterial designed as a barrier membrane with superior spatial stability and optimal degradation properties. The GelMA/HAp/HAM composite features a bilayer structure, with each layer possessing distinct properties: the dense hydroxyapatite membrane (HAM) acts as a barrier to prevent connective tissue infiltration. In contrast, the porous gelatin methacrylate/hydroxyapatite (GelMA/HAp) hydrogel layer promotes osteogenesis. Studies have demonstrated the composite's excellent biocompatibility and its significant osteogenic differentiation enhancement. This composite membrane holds great promise for clinical applications in bone defect repair, providing a new avenue for improving patient outcomes in regenerative medicine.
Collapse
Affiliation(s)
- Qingbin Han
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
- Department of Oral and Maxillofacial Surgery, Linyi People's Hospital Lin'yi 276000 China
| | - Delu Zhao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
| | - Xiaohong Wang
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
| | - Mengyao Shang
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
| | - Wenbin Zhou
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
| | - Qing Li
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
| | - Hui Song
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneratioon, Shandong Provincial Clinical Research Center for Oral Diseases Ji'nan 250012 China
| |
Collapse
|
14
|
Nayak VV, Bergamo ETP, Sanjairaj V, Behera RK, Gupta N, Coelho PG, Witek L. Effect of bioceramic inclusions on gel-cast aliphatic polymer membranes for bone tissue engineering applications: An in vitro study. Biomed Mater Eng 2025; 36:15-33. [PMID: 39331087 DOI: 10.3233/bme-240079] [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: 09/28/2024]
Abstract
BACKGROUND Polylactic acid (PLA) has been extensively used in tissue engineering. However, poor mechanical properties and low cell affinity have limited its pertinence in load bearing bone tissue regeneration (BTR) devices. OBJECTIVE Augmenting PLA with β-Tricalcium Phosphate (β-TCP), a calcium phosphate-based ceramic, could potentially improve its mechanical properties and enhance its osteogenic potential. METHODS Gels of PLA and β-TCP were prepared of different % w/w ratios through polymer dissolution in acetone, after which polymer-ceramic membranes were synthesized using the gel casting workflow and subjected to characterization. RESULTS Gel-cast polymer-ceramic constructs were associated with significantly higher osteogenic capacity and calcium deposition in differentiated osteoblasts compared to pure polymer counterparts. Immunocytochemistry revealed cell spreading over the gel-cast membrane surfaces, characterized by trapezoidal morphology, distinct rounded nuclei, and well-aligned actin filaments. However, groups with higher ceramic loading expressed significantly higher levels of osteogenic markers relative to pure PLA membranes. Rule of mixtures and finite element models indicated an increase in theoretical mechanical strength with an increase in β-TCP concentration. CONCLUSION This study potentiates the use of PLA/β-TCP composites in load bearing BTR applications and the ability to be used as customized patient-specific shape memory membranes in guided bone regeneration.
Collapse
Affiliation(s)
- Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Edmara T P Bergamo
- Biomaterials Division, NYU Dentistry, New York, NY, USA
- Department of Prosthodontics, NYU Dentistry, New York, NY, USA
| | | | - Rakesh Kumar Behera
- Department of Mechanical and Aerospace Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Nikhil Gupta
- Department of Mechanical and Aerospace Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Paulo G Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
- Hansjörg Wyss Department of Plastic Surgery, NYU Grossman School of Medicine, New York University, New York, NY, USA
| |
Collapse
|
15
|
Ksouri R, Odabas S, Yar Sağlam AS. Exosome loaded 3D printed magnetic PLA constructs: a candidate for bone tissue engineering. PROGRESS IN ADDITIVE MANUFACTURING 2025; 10:247-260. [DOI: 10.1007/s40964-024-00619-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/07/2024] [Indexed: 01/05/2025]
|
16
|
Mohammadi S, Ghasemi F, Alavi G SA, Alemzadeh E. Investigate the in vitro biocompatibility, biodegradation, cytotoxicity, and differentiation potential of 3-D gelatin-nanocellulose composite scaffolds loaded with nanohydroxyapatite and simvastatin. Tissue Cell 2024; 91:102536. [PMID: 39236521 DOI: 10.1016/j.tice.2024.102536] [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: 01/07/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/07/2024]
Abstract
Bone tissue engineering has been proposed as a promising solution for healing of bone fractures. An important aspect of bone tissue engineering is the implantable scaffolds that participate in the regeneration and repair of bone tissue. In this study, the composite scaffolds of gelatin- nanocellulose loaded with nanohydroxyapatite and simvastatin (as the osteoinductive component) were fabricated using freeze- drying method. Scaffolds were characterized in terms of morphology, mechanical, biodegradability, water absorption capacity, and simvastatin release characteristics. Also, the biocompatibility and differentiation potential of the scaffolds were evaluated on human bone marrow-derived mesenchymal stem cells using the MTT assay and alizarin red staining, respectively. The simvastatin loaded scaffolds showed a sustained release profile in vitro up to 216 h. The results of BMSCs differentiation by alizarin red staining showed significant differences between the simvastatin loaded group and other groups. Moreover, the results of MTT assay verified cytocompatibility and non-toxicity of the scaffolds. Therefore, the gelatin-nano cellulose composite scaffolds loaded with hydroxyapatite and simvastatin may be considered promising for use in bone tissue engineering.
Collapse
Affiliation(s)
- Soroush Mohammadi
- Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Fahimeh Ghasemi
- Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Esmat Alemzadeh
- Infectious Diseases Research Center, Department of Medical Biotechnology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
17
|
Łuczak JW, Palusińska M, Matak D, Pietrzak D, Nakielski P, Lewicki S, Grodzik M, Szymański Ł. The Future of Bone Repair: Emerging Technologies and Biomaterials in Bone Regeneration. Int J Mol Sci 2024; 25:12766. [PMID: 39684476 DOI: 10.3390/ijms252312766] [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: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Bone defects and fractures present significant clinical challenges, particularly in orthopedic and maxillofacial applications. While minor bone defects may be capable of healing naturally, those of a critical size necessitate intervention through the use of implants or grafts. The utilization of traditional methodologies, encompassing autografts and allografts, is constrained by several factors. These include the potential for donor site morbidity, the restricted availability of suitable donors, and the possibility of immune rejection. This has prompted extensive research in the field of bone tissue engineering to develop advanced synthetic and bio-derived materials that can support bone regeneration. The optimal bone substitute must achieve a balance between biocompatibility, bioresorbability, osteoconductivity, and osteoinductivity while simultaneously providing mechanical support during the healing process. Recent innovations include the utilization of three-dimensional printing, nanotechnology, and bioactive coatings to create scaffolds that mimic the structure of natural bone and enhance cell proliferation and differentiation. Notwithstanding the advancements above, challenges remain in optimizing the controlled release of growth factors and adapting materials to various clinical contexts. This review provides a comprehensive overview of the current advancements in bone substitute materials, focusing on their biological mechanisms, design considerations, and clinical applications. It explores the role of emerging technologies, such as additive manufacturing and stem cell-based therapies, in advancing the field. Future research highlights the need for multidisciplinary collaboration and rigorous testing to develop advanced bone graft substitutes, improving outcomes and quality of life for patients with complex defects.
Collapse
Affiliation(s)
- Julia Weronika Łuczak
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, Bldg. 23, 02-786 Warsaw, Poland
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Damian Matak
- European Biomedical Institute, 05-410 Jozefów, Poland
| | - Damian Pietrzak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Paweł Nakielski
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Sławomir Lewicki
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Pl. Żelaznej Bramy 10, 00-136 Warsaw, Poland
| | - Marta Grodzik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, Bldg. 23, 02-786 Warsaw, Poland
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
- European Biomedical Institute, 05-410 Jozefów, Poland
| |
Collapse
|
18
|
Singhatanadgit W, Sungkhaphan P, Thavornyutikarn B, Kitpakornsanti S, Young A, Janvikul W. In Vitro Osteo-Immunological Responses of Bioactive Calcium Phosphate-Containing Urethane Dimethacrylate-Based Composites: A Potential Alternative to Poly(methyl methacrylate) Bone Cement. ACS MATERIALS AU 2024; 4:612-627. [PMID: 39554857 PMCID: PMC11565289 DOI: 10.1021/acsmaterialsau.4c00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 11/19/2024]
Abstract
This investigation developed new composite bone cements using urethane dimethacrylate (UDMA), poly(propylene glycol) dimethacrylate (PPGDMA), and hydroxyethyl methacrylate (HEMA), with micrometer-sized aluminosilicate glass filler. Monocalcium phosphate monohydrate (MCPM) and hydroxyapatite (HA) particles were added to enhance biological performance, particularly osteo-immunomodulation. Free radical polymerization was triggered by mixing two pastes containing either benzoyl peroxide (BPO, an initiator) or N-tolyglycine glycidyl methacrylate (NTGGMA, an activator). Increasing butylated hydroxytoluene (BHT, an inhibitor) enabled a suitable delay after mixing at 25 °C for placement. At 37 °C, the delay time was reduced and the final conversion was enhanced. Findings also demonstrated the biocompatibility of the developed bone cement toward osteo-immunological cell lineages, including mesenchymal stem cells (MSCs), fibroblasts, osteoclast precursor RAW 246.7 cells, and peripheral blood mononuclear cells (PBMCs). Notably, the cement with both MCPM and HA combined facilitated sufficient MSC growth, enabling subsequent mineralization while concurrently suppressing the proliferation of fibroblasts, osteoclast progenitors, and PBMCs. Furthermore, composite cement exhibited the capacity to differentially regulate osteoblast differentiation, cell-(in)dependent mineralization, osteoclastogenesis, and PBMC-mediated inflammatory responses at both cellular and molecular levels in vitro. These observations suggested their potential use for bone repair, especially in cases of inflammation-associated bone defects.
Collapse
Affiliation(s)
- Weerachai Singhatanadgit
- Faculty
of Dentistry and Research Unit in Mineralized Tissue Reconstruction, Thammasat University (Rangsit Campus), Pathum-thani 12121, Thailand
| | - Piyarat Sungkhaphan
- National
Metal and Materials Technology Center, National
Science and Technology Development Agency, Khlong Luang 12120, Thailand
| | - Boonlom Thavornyutikarn
- National
Metal and Materials Technology Center, National
Science and Technology Development Agency, Khlong Luang 12120, Thailand
| | - Setthawut Kitpakornsanti
- Faculty
of Dentistry and Research Unit in Mineralized Tissue Reconstruction, Thammasat University (Rangsit Campus), Pathum-thani 12121, Thailand
| | - Anne Young
- Division
of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, Royal Free Hospital, Hampstead, London NW3 2PF, U.K.
| | - Wanida Janvikul
- National
Metal and Materials Technology Center, National
Science and Technology Development Agency, Khlong Luang 12120, Thailand
| |
Collapse
|
19
|
Ma Y, Wang Y, Tong S, Wang Y, Wang Z, Sui R, Yang K, Witte F, Yang S. Porous metal materials for applications in orthopedic field: A review on mechanisms in bone healing. J Orthop Translat 2024; 49:135-155. [PMID: 40226784 PMCID: PMC11993841 DOI: 10.1016/j.jot.2024.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 04/15/2025] Open
Abstract
Background Porous metal materials have been widely studied for applications in orthopedic field, owing to their excellent features and properties in bone healing. Porous metal materials with different compositions, manufacturing methods, and porosities have been developed. Whereas, the systematic mechanisms on how porous metal materials promote bone healing still remain unclear. Methods This review is concerned on the porous metal materials from three aspects with accounts of specific mechanisms, inflammatory regulation, angiogenesis and osteogenesis. We place great emphasis on different cells regulated by porous metal materials, including mesenchymal stem cells (MSCs), macrophages, endothelial cells (ECs), etc. Result The design of porous metal materials is diversified, with its varying pore sizes, porosity material types, modification methods and coatings help researchers create the most experimentally suitable and clinically effective scaffolds. Related signal pathways presented from different functions showed that porous metal materials could change the behavior of cells and the amount of cytokines, achieving good influence on osteogenesis. Conclusion This article summarizes the current progress achieved in the mechanism of porous metal materials promoting bone healing. By modulating the cellular behavior and physiological status of a spectrum of cellular constituents, such as macrophages, osteoblasts, and osteoclasts, porous metal materials are capable of activating different pathways and releasing regulatory factors, thus exerting pivotal influence on improving the bone healing effect. The translational potential of this article Porous metal materials play a vital role in the treatment of bone defects. Unfortunately, although an increasing number of studies have been concentrated on the effect of porous metal materials on osteogenesis-related cells, the comprehensive regulation of porous metal materials on the host cell functions during bone regeneration and the related intrinsic mechanisms remain unclear. This review summarizes different design methods for porous metal materials to fabricate the most suitable scaffolds for bone remodeling, and systematically reviews the corresponding mechanisms on inflammation, angiogenesis and osteogenesis of porous metal materials. This review can provide more theoretical framework and innovative optimization for the application of porous metal materials in orthopedics, dentistry, and other areas, thereby advancing their clinical utility and efficacy.
Collapse
Affiliation(s)
- Yutong Ma
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yi Wang
- The First Clinical College of China Medical University, Shenyang, 110001, China
| | - Shuang Tong
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yuehan Wang
- The First Clinical College of China Medical University, Shenyang, 110001, China
| | - Zhuoya Wang
- The First Clinical College of China Medical University, Shenyang, 110001, China
| | - Rongze Sui
- The First Clinical College of China Medical University, Shenyang, 110001, China
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Frank Witte
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, Charité Medical University, Assmannshauser Strasse 4–6, 14197, Berlin, Germany
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
20
|
da Silva VC, Gomes DDS, de Medeiros ELG, Santos AMDC, de Lima IL, Rosa TP, Rocha FS, Filice LDSC, Neves GDA, Menezes RR. Highly Porous 3D Nanofibrous Scaffold of Polylactic Acid/Polyethylene Glycol/Calcium Phosphate for Bone Regeneration by a Two-Step Solution Blow Spinning (SBS) Facile Route. Polymers (Basel) 2024; 16:3041. [PMID: 39518250 PMCID: PMC11548267 DOI: 10.3390/polym16213041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
This work presents the successful production of highly porous 3D nanofibrous hybrid scaffolds of polylactic acid (PLA)/polyethylene glycol (PEG) blends with the incorporation of calcium phosphate (CaP) bioceramics by a facile two-step process using the solution blow spinning (SBS) technique. CaP nanofibers were obtained at two calcium/phosphorus (Ca/P) ratios, 1.67 and 1.1, by SBS and calcination at 1000 °C. They were incorporated in PLA/PEG blends by SBS at 10 and 20 wt% to form 3D hybrid cotton-wool-like scaffolds. Morphological analysis showed that the fibrous scaffolds obtained had a randomly interconnected and highly porous structure. Also, the mean fiber diameter ranged from 408 ± 141 nm to 893 ± 496 nm. Apatite deposited considerably within 14 days in a simulated body fluid (SBF) test for hybrid scaffolds containing a mix of hydroxyapatite (HAp) and tri-calcium phosphate-β (β-TCP) phases. The scaffolds with 20 wt% CaP and a Ca/P ration of 1.1 showed better in vitro bioactivity to induce calcium mineralization for bone regeneration. Cellular tests evidenced that the developed scaffolds can support the osteogenic differentiation and proliferation of pre-osteoblastic MC3T3-E1 cells into mature osteoblasts. The results showed that the developed 3D scaffolds have potential applications for bone tissue engineering.
Collapse
Affiliation(s)
- Vanderlane Cavalcanti da Silva
- Graduate Program in Materials Science and Engineering, Department of Materials Engineering, Federal University of Campina Grande, Campina Grande 58429-900, Brazil;
- Laboratory of Materials Technology (LTM), Department of Materials Engineering, Federal University of Campina Grande, Av. Aprígio Veloso 882, Campina Grande 58429-900, Brazil; (E.L.G.d.M.); (G.d.A.N.)
| | - Déborah dos Santos Gomes
- Laboratory of Materials Technology (LTM), Department of Materials Engineering, Federal University of Campina Grande, Av. Aprígio Veloso 882, Campina Grande 58429-900, Brazil; (E.L.G.d.M.); (G.d.A.N.)
| | - Eudes Leonan Gomes de Medeiros
- Laboratory of Materials Technology (LTM), Department of Materials Engineering, Federal University of Campina Grande, Av. Aprígio Veloso 882, Campina Grande 58429-900, Brazil; (E.L.G.d.M.); (G.d.A.N.)
| | - Adillys Marcelo da Cunha Santos
- Center for Science and Technology in Energy and Sustainability (CETENS), Federal University of Recôncavo of Bahia, Feira de Santana 44042-280, Brazil;
| | - Isabela Lemos de Lima
- Nanobiotechnology Laboratory, Federal University of Uberlandia, Uberlandia 38408-100, Brazil; (I.L.d.L.); (T.P.R.); (L.d.S.C.F.)
| | - Taciane Pedrosa Rosa
- Nanobiotechnology Laboratory, Federal University of Uberlandia, Uberlandia 38408-100, Brazil; (I.L.d.L.); (T.P.R.); (L.d.S.C.F.)
| | - Flaviana Soares Rocha
- Department of Oral and Maxillofacial Surgery and Implantology, Federal University of Uberlandia, Uberlandia 38408-100, Brazil;
| | - Leticia de Souza Castro Filice
- Nanobiotechnology Laboratory, Federal University of Uberlandia, Uberlandia 38408-100, Brazil; (I.L.d.L.); (T.P.R.); (L.d.S.C.F.)
| | - Gelmires de Araújo Neves
- Laboratory of Materials Technology (LTM), Department of Materials Engineering, Federal University of Campina Grande, Av. Aprígio Veloso 882, Campina Grande 58429-900, Brazil; (E.L.G.d.M.); (G.d.A.N.)
| | - Romualdo Rodrigues Menezes
- Laboratory of Materials Technology (LTM), Department of Materials Engineering, Federal University of Campina Grande, Av. Aprígio Veloso 882, Campina Grande 58429-900, Brazil; (E.L.G.d.M.); (G.d.A.N.)
| |
Collapse
|
21
|
Raja S, Paschoalin RT, Terra IAA, Schalla C, Guimarães F, Periyasami G, Mattoso LHC, Sechi A. Highly fluorescent hybrid nanofibers as potential nanofibrous scaffolds for studying cell-fiber interactions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 319:124535. [PMID: 38830327 DOI: 10.1016/j.saa.2024.124535] [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: 01/23/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
In this study, we report on the fabrication of hybrid nanofibers for labeling and bioimaging applications. Our approach is involved for developing highly fluorescent nanofibers using a blend of polylactic acid, polyethyleneglycol, and perylenediimide dyes, through the solution blow spinning technique. The nanofibers are exhibited diameters ranging from 330 nm to 420 nm. Nanofibers showed excellent red and near-infrared fluorescence emissive properties in fluorescent spectroscopy. Moreover, the strong two-photon absorption phenomenon was observed for nanofibers under confocal microscopy. To assess the applicability of these fluorescent nanofibers in bioimaging settings, we employ two types of mammalian cells B16F1 melanoma cells and J774.A1 macrophages. Both cell types exhibit negligible cytotoxicity after 24 h incubation with the nanofibers, indicating the suitability of nanofibers for cell-based experiments. We also observe strong interactions between the nanofibers and cells, as evidenced by two major events: a) the acquisition of an elongated cellular morphology with the major cellular axis parallel to the nanofibers and b) the accumulation of actin filaments along the points of contact of the cells with the fibers. Our findings demonstrate the suitability of these newly developed fluorescent nanofibers in cell-based applications for guiding cellular behavior. We expect that these fluorescent nanofibers have the potential to serve as scaffold materials for long-time tracking of cell-fiber interactions in fluorescence microscopy.
Collapse
Affiliation(s)
- Sebastian Raja
- National Nanotechnology Laboratory for Agribusiness (LNNA), Embrapa Instrumentation, 13560-970 São Carlos, SP, Brazil; Dept. of Cell and Tumor Biology, Medical Faculty, RWTH Aachen University, Pauwelsstrasse, 30, D-52074 Aachen, Germany; Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Marcina Strzody Street 9, Gliwice 44-100, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego Street 22b, Gliwice 44-100, Poland.
| | - Rafaella T Paschoalin
- National Nanotechnology Laboratory for Agribusiness (LNNA), Embrapa Instrumentation, 13560-970 São Carlos, SP, Brazil
| | - Idelma A A Terra
- National Nanotechnology Laboratory for Agribusiness (LNNA), Embrapa Instrumentation, 13560-970 São Carlos, SP, Brazil
| | - Carmen Schalla
- Dept. of Cell and Tumor Biology, Medical Faculty, RWTH Aachen University, Pauwelsstrasse, 30, D-52074 Aachen, Germany
| | - Francisco Guimarães
- São Carlos Institute of Physics, University of São Paulo, Av. Trabalhador São-Carlense, 400, 13566-590 São Carlos, São Paulo, Brazil
| | - Govindasami Periyasami
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Luiz H C Mattoso
- National Nanotechnology Laboratory for Agribusiness (LNNA), Embrapa Instrumentation, 13560-970 São Carlos, SP, Brazil
| | - Antonio Sechi
- Dept. of Cell and Tumor Biology, Medical Faculty, RWTH Aachen University, Pauwelsstrasse, 30, D-52074 Aachen, Germany
| |
Collapse
|
22
|
Maduka CV, Makela AV, Tundo A, Ural E, Stivers KB, Kuhnert MM, Alhaj M, Hoque Apu E, Ashammakhi N, Hankenson KD, Narayan R, Elisseeff JH, Contag CH. Regulating the proinflammatory response to composite biomaterials by targeting immunometabolism. Bioact Mater 2024; 40:64-73. [PMID: 38948254 PMCID: PMC11214186 DOI: 10.1016/j.bioactmat.2024.05.046] [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: 03/11/2024] [Revised: 05/03/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024] Open
Abstract
Composite biomaterials comprising polylactide (PLA) and hydroxyapatite (HA) are applied in bone, cartilage and dental regenerative medicine, where HA confers osteoconductive properties. However, after surgical implantation, adverse immune responses to these composites can occur, which have been attributed to size and morphology of HA particles. Approaches to effectively modulate these adverse immune responses have not been described. PLA degradation products have been shown to alter immune cell metabolism (immunometabolism), which drives the inflammatory response. Accordingly, to modulate the inflammatory response to composite biomaterials, inhibitors were incorporated into composites comprised of amorphous PLA (aPLA) and HA (aPLA + HA) to regulate glycolytic flux. Inhibition at specific steps in glycolysis reduced proinflammatory (CD86+CD206-) and increased pro-regenerative (CD206+) immune cell populations around implanted aPLA + HA. Notably, neutrophil and dendritic cell (DC) numbers along with proinflammatory monocyte and macrophage populations were decreased, and Arginase 1 expression among DCs was increased. Targeting immunometabolism to control the proinflammatory response to biomaterial composites, thereby creating a pro-regenerative microenvironment, is a significant advance in tissue engineering where immunomodulation enhances osseointegration and angiogenesis, which could lead to improved bone regeneration.
Collapse
Affiliation(s)
- Chima V. Maduka
- Comparative Medicine & Integrative Biology, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Ashley V. Makela
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Anthony Tundo
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Evran Ural
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Katlin B. Stivers
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Maxwell M. Kuhnert
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Mohammed Alhaj
- Department of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Ehsanul Hoque Apu
- Department of Biomedical Sciences, College of Dental Medicine, Lincoln Memorial University, Knoxville, TN, 37917, USA
| | - Nureddin Ashammakhi
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Ramani Narayan
- Department of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Christopher H. Contag
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, 48864, USA
| |
Collapse
|
23
|
Gong T, Lu M, Sheng H, Li Z, Zhou Y, Luo Y, Min L, Tu C. Survivorship, complications, and functional outcomes of uncemented distal femoral endoprosthesis with short, curved stem for patients with bone tumours. Bone Joint J 2024; 106-B:1000-1007. [PMID: 39216865 DOI: 10.1302/0301-620x.106b9.bjj-2024-0109.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Aims Endoprosthetic reconstruction following distal femur tumour resection has been widely advocated. In this paper, we present the design of an uncemented endoprosthesis system featuring a short, curved stem, with the goal of enhancing long-term survivorship and functional outcomes. Methods This study involved patients who underwent implantation of an uncemented distal femoral endoprosthesis with a short and curved stem between 2014 and 2019. Functional outcomes were assessed using the 1993 version of the Musculoskeletal Tumour Society (MSTS-93) score. Additionally, we quantified five types of complications and assessed osseointegration radiologically. The survivorship of the endoprosthesis was evaluated according to two endpoints. A total of 134 patients with a median age of 26 years (IQR 16 to 41) were included in our study. The median follow-up time was 61 months (IQR 56 to 76), and the median functional MSTS-93 was 83% (IQR 73 to 91) postoperatively. Results Overall, 21 patients (16%) encountered complications, and the rate of aseptic loosening was 7% (9/134). The survival rate up to 8.5 years was 93% for aseptic loosening as the endpoint, and 88% for any reason as the endpoint, retrospectively. Conclusion The use of an uncemented distal femoral endoprosthesis with a short, curved stem demonstrated a low incidence of aseptic loosening and achieved long-term survivorship of up to nine years. Meanwhile, aseptic loosening typically occurs in the early stage postoperatively.
Collapse
Affiliation(s)
- Taojun Gong
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Minxun Lu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Hongtao Sheng
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Zhuangzhuang Li
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Yong Zhou
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Yi Luo
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Li Min
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| | - Chongqi Tu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Model Worker and Craftsman Talent Innovation Workshop of Sichuan province, Chengdu, China
| |
Collapse
|
24
|
Aslanbay Guler B, Morçimen ZG, Taşdemir Ş, Demirel Z, Turunç E, Şendemir A, Imamoglu E. Design of chemobrionic and biochemobrionic scaffolds for bone tissue engineering. Sci Rep 2024; 14:13764. [PMID: 38877025 PMCID: PMC11178857 DOI: 10.1038/s41598-024-63171-z] [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: 02/02/2024] [Accepted: 05/27/2024] [Indexed: 06/16/2024] Open
Abstract
Chemobrionic systems have attracted great attention in material science for development of novel biomimetic materials. This study aims to design a new bioactive material by integrating biosilica into chemobrionic structure, which will be called biochemobrionic, and to comparatively investigate the use of both chemobrionic and biochemobrionic materials as bone scaffolds. Biosilica, isolated from Amphora sp. diatom, was integrated into chemobrionic structure, and a comprehensive set of analysis was conducted to evaluate their morphological, chemical, mechanical, thermal, and biodegradation properties. Then, the effects of both scaffolds on cell biocompatibility and osteogenic differentiation capacity were assessed. Cells attached to the scaffolds, spread out, and covered the entire surface, indicating the absence of cytotoxicity. Biochemobrionic scaffold exhibited a higher level of mineralization and bone formation than the chemobrionic structure due to the osteogenic activity of biosilica. These results present a comprehensive and pioneering understanding of the potential of (bio)chemobrionics for bone regeneration.
Collapse
Affiliation(s)
- Bahar Aslanbay Guler
- Bioengineering Department, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Zehra Gül Morçimen
- Bioengineering Department, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Şeyma Taşdemir
- Ioengineering Department, Faculty of Engineering, Manisa Celal Bayar University, Manisa, Turkey
| | - Zeliha Demirel
- Bioengineering Department, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Ezgi Turunç
- Department of Biochemistry, Faculty of Pharmacy, İzmir Katip Çelebi University, İzmir, Turkey
| | - Aylin Şendemir
- Bioengineering Department, Faculty of Engineering, Ege University, Izmir, Turkey
| | - Esra Imamoglu
- Bioengineering Department, Faculty of Engineering, Ege University, Izmir, Turkey.
| |
Collapse
|
25
|
Dacrory S. Anti-proliferative, antimicrobial, DFT calculations, and molecular docking 3D scaffold based on sodium alginate, chitosan, neomycin sulfate and hydroxyapatite. Int J Biol Macromol 2024; 270:132297. [PMID: 38744365 DOI: 10.1016/j.ijbiomac.2024.132297] [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: 10/02/2023] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
3D multifunctional scaffold has been designed based on Cs/SA/NS/NPHA. Nanoparticles hydroxyapatite (NPHA) was prepared via precipitation method of sodium dihydrogen phosphate in presence calcium chloride. Different ratios of Chitosan (CS)/Sodium Alginate (SA) were used to prepare Cs/SA scaffolds in presence of CaCl2 as a cross linker. NPHA was incorporated in CS/SA scaffold and neomycin sulfate (NS) was added as an antimicrobial agent. The structure and surface morphology of the scaffolds were investigated via infrared spectroscopy (FTIR), X-ray diffraction (XRD), scanning electron microscope (SEM) and thermal gravimetric analysis (TGA) techniques. Additionally, Antimicrobial activity of the scaffold has evaluated against Gram- negative and Gram- positive bacteria. The result showed promising antimicrobial activity against Bacillus subtilis, Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa and Candida albicans. Furthermore, cytotoxicity against MG63 osteosarcoma cell and fibroblast normal cell line has investigated. The result showed anti-proliferative against MG63. DFT calculations and molecular docking were used to study the reactivity of the compounds. The results exhibited that Cs/SA/NS/NPHA is potent expected to be used in bone tissue regeneration.
Collapse
Affiliation(s)
- Sawsan Dacrory
- Cellulose & Paper Department, National Research Centre, Dokki, Giza, Egypt.
| |
Collapse
|
26
|
Zhou Z, Feng W, Moghadas BK, Baneshi N, Noshadi B, Baghaei S, Dehkordi DA. Review of recent advances in bone scaffold fabrication methods for tissue engineering for treating bone diseases and sport injuries. Tissue Cell 2024; 88:102390. [PMID: 38663113 DOI: 10.1016/j.tice.2024.102390] [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: 03/04/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 06/17/2024]
Abstract
Despite advancements in medical care, the management of bone injuries remains one of the most significant challenges in the fields of medicine and sports medicine globally. Bone tissue damage is often associated with aging, reduced quality of life, and various conditions such as trauma, cancer, and infection. While bone tissue possesses the natural capacity for self-repair and regeneration, severe damage may render conventional treatments ineffective, and bone grafting may be limited due to secondary surgical procedures and potential disease transmission. In such cases, bone tissue engineering has emerged as a viable approach, utilizing cells, scaffolds, and growth factors to repair damaged bone tissue. This research shows a comprehensive review of the current literature on the most important and effective methods and materials for improving the treatment of these injuries. Commonly employed cell types include osteogenic cells, embryonic stem cells, and mesenchymal cells, while scaffolds play a crucial role in bone tissue regeneration. To create an effective bone scaffold, a thorough understanding of bone structure, material selection, and examination of scaffold fabrication techniques from inception to the present day is necessary. By gaining insights into these three key components, the ability to design and construct appropriate bone scaffolds can be achieved. Bone tissue engineering scaffolds are evaluated based on factors such as strength, porosity, cell adhesion, biocompatibility, and biodegradability. This article examines the diverse categories of bone scaffolds, the materials and techniques used in their fabrication, as well as the associated merits and drawbacks of these approaches. Furthermore, the review explores the utilization of various scaffold types in bone tissue engineering applications.
Collapse
Affiliation(s)
- Zeng Zhou
- Department of Physical Education, Central South University, Changsha, Hunan 4100083, China
| | - Wei Feng
- Department of Physical Education, Central South University, Changsha, Hunan 4100083, China.
| | - B Kamyab Moghadas
- Department of Chemical Engineering, Shiraz Branch, Islamic Azad University, Shiraz, Iran; Department of Applied Researches, Chemical, Petroleum & Polymer Engineering Research Center, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - N Baneshi
- Department of Chemical Engineering, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - B Noshadi
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Eastern Mediterranean University, via Mersin 10, TR-99628 Famagusta, North Cyprus, Turkey
| | - Sh Baghaei
- Medical Doctor, Isfahan University of Medical Science, Isfahan, Iran
| | - D Abasi Dehkordi
- Medical Doctor, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
27
|
Kosinski J, Sechi A, Hain J, Villwock S, Ha SA, Hauschulz M, Rose M, Steib F, Ortiz‐Brüchle N, Heij L, Maas SL, van der Vorst EPC, Knoesel T, Altendorf‐Hofmann A, Simon R, Sauter G, Bednarsch J, Jonigk D, Dahl E. ITIH5 as a multifaceted player in pancreatic cancer suppression, impairing tyrosine kinase signaling, cell adhesion and migration. Mol Oncol 2024; 18:1486-1509. [PMID: 38375974 PMCID: PMC11161730 DOI: 10.1002/1878-0261.13609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
Inter-alpha-trypsin inhibitor heavy chain 5 (ITIH5) has been identified as a metastasis suppressor gene in pancreatic cancer. Here, we analyzed ITIH5 promoter methylation and protein expression in The Cancer Genome Atlas (TCGA) dataset and three tissue microarray cohorts (n = 618), respectively. Cellular effects, including cell migration, focal adhesion formation and protein tyrosine kinase activity, induced by forced ITIH5 expression in pancreatic cancer cell lines were studied in stable transfectants. ITIH5 promoter hypermethylation was associated with unfavorable prognosis, while immunohistochemistry demonstrated loss of ITIH5 in the metastatic setting and worsened overall survival. Gain-of-function models showed a significant reduction in migration capacity, but no alteration in proliferation. Focal adhesions in cells re-expressing ITIH5 exhibited a smaller and more rounded phenotype, typical for slow-moving cells. An impressive increase of acetylated alpha-tubulin was observed in ITIH5-positive cells, indicating more stable microtubules. In addition, we found significantly decreased activities of kinases related to focal adhesion. Our results indicate that loss of ITIH5 in pancreatic cancer profoundly affects its molecular profile: ITIH5 potentially interferes with a variety of oncogenic signaling pathways, including the PI3K/AKT pathway. This may lead to altered cell migration and focal adhesion formation. These cellular alterations may contribute to the metastasis-inhibiting properties of ITIH5 in pancreatic cancer.
Collapse
Affiliation(s)
- Jennifer Kosinski
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Antonio Sechi
- Department of Cell and Tumor BiologyRWTH Aachen UniversityGermany
| | - Johanna Hain
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Sophia Villwock
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Stefanie Anh Ha
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Maximilian Hauschulz
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Michael Rose
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Florian Steib
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Nadina Ortiz‐Brüchle
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Lara Heij
- Institute of PathologyUniversity Hospital EssenGermany
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
- Department of PathologyErasmus Medical Center RotterdamThe Netherlands
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityThe Netherlands
| | - Sanne L. Maas
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐University MunichGermany
| | - Thomas Knoesel
- Institute of PathologyLudwig‐Maximilians‐University MunichGermany
| | | | - Ronald Simon
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Guido Sauter
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Jan Bednarsch
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
| | - Danny Jonigk
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
- German Center for Lung Research (DZL), BREATHHanoverGermany
| | - Edgar Dahl
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
| |
Collapse
|
28
|
Avinashi SK, Shweta, Bohra B, Mishra RK, Kumari S, Fatima Z, Hussain A, Saxena B, Kumar S, Banerjee M, Gautam CR. Fabrication of Novel 3-D Nanocomposites of HAp-TiC-h-BN-ZrO 2: Enhanced Mechanical Performances and In Vivo Toxicity Study for Biomedical Applications. ACS Biomater Sci Eng 2024; 10:2116-2132. [PMID: 38498674 DOI: 10.1021/acsbiomaterials.3c01478] [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: 03/20/2024]
Abstract
Due to excellent biocompatibility, bioactivities, and osteoconductivity, hydroxyapatite (HAp) is considered as one of the most suitable biomaterials for numerous biomedical applications. Herein, HAp was fabricated using a bottom-up approach, i.e., a wet chemical method, and its composites with TiC, h-BN, and ZrO2 were fabricated by a solid-state reaction method with enhanced mechanical and biological performances. Structural, surface morphology, and mechanical behavior of the fabricated composites were characterized using various characterization techniques. Furthermore, transmission electron microscopy study revealed a randomly oriented rod-like morphology, with the length and width of these nanorods ranging from 78 to 122 and from 9 to 13 nm. Moreover, the mechanical characterizations of the composite HZBT4 (80HAp-10TiC-5h-BN-5ZrO2) reveal a very high compressive strength (246 MPa), which is comparable to that of the steel (250 MPa), fracture toughness (14.78 MPa m1/2), and Young's modulus (1.02 GPa). In order to check the biocompatibility of the composites, numerous biological tests were also performed on different body organs of healthy adult Sprague-Dawley rats. This study suggests that the composite HZBT4 could not reveal any significant influence on the hematological, serum biochemical, and histopathological parameters. Hence, the fabricated composite can be used for several biological applications, such as bone implants, bone grafting, and bone regeneration.
Collapse
Affiliation(s)
- Sarvesh Kumar Avinashi
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
| | - Shweta
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
| | - Bhavna Bohra
- Department of Pharmacology, Institute of Pharmacy, Nirma University, S.G. Highway, Ahmedabad 382481, India
| | - Rajat Kumar Mishra
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
| | - Savita Kumari
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
| | - Zaireen Fatima
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
- Department of Physics, Integral University, Lucknow 226026, India
| | - Ajaz Hussain
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
| | - Bhagawati Saxena
- Department of Pharmacology, Institute of Pharmacy, Nirma University, S.G. Highway, Ahmedabad 382481, India
| | - Saurabh Kumar
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Monisha Banerjee
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Chandki Ram Gautam
- Advanced Glass and Glass Ceramics Research Laboratory, Department of Physics, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
29
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
30
|
Štěpánková K, Ozaltin K, Sáha P, Vargun E, Domincová-Bergerová E, Vesel A, Mozetič M, Lehocký M. Carboxymethylated and Sulfated Furcellaran from Furcellaria lumbricalis and Its Immobilization on PLA Scaffolds. Polymers (Basel) 2024; 16:720. [PMID: 38475404 PMCID: PMC10934783 DOI: 10.3390/polym16050720] [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: 01/03/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
This study involved the creation of highly porous PLA scaffolds through the porogen/leaching method, utilizing polyethylene glycol as a porogen with a 75% mass ratio. The outcome achieved a highly interconnected porous structure with a thickness of 25 μm. To activate the scaffold's surface and improve its hydrophilicity, radiofrequency (RF) air plasma treatment was employed. Subsequently, furcellaran subjected to sulfation or carboxymethylation was deposited onto the RF plasma treated surfaces with the intention of improving bioactivity. Surface roughness and water wettability experienced enhancement following the surface modification. The incorporation of sulfate/carboxymethyl group (DS = 0.8; 0.3, respectively) is confirmed by elemental analysis and FT-IR. Successful functionalization of PLA scaffolds was validated by SEM and XPS analysis, showing changes in topography and increases in characteristic elements (N, S, Na) for sulfated (SF) and carboxymethylated (CMF). Cytocompatibility was evaluated by using mouse embryonic fibroblast cells (NIH/3T3).
Collapse
Affiliation(s)
- Kateřina Štěpánková
- Centre of Polymer Systems, University Institute, Tomas Bata University in Zlín, Trida Tomase Bati 5678, 760 01 Zlin, Czech Republic; (K.O.); (P.S.); (E.D.-B.)
| | - Kadir Ozaltin
- Centre of Polymer Systems, University Institute, Tomas Bata University in Zlín, Trida Tomase Bati 5678, 760 01 Zlin, Czech Republic; (K.O.); (P.S.); (E.D.-B.)
| | - Petr Sáha
- Centre of Polymer Systems, University Institute, Tomas Bata University in Zlín, Trida Tomase Bati 5678, 760 01 Zlin, Czech Republic; (K.O.); (P.S.); (E.D.-B.)
| | - Elif Vargun
- Department of Chemistry, Mugla Sitki Kocman University, Kotekli, 48000 Mugla, Turkey;
| | - Eva Domincová-Bergerová
- Centre of Polymer Systems, University Institute, Tomas Bata University in Zlín, Trida Tomase Bati 5678, 760 01 Zlin, Czech Republic; (K.O.); (P.S.); (E.D.-B.)
| | - Alenka Vesel
- Department of Surface Engineering, Jozef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia; (A.V.); (M.M.)
| | - Miran Mozetič
- Department of Surface Engineering, Jozef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia; (A.V.); (M.M.)
| | - Marian Lehocký
- Centre of Polymer Systems, University Institute, Tomas Bata University in Zlín, Trida Tomase Bati 5678, 760 01 Zlin, Czech Republic; (K.O.); (P.S.); (E.D.-B.)
| |
Collapse
|
31
|
Harb SV, Kolanthai E, Pugazhendhi AS, Beatrice CA, Pinto LA, Neal CJ, Backes EH, Nunes AC, Selistre-de-Araújo HS, Costa LC, Coathup MJ, Seal S, Pessan LA. 3D printed bioabsorbable composite scaffolds of poly (lactic acid)-tricalcium phosphate-ceria with osteogenic property for bone regeneration. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100086. [PMID: 38213985 PMCID: PMC10776431 DOI: 10.1016/j.bbiosy.2023.100086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/28/2023] [Accepted: 12/17/2023] [Indexed: 01/13/2024] Open
Abstract
The fabrication of customized implants by additive manufacturing has allowed continued development of the personalized medicine field. Herein, a 3D-printed bioabsorbable poly (lactic acid) (PLA)- β-tricalcium phosphate (TCP) (10 wt %) composite has been modified with CeO2 nanoparticles (CeNPs) (1, 5 and 10 wt %) for bone repair. The filaments were prepared by melt extrusion and used to print porous scaffolds. The nanocomposite scaffolds possessed precise structure with fine print resolution, a homogenous distribution of TCP and CeNP components, and mechanical properties appropriate for bone tissue engineering applications. Cell proliferation assays using osteoblast cultures confirmed the cytocompatibility of the composites. In addition, the presence of CeNPs enhanced the proliferation and differentiation of mesenchymal stem cells; thereby, increasing alkaline phosphatase (ALP) activity, calcium deposition and bone-related gene expression. Results from this study have shown that the 3D printed PLA-TCP-10%CeO2 composite scaffold could be used as an alternative polymeric implant for bone tissue engineering applications: avoiding additional/revision surgeries and accelerating the regenerative process.
Collapse
Affiliation(s)
- Samarah V. Harb
- Department of Materials Engineering (DEMa), Federal University of Sao Carlos (UFSCar), São Carlos, SP, Brazil
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | | | - Cesar A.G. Beatrice
- Department of Materials Engineering (DEMa), Federal University of Sao Carlos (UFSCar), São Carlos, SP, Brazil
| | - Leonardo A. Pinto
- Graduate Program in Materials Science and Engineering, Department of Materials Engineering (DEMa), Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil
| | - Craig J. Neal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Eduardo H. Backes
- Department of Materials Engineering (DEMa), Federal University of Sao Carlos (UFSCar), São Carlos, SP, Brazil
| | - Ana C.C. Nunes
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil
| | | | - Lidiane C. Costa
- Department of Materials Engineering (DEMa), Federal University of Sao Carlos (UFSCar), São Carlos, SP, Brazil
| | - Melanie J. Coathup
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Luiz A. Pessan
- Department of Materials Engineering (DEMa), Federal University of Sao Carlos (UFSCar), São Carlos, SP, Brazil
| |
Collapse
|
32
|
Pudełko-Prażuch I, Balasubramanian M, Ganesan SM, Marecik S, Walczak K, Pielichowska K, Chatterjee S, Kandaswamy R, Pamuła E. Characterization and In Vitro Evaluation of Porous Polymer-Blended Scaffolds Functionalized with Tricalcium Phosphate. J Funct Biomater 2024; 15:57. [PMID: 38535250 PMCID: PMC10970789 DOI: 10.3390/jfb15030057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 11/11/2024] Open
Abstract
Bone tissue is one of the most transplanted tissues. The ageing population and bone diseases are the main causes of the growing need for novel treatments offered by bone tissue engineering. Three-dimensional (3D) scaffolds, as artificial structures that fulfil certain characteristics, can be used as a temporary matrix for bone regeneration. In this study, we aimed to fabricate 3D porous polymer scaffolds functionalized with tricalcium phosphate (TCP) particles for applications in bone tissue regeneration. Different combinations of poly(lactic acid) (PLA), poly(ethylene glycol) (PEG with molecular weight of 600 or 2000 Da) and poly(ε-caprolactone) (PCL) with TCP were blended by a gel-casting method combined with rapid heating. Porous composite scaffolds with pore sizes from 100 to 1500 µm were obtained. ATR-FTIR, DSC, and wettability tests were performed to study scaffold composition, thermal properties, and hydrophilicity, respectively. The samples were observed with the use of optical and scanning electron microscopes. The addition of PCL to PLA increased the hydrophobicity of the composite scaffolds and reduced their susceptibility to degradation, whereas the addition of PEG increased the hydrophilicity and degradation rates but concomitantly resulted in enhanced creation of rounded mineral deposits. The scaffolds were not cytotoxic according to an indirect test in L929 fibroblasts, and they supported adhesion and growth of MG-63 cells when cultured in direct contact.
Collapse
Affiliation(s)
- Iwona Pudełko-Prażuch
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, Al. Mickiewicza 30, 30-059 Krakow, Poland; (I.P.-P.); (S.M.); (K.W.); (K.P.)
| | - Mareeswari Balasubramanian
- Department of Rubber and Plastics Technology, Madras Institute of Technology Campus, Anna University, Chromepet, Chennai 600 044, Tamil Nadu, India; (M.B.); (S.M.G.)
| | - Sundara Moorthi Ganesan
- Department of Rubber and Plastics Technology, Madras Institute of Technology Campus, Anna University, Chromepet, Chennai 600 044, Tamil Nadu, India; (M.B.); (S.M.G.)
| | - Stanisław Marecik
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, Al. Mickiewicza 30, 30-059 Krakow, Poland; (I.P.-P.); (S.M.); (K.W.); (K.P.)
| | - Kamila Walczak
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, Al. Mickiewicza 30, 30-059 Krakow, Poland; (I.P.-P.); (S.M.); (K.W.); (K.P.)
| | - Kinga Pielichowska
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, Al. Mickiewicza 30, 30-059 Krakow, Poland; (I.P.-P.); (S.M.); (K.W.); (K.P.)
| | - Suvro Chatterjee
- Department of Biotechnology, Golapbag Campus, University of Burdwan, Burdwan 713 104, West Bengal, India;
| | - Ravichandran Kandaswamy
- Department of Rubber and Plastics Technology, Madras Institute of Technology Campus, Anna University, Chromepet, Chennai 600 044, Tamil Nadu, India; (M.B.); (S.M.G.)
| | - Elżbieta Pamuła
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, Al. Mickiewicza 30, 30-059 Krakow, Poland; (I.P.-P.); (S.M.); (K.W.); (K.P.)
| |
Collapse
|
33
|
Klimczuk B, Rudnicka A, Owczarek O, Puszkarz AK, Szparaga G, Puchalski M. Investigation of the Hydrolytic Degradation Kinetics of 3D-Printed PLA Structures under a Thermally Accelerated Regime. MATERIALS (BASEL, SWITZERLAND) 2024; 17:1043. [PMID: 38473515 DOI: 10.3390/ma17051043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
The application of biobased and biodegradable polymers, such as polylactide (PLA), in fused deposition modeling (FDM) 3D-printing technology creates a new prospect for rapid prototyping and other applications in the context of ecology. The popularity of the FDM method and its significance in material engineering not only creates new prospects for the development of technical sciences on an industrial scale, but also introduces new technologies into households. In this study, the kinetics of the hydrolytic degradation of samples obtained by the FDM method from commercially available PLA filaments under a thermally accelerated regime were analyzed. The investigation was conducted at the microstructural, supramolecular, and molecular levels by using methods such as micro-computed tomography (micro-CT), wide-angle X-ray diffraction (WAXD), viscosimetry, and mass erosion measurements. The obtained results clearly present the rapid structural changes in 3D-printed materials during degradation due to their amorphous initial structure. The complementary studies carried out at different scale levels allowed us to demonstrate the relationship between the observed structural changes in the samples and the hydrolytic decomposition of the polymer chains, which made it possible to scientifically understand the process and expand the knowledge on biodegradation.
Collapse
Affiliation(s)
- Bartłomiej Klimczuk
- ECOResearch Student Research Group, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
| | - Aleksandra Rudnicka
- ECOResearch Student Research Group, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
| | - Oliwia Owczarek
- ECOResearch Student Research Group, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
| | - Adam K Puszkarz
- Division of Materials Science, Commodity Science and Textile Metrology, Textile Institute, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
| | - Grzegorz Szparaga
- Division of Technology of Yarns, Commodity Science and Textile Metrology, Textile Institute, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
| | - Michał Puchalski
- ECOResearch Student Research Group, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
- Division of Materials Science, Commodity Science and Textile Metrology, Textile Institute, Faculty of Material Technologies and Textile Design, Lodz University of Technology, 116 Zeromskiego Str., 90-924 Lodz, Poland
| |
Collapse
|
34
|
Liang W, Zhou C, Bai J, Zhang H, Long H, Jiang B, Liu L, Xia L, Jiang C, Zhang H, Zhao J. Nanotechnology-based bone regeneration in orthopedics: a review of recent trends. Nanomedicine (Lond) 2024; 19:255-275. [PMID: 38275154 DOI: 10.2217/nnm-2023-0187] [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: 01/27/2024] Open
Abstract
Nanotechnology has revolutionized the field of bone regeneration, offering innovative solutions to address the challenges associated with conventional therapies. This comprehensive review explores the diverse landscape of nanomaterials - including nanoparticles, nanocomposites and nanofibers - tailored for bone tissue engineering. We delve into the intricate design principles, structural mimicry of native bone and the crucial role of biomaterial selection, encompassing bioceramics, polymers, metals and their hybrids. Furthermore, we analyze the interface between cells and nanostructured materials and their pivotal role in engineering and regenerating bone tissue. In the concluding outlook, we highlight emerging frontiers and potential research directions in harnessing nanomaterials for bone regeneration.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua hospital, Zhoushan, 316000, China
| | - Juqin Bai
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hongwei Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hengguo Long
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Bo Jiang
- Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Chanyi Jiang
- Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hengjian Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| |
Collapse
|
35
|
Kauke-Navarro M, Knoedler L, Knoedler S, Deniz C, Stucki L, Safi AF. Balancing beauty and science: a review of facial implant materials in craniofacial surgery. Front Surg 2024; 11:1348140. [PMID: 38327548 PMCID: PMC10847330 DOI: 10.3389/fsurg.2024.1348140] [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: 12/01/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024] Open
Abstract
Facial reconstruction and augmentation, integral in facial plastic surgery, address defects related to trauma, tumors infections, and congenital skeletal deficiencies. Aesthetic considerations, including age-related facial changes, involve volume loss and diminished projection, often associated with predictable changes in the facial skeleton. Autologous, allogeneic, and alloplastic implants are used to address these concerns. Autologous materials such as bone, cartilage, and fat, while longstanding options, have limitations, including unpredictability and resorption rates. Alloplastic materials, including metals, polymers, and ceramics, offer alternatives. Metals like titanium are biocompatible and used primarily in fracture fixation. Polymers, such as silicone and polyethylene, are widely used, with silicone presenting migration, bony resorption, and visibility issues. Polyethylene, particularly porous polyethylene (MedPor), was reported to have one of the lowest infection rates while it becomes incorporated into the host. Polyether-ether-ketone (PEEK) exhibits mechanical strength and compatibility with imaging modalities, with custom PEEK implants providing stable results. Acrylic materials, like poly-methylmethacrylate (PMMA), offer strength and is thus mostly used in the case of cranioplasty. Bioceramics, notably hydroxyapatite (HaP), offer osteoconductive and inductive properties, and HaP granules demonstrate stable volume retention in facial aesthetic augmentation. Combining HaP with other materials, such as PLA, may enhance mechanical stability. 3D bioprinting with HaP-based bioinks presents a promising avenue for customizable and biocompatible implants. In conclusion, various materials have been used for craniofacial augmentation, but none have definitively demonstrated superiority. Larger randomized controlled trials are essential to evaluate short- and long-term complications comprehensively, potentially revolutionizing facial balancing surgery.
Collapse
Affiliation(s)
- Martin Kauke-Navarro
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
- Craniologicum, Center for Craniomaxillofacial Surgery, Bern, Switzerland
| | - Leonard Knoedler
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Samuel Knoedler
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Can Deniz
- Craniologicum, Center for Craniomaxillofacial Surgery, Bern, Switzerland
| | - Lars Stucki
- Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| | - Ali-Farid Safi
- Craniologicum, Center for Craniomaxillofacial Surgery, Bern, Switzerland
- Medical Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
36
|
Stogov MV, Kireeva EA, Dubinenko GE, Tverdokhlebov SI. The influence of polylactide/hydroxyapatite composite material crystallinity on the polymer structure degradation rate. GENIJ ORTOPEDII 2023; 29:591-595. [DOI: 10.18019/1028-4427-2023-29-6-591-595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Introduction Assessment of biological characteristics of polylactide/hydroxyapatite (PLLA/HA) biodegradable materials is requiered to specify indications for the use of PLLA/HA composite implants in clinical practice.The present study was aimed to measure the kinetics of calcium and phosphate release from PLLA and its dependence on polymer structure crystallinity. Material and methods Four types of biodegradable materials were studied in vitro. Samples of type 1 and type 3 made of crystalline PLLA after annealing contained 25 % and 50 % of HA mass fraction, respectively. Samples of type 2 and type 4 made of amorphous PLLA (without annealing) contained 25 % and 50 % of HA mass fraction, respectively. In every group, 6 samples were tested. The samples were incubated in an aqueous medium at 37 °C for 52 weeks. The rate of PLLA degradation was assessed by the accumulation of lactate monomer in the hydrolysate. The concentrations of calcium ions and phosphate ions were determined for assessment the HA hydrolysis rate. The degree of crystallinity of the polymer matrix was evaluated by scanning calorimetry.Results The hydrolysis of PLLA and HA in the samples was not simultaneous. The PLLA was hydrolyzed first followed by HA hydrolysis. By the moment of complete hydrolysis of PLLA, there was only 15 % of hydrolyzed HA. The release of calcium ions occurred from the sixth week of incubation for all tested samples, that of phosphate ions from the third week. The total amount of the released calcium ions and phosphate ions decreased in the line: material 3 > material 4 > material 1 > material 2. Calcium ions in the hydrolysates were detected up to 42 weeks of incubation, phosphate ions up to the 52nd week.Conclusion Higher crystallinity of PLLA achieved by annealing results in increased rate of hydrolysis of HA from PLLA matrix. Biological activity of PLLA/HA implants can be determined by degree of polymer crystallinity and saturation with HA.
Collapse
Affiliation(s)
- M. V. Stogov
- Ilizarov National Medical Research Centre for Traumatology and Orthopedics
| | - E. A. Kireeva
- Ilizarov National Medical Research Centre for Traumatology and Orthopedics
| | | | | |
Collapse
|
37
|
Liu YC, Lo GJ, Shyu VBH, Tsai CH, Chen CH, Chen CT. Surface Modification of Polylactic Acid Bioscaffold Fabricated via 3D Printing for Craniofacial Bone Tissue Engineering. Int J Mol Sci 2023; 24:17410. [PMID: 38139240 PMCID: PMC10744214 DOI: 10.3390/ijms242417410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Bone tissue engineering is a promising solution for advanced bone defect reconstruction after severe trauma. In bone tissue engineering, scaffolds in three-dimensional (3D) structures are crucial components for cell growth, migration, and infiltration. The three-dimensional printing technique is well suited to manufacturing scaffolds since it can fabricate scaffolds with highly complex designs under good internal structural control. In the current study, the 3D printing technique was utilized to produce polylactic acid (PLA) scaffolds. BMSCs were seeded onto selected scaffolds, either hydrogel-mixed or not, and cultivated in vitro to investigate the osteogenic potential in each group. After osteogenic incubation in vitro, BMSC-seeded scaffolds were implanted onto rat cranium defects, and bone regeneration was observed after 12 weeks. Our results demonstrated that BMSCs were able to seed onto 3D-printed PLA scaffolds under high-resolution observation. Real-time PCR analysis showed their osteogenic ability, which could be further improved after BMSCs were mixed with hydrogel. The in vivo study showed significantly increased bone regeneration when rats' cranium defects were implanted with a hydrogel-mixed BMSC-seeded scaffold compared to the control and those without cell or hydrogel groups. This study showed that 3D-printed PLA scaffolds are a feasible option for BMSC cultivation and osteogenic differentiation. After mixing with hydrogel, BMSC-seeded 3D-printed scaffolds can facilitate bone regeneration.
Collapse
Affiliation(s)
- Yao-Chang Liu
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (Y.-C.L.); (G.-J.L.); (V.B.-H.S.); (C.-H.T.)
| | - Guan-Jie Lo
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (Y.-C.L.); (G.-J.L.); (V.B.-H.S.); (C.-H.T.)
| | - Victor Bong-Hang Shyu
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (Y.-C.L.); (G.-J.L.); (V.B.-H.S.); (C.-H.T.)
| | - Chia-Hsuan Tsai
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (Y.-C.L.); (G.-J.L.); (V.B.-H.S.); (C.-H.T.)
| | - Chih-Hao Chen
- Department of Plastic and Reconstructive Surgery, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; (Y.-C.L.); (G.-J.L.); (V.B.-H.S.); (C.-H.T.)
| | - Chien-Tzung Chen
- Division of Trauma Plastic Surgery, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Craniofacial Research Center at Taoyuan, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
38
|
García-Sobrino R, Muñoz M, Rodríguez-Jara E, Rams J, Torres B, Cifuentes SC. Bioabsorbable Composites Based on Polymeric Matrix (PLA and PCL) Reinforced with Magnesium (Mg) for Use in Bone Regeneration Therapy: Physicochemical Properties and Biological Evaluation. Polymers (Basel) 2023; 15:4667. [PMID: 38139919 PMCID: PMC10747080 DOI: 10.3390/polym15244667] [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: 11/13/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Improvements in Tissue Engineering and Regenerative Medicine (TERM)-type technologies have allowed the development of specific materials that, together with a better understanding of bone tissue structure, have provided new pathways to obtain biomaterials for bone tissue regeneration. In this manuscript, bioabsorbable materials are presented as emerging materials in tissue engineering therapies related to bone lesions because of their ability to degrade in physiological environments while the regeneration process is completed. This comprehensive review aims to explore the studies, published since its inception (2010s) to the present, on bioabsorbable composite materials based on PLA and PCL polymeric matrix reinforced with Mg, which is also bioabsorbable and has recognized osteoinductive capacity. The research collected in the literature reveals studies based on different manufacturing and dispersion processes of the reinforcement as well as the physicochemical analysis and corresponding biological evaluation to know the osteoinductive capacity of the proposed PLA/Mg and PCL/Mg composites. In short, this review shows the potential of these composite materials and serves as a guide for those interested in bioabsorbable materials applied in bone tissue engineering.
Collapse
Affiliation(s)
- Rubén García-Sobrino
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain; (M.M.); (J.R.); (B.T.)
| | - Marta Muñoz
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain; (M.M.); (J.R.); (B.T.)
| | - Elías Rodríguez-Jara
- Instituto de Cerámica y Vidrio, Consejo Superior de Investigaciones Científicas (CSIC), Campus de Cantoblanco, c/Kelsen 5, 28049 Madrid, Spain;
| | - Joaquín Rams
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain; (M.M.); (J.R.); (B.T.)
| | - Belén Torres
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain; (M.M.); (J.R.); (B.T.)
| | - Sandra C. Cifuentes
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain; (M.M.); (J.R.); (B.T.)
| |
Collapse
|
39
|
Zhang C, Hayashi K, Ishikawa K. Osseointegration enhancement by controlling dispersion state of carbonate apatite in polylactic acid implant. Colloids Surf B Biointerfaces 2023; 232:113588. [PMID: 37844475 DOI: 10.1016/j.colsurfb.2023.113588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Osteoconductive ceramics (OCs) are often used to endow polylactic acid (PLA) with osseointegration ability. Conventionally, OC powder is dispersed in PLA. However, considering cell attachment to the implant, OCs may be more favorable when they exist in the form of aggregations, such as granules, and are larger than the cells rather than being dispersed like a powder. In this study, to clarify the effects of the dispersion state of OCs on the osseointegration ability, carbonate apatite (CAp), a bone mineral analog that is osteoconductive and bioresorbable, powder-PLA (P-PLA), and CAp granule-PLA (G-PLA) composite implants were fabricated via thermal pressing. The powder and granule sizes of CAp were approximately 1 and 300-600 µm, respectively. G-PLA exhibited a higher water wettability and released calcium and phosphate ions faster than P-PLA. When cylindrical G-PLA, P-PLA, and PLA were implanted in rabbit tibial bone defects, G-PLA promoted bone maturation compared to P-PLA and pure PLA. Furthermore, G-PLA bonded directly to the host bone, whereas P-PLA bonded across the osteoid layers. Consequently, the bone-to-implant contact of G-PLA was 1.8- and 5.6-fold higher than those of P-PLA and PLA, respectively. Furthermore, the adhesive shear strength of G-PLA was 1.9- and 3.0-fold higher than those of P-PLA and PLA, respectively. Thus, G-PLA achieved earlier and stronger osseointegration than P-PLA or PLA. The findings of this study highlight the significance of the state of dispersion of OCs in implants as a novel strategy for material development.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biomaterials Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Koichiro Hayashi
- Department of Biomaterials Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Kunio Ishikawa
- Department of Biomaterials Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
40
|
Janmohammadi M, Nourbakhsh MS, Bahraminasab M, Tayebi L. Enhancing bone tissue engineering with 3D-Printed polycaprolactone scaffolds integrated with tragacanth gum/bioactive glass. Mater Today Bio 2023; 23:100872. [PMID: 38075257 PMCID: PMC10709082 DOI: 10.1016/j.mtbio.2023.100872] [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: 06/28/2023] [Revised: 10/26/2023] [Accepted: 11/15/2023] [Indexed: 02/12/2024] Open
Abstract
Tissue-engineered bone substitutes, characterized by favorable physicochemical, mechanical, and biological properties, present a promising alternative for addressing bone defects. In this study, we employed an innovative 3D host-guest scaffold model, where the host component served as a mechanical support, while the guest component facilitated osteogenic effects. More specifically, we fabricated a triangular porous polycaprolactone framework (host) using advanced 3D printing techniques, and subsequently filled the framework's pores with tragacanth gum-45S5 bioactive glass as the guest component. Comprehensive assessments were conducted to evaluate the physical, mechanical, and biological properties of the designed scaffolds. Remarkably, successful integration of the guest component within the framework was achieved, resulting in enhanced bioactivity and increased strength. Our findings demonstrated that the scaffolds exhibited ion release (Si, Ca, and P), surface apatite formation, and biodegradation. Additionally, in vitro cell culture assays revealed that the scaffolds demonstrated significant improvements in cell viability, proliferation, and attachment. Significantly, the multi-compartment scaffolds exhibited remarkable osteogenic properties, indicated by a substantial increase in the expression of osteopontin, osteocalcin, and matrix deposition. Based on our results, the framework provided robust mechanical support during the new bone formation process, while the guest component matrix created a conducive micro-environment for cellular adhesion, osteogenic functionality, and matrix production. These multi-compartment scaffolds hold great potential as a viable alternative to autografts and offer promising clinical applications for bone defect repair in the future.
Collapse
Affiliation(s)
- Mahsa Janmohammadi
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, Semnan University, Semnan, Iran
| | | | - Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, 3513138111, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| |
Collapse
|
41
|
Chen PH, Chen CW, Mao HI, Dai CA, Su CS, Tsai JC, Lin FH. Bio-Based PLA/PBS/PBAT Ternary Blends with Added Nanohydroxyapatite: A Thermal, Physical, and Mechanical Study. Polymers (Basel) 2023; 15:4585. [PMID: 38232012 PMCID: PMC10708084 DOI: 10.3390/polym15234585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024] Open
Abstract
The physical and mechanical properties of novel bio-based polymer blends of polylactic acid (PLA), poly(butylene succinate) (PBS), and poly (butylene adipate-co-terephthalate) (PBAT) with various added amounts of nanohydroxyapatite (nHA) were investigated in this study. The formulations of PLA/PBS/PBAT/nHA blends were divided into two series, A and B, containing 70 or 80 wt% PLA, respectively. Samples of four specimens per series were prepared using a twin-screw extruder, and different amounts of nHA were added to meet the regeneration needs of bone graft materials. FTIR and XRD analyses were employed to identify the presence of each polymer and nHA in the various blends. The crystallization behavior of these blends was examined using DSC. Tensile and impact strength tests were performed on all samples to screen feasible formulations of polymer blends for bone graft material applications. Surface morphology analyses were conducted using SEM, and the dispersion of nHA particles in the blends was further tested using TEM. The added nHA also served as a nucleating agent aimed at improving the crystallinity and mechanical properties of the blends. Through the above analyses, the physical and mechanical properties of the polymer blends are reported and the most promising bone graft material formulations are suggested. All blends were tested for thermal degradation analysis using TGA and thermal stability was confirmed. The water absorption experiments carried out in this study showed that the addition of nHA could improve the hydrophilicity of the blends.
Collapse
Affiliation(s)
- Pei-Hua Chen
- Department of Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan;
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan
| | - Chin-Wen Chen
- Department of Molecular Science and Engineering, Institute of Organic and Polymeric Materials, Research and Development Center of Smart Textile Technology, National Taipei University of Technology, Taipei 106344, Taiwan;
| | - Hsu-I Mao
- Department of Molecular Science and Engineering, Institute of Organic and Polymeric Materials, Research and Development Center of Smart Textile Technology, National Taipei University of Technology, Taipei 106344, Taiwan;
| | - Chi-An Dai
- Department of Chemical Engineering, Institute of Polymer Science and Engineering, National Taiwan University, Taipei 106319, Taiwan;
| | - Chie-Shaan Su
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106344, Taiwan;
| | - Jung-Chin Tsai
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 243303, Taiwan;
| | - Feng-Huei Lin
- Department of Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan;
| |
Collapse
|
42
|
Shaikh S, Baniasadi H, Mehrotra S, Ghosh R, Singh P, Seppälä JV, Kumar A. Strontium-Substituted Nanohydroxyapatite-Incorporated Poly(lactic acid) Composites for Orthopedic Applications: Bioactive, Machinable, and High-Strength Properties. Biomacromolecules 2023; 24:4901-4914. [PMID: 37874127 DOI: 10.1021/acs.biomac.3c00610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Traditional metal-alloy bone fixation devices provide structural support for bone repair but have limitations in actively promoting bone healing and often require additional surgeries for implant removal. In this study, we focused on addressing these challenges by fabricating biodegradable composites using poly(lactic acid) (PLA) and strontium-substituted nanohydroxyapatite (SrHAP) via melt compounding and injection molding. Various percentages of SrHAP (5, 10, 20, and 30% w/w) were incorporated into the PLA matrix. We systematically investigated the structural, morphological, thermal, mechanical, rheological, and dynamic mechanical properties of the prepared composites. Notably, the tensile modulus, a critical parameter for orthopedic implants, significantly improved from 2.77 GPa in pristine PLA to 3.73 GPa in the composite containing 10% w/w SrHAP. The incorporation of SrHAP (10% w/w) into the PLA matrix led to an increased storage modulus, indicating a uniform dispersion of SrHAP within the PLA and good compatibility between the polymer and nanoparticles. Moreover, we successfully fabricated screws using PLA composites with 10% (w/w) SrHAP, demonstrating their formability at room temperature and radiopacity when observed under X-ray microtomography (micro-CT). Furthermore, the water contact angle decreased from 93 ± 2° for pristine PLA to 75 ± 3° for the composite containing SrHAP, indicating better surface wettability. To assess the biological behavior of the composites, we conducted in vitro cell-material tests, which confirmed their osteoconductive and osteoinductive properties. These findings highlight the potential of our developed PLA/SrHAP10 (10% w/w) composites as machinable implant materials for orthopedic applications. In conclusion, our study presents the fabrication and comprehensive characterization of biodegradable composites comprising PLA and strontium-substituted nanohydroxyapatite (SrHAP). These composites exhibit improved mechanical properties, formability, and radiopacity while also demonstrating desirable biological behavior. Our results suggest that these PLA/SrHAP10 composites hold promise as machinable implant materials for orthopedic applications.
Collapse
Affiliation(s)
- Shazia Shaikh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Hossein Baniasadi
- Polymer Technology, School of Chemical Engineering, Aalto University, P.O. Box 16100, Espoo, FI-00076 Aalto, Finland
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Rupita Ghosh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Jukka V Seppälä
- Polymer Technology, School of Chemical Engineering, Aalto University, P.O. Box 16100, Espoo, FI-00076 Aalto, Finland
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center of Excellence for Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| |
Collapse
|
43
|
Maduka CV, Alhaj M, Ural E, Habeeb OM, Kuhnert MM, Smith K, Makela AV, Pope H, Chen S, Hix JM, Mallett CL, Chung S, Hakun M, Tundo A, Zinn KR, Hankenson KD, Goodman SB, Narayan R, Contag CH. Polylactide Degradation Activates Immune Cells by Metabolic Reprogramming. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304632. [PMID: 37737614 PMCID: PMC10625072 DOI: 10.1002/advs.202304632] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/04/2023] [Indexed: 09/23/2023]
Abstract
Polylactide (PLA) is the most widely utilized biopolymer in medicine. However, chronic inflammation and excessive fibrosis resulting from its degradation remain significant obstacles to extended clinical use. Immune cell activation has been correlated to the acidity of breakdown products, yet methods to neutralize the pH have not significantly reduced adverse responses. Using a bioenergetic model, delayed cellular changes were observed that are not apparent in the short-term. Amorphous and semi-crystalline PLA degradation products, including monomeric l-lactic acid, mechanistically remodel metabolism in cells leading to a reactive immune microenvironment characterized by elevated proinflammatory cytokines. Selective inhibition of metabolic reprogramming and altered bioenergetics both reduce these undesirable high cytokine levels and stimulate anti-inflammatory signals. The results present a new biocompatibility paradigm by identifying metabolism as a target for immunomodulation to increase tolerance to biomaterials, ensuring safe clinical application of PLA-based implants for soft- and hard-tissue regeneration, and advancing nanomedicine and drug delivery.
Collapse
Affiliation(s)
- Chima V. Maduka
- Comparative Medicine & Integrative BiologyMichigan State UniversityEast LansingMI48824USA
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Mohammed Alhaj
- Department of Chemical Engineering & Materials ScienceMichigan State UniversityEast LansingMI48824USA
| | - Evran Ural
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Oluwatosin M. Habeeb
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Maxwell M. Kuhnert
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Kylie Smith
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Ashley V. Makela
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Hunter Pope
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Shoue Chen
- School of PackagingMichigan State UniversityEast LansingMI48824USA
| | - Jeremy M. Hix
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Christiane L. Mallett
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Seock‐Jin Chung
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Maxwell Hakun
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Anthony Tundo
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Kurt R. Zinn
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Kurt D. Hankenson
- Department of Orthopedic SurgeryUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Stuart B. Goodman
- Department of Orthopedic SurgeryStanford UniversityStanfordCA94063USA
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Ramani Narayan
- Department of Chemical Engineering & Materials ScienceMichigan State UniversityEast LansingMI48824USA
| | - Christopher H. Contag
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
- Department of Microbiology & Molecular GeneticsMichigan State UniversityEast LansingMI48864USA
| |
Collapse
|
44
|
Ghuloum FI, Stevens LA, Johnson CA, Riobo-Del Galdo NA, Amer MH. Towards modular engineering of cell signalling: Topographically-textured microparticles induce osteogenesis via activation of canonical hedgehog signalling. BIOMATERIALS ADVANCES 2023; 154:213652. [PMID: 37837904 DOI: 10.1016/j.bioadv.2023.213652] [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: 07/19/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Polymer microparticles possess great potential as functional building blocks for advanced bottom-up engineering of complex tissues. Tailoring the three-dimensional architectural features of culture substrates has been shown to induce osteogenesis in mesenchymal stem cells in vitro, but the molecular mechanisms underpinning this remain unclear. This study proposes a mechanism linking the activation of Hedgehog signalling to the osteoinductive effect of surface-engineered, topographically-textured polymeric microparticles. In this study, mesenchymal progenitor C3H10T1/2 cells were cultured on smooth and dimpled poly(D,l-lactide) microparticles to assess differences in viability, cellular morphology, proliferation, and expression of a range of Hedgehog signalling components and osteogenesis-related genes. Dimpled microparticles induced osteogenesis and activated the Hedgehog signalling pathway relative to smooth microparticles and 2D-cultured controls without the addition of exogenous biochemical factors. We observed upregulation of the osteogenesis markers Runt-related transcription factor2 (Runx2) and bone gamma-carboxyglutamate protein 2 (Bglap2), as well as the Hedgehog signalling components, glioma associated oncogene homolog 1 (Gli1), Patched1 (Ptch1), and Smoothened (Smo). Treatment with the Smo antagonist KAAD-cyclopamine confirmed the involvement of Smo in Gli1 target gene activation, with a significant reduction in the expression of Gli1, Runx2 and Bglap2 (p ≤ 0.001) following KAAD-cyclopamine treatment. Overall, our study demonstrates the role of the topographical microenvironment in the modulation of Hedgehog signalling, highlighting the potential for tailoring substrate topographical design to offer cell-instructive 3D microenvironments. Topographically-textured microparticles allow the modulation of Hedgehog signalling in vitro without adding exogenous biochemical agonists, thereby eliminating potential confounding artefacts in high-throughput drug screening applications.
Collapse
Affiliation(s)
- Fatmah I Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom; Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Lee A Stevens
- Low Carbon Energy and Resources Technologies Research Group, Faculty of Engineering, University of Nottingham, UK
| | - Colin A Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom; Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
45
|
Hatt LP, van der Heide D, Armiento AR, Stoddart MJ. β-TCP from 3D-printed composite scaffolds acts as an effective phosphate source during osteogenic differentiation of human mesenchymal stromal cells. Front Cell Dev Biol 2023; 11:1258161. [PMID: 37965582 PMCID: PMC10641282 DOI: 10.3389/fcell.2023.1258161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction: Human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) are often combined with calcium phosphate (CaP)-based 3D-printed scaffolds with the goal of creating a bone substitute that can repair segmental bone defects. In vitro, the induction of osteogenic differentiation traditionally requires, among other supplements, the addition of β-glycerophosphate (BGP), which acts as a phosphate source. The aim of this study is to investigate whether phosphate contained within the 3D-printed scaffolds can effectively be used as a phosphate source during hBM-MSC in vitro osteogenesis. Methods: hBM-MSCs are cultured on 3D-printed discs composed of poly (lactic-co-glycolic acid) (PLGA) and β-tricalcium phosphate (β-TCP) for 28 days under osteogenic conditions, with and without the supplementation of BGP. The effects of BGP removal on various cellular parameters, including cell metabolic activity, alkaline phosphatase (ALP) presence and activity, proliferation, osteogenic gene expression, levels of free phosphate in the media and mineralisation, are assessed. Results: The removal of exogenous BGP increases cell metabolic activity, ALP activity, proliferation, and gene expression of matrix-related (COL1A1, IBSP, SPP1), transcriptional (SP7, RUNX2/SOX9, PPARγ) and phosphate-related (ALPL, ENPP1, ANKH, PHOSPHO1) markers in a donor dependent manner. BGP removal leads to decreased free phosphate concentration in the media and maintained of mineral deposition staining. Discussion: Our findings demonstrate the detrimental impact of exogenous BGP on hBM-MSCs cultured on a phosphate-based material and propose β-TCP embedded within 3D-printed scaffold as a sufficient phosphate source for hBM-MSCs during osteogenesis. The presented study provides novel insights into the interaction of hBM-MSCs with 3D-printed CaP based materials, an essential aspect for the advancement of bone tissue engineering strategies aimed at repairing segmental defects.
Collapse
Affiliation(s)
- Luan P. Hatt
- AO Research Institute Davos, Davos, Switzerland
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Daphne van der Heide
- AO Research Institute Davos, Davos, Switzerland
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | | | | |
Collapse
|
46
|
Cárdenas-Aguazaco W, Camacho B, Gómez-Pachón EY, Lara-Bertrand AL, Silva-Cote I. Electrospun Scaffolds of Polylactic Acid, Collagen, and Amorphous Calcium Phosphate for Bone Repair. Pharmaceutics 2023; 15:2529. [PMID: 38004509 PMCID: PMC10674189 DOI: 10.3390/pharmaceutics15112529] [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: 08/31/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 11/26/2023] Open
Abstract
Most electrospun scaffolds for bone tissue engineering typically use hydroxyapatite (HA) or beta tricalcium phosphate (β-TCP). However, the biological activity of these crystalline compounds can be limited due to their low solubility. Therefore, amorphous calcium phosphate (ACP) may be an alternative in bone repair scaffolds. This study analyzes the morphology, porosity, mechanical strength, and surface chemistry of electrospun scaffolds composed of polylactic acid and collagen integrated with hydroxyapatite (MHAP) or amorphous calcium phosphate (MACP). In addition, the in vitro biocompatibility, osteogenic differentiation, and growth factor production associated with bone repair using human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) are evaluated. The results show that the electrospun MHAP and MACP scaffolds exhibit a fibrous morphology with interconnected pores. Both scaffolds exhibit favorable biocompatibility and stimulate the proliferation and osteogenesis of hWJ-MSCs. However, cell adhesion and osteocalcin production are greater in the MACP scaffold compared to the MHAP scaffold. In addition, the MACP scaffold shows significant production of bone-repair-related growth factors such as transforming growth factor-beta 1 (TGF-β1), providing a solid basis for its use in bone tissue engineering.
Collapse
Affiliation(s)
- William Cárdenas-Aguazaco
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| | - Bernardo Camacho
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| | - Edwin Yesid Gómez-Pachón
- Facultad Duitama, Universidad Pedagógica y Tecnológica de Colombia-UPTC, Duitama 150462, Colombia;
| | - Adriana Lorena Lara-Bertrand
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| | - Ingrid Silva-Cote
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud-IDCBIS, Bogotá 111611, Colombia; (W.C.-A.); (B.C.); (A.L.L.-B.)
| |
Collapse
|
47
|
Moazeni N, Hesaraki S, Behnamghader A, Esmaeilzadeh J, Orive G, Dolatshahi-Pirouz A, Borhan S. Design and Manufacture of Bone Cements Based on Calcium Sulfate Hemihydrate and Mg, Sr-Doped Bioactive Glass. Biomedicines 2023; 11:2833. [PMID: 37893206 PMCID: PMC10604917 DOI: 10.3390/biomedicines11102833] [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: 09/11/2023] [Revised: 10/04/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
In the present study, a novel composite bone cement based on calcium sulfate hemihydrate (CSH) and Mg, Sr-containing bioactive glass (BG) as solid phase, and solution of chitosan as liquid phase were developed. The phase composition, morphology, setting time, injectability, viscosity, and cellular responses of the composites with various contents of BG (0, 10, 20, and 30 wt.%) were investigated. The pure calcium sulfate cement was set at approximately 180 min, whereas the setting time was drastically decreased to 6 min by replacing 30 wt.% glass powder for CSH in the cement solid phase. BG changed the microscopic morphology of the set cement and decreased the size and compaction of the precipitated gypsum phase. Replacing the CSH phase with BG increased injection force of the produced cement; however, all the cements were injected at a nearly constant force, lower than 20 N. The viscosity measurements in oscillatory mode determined the shear-thinning behavior of the pastes. Although the viscosity of the pastes increased with increasing BG content, it was influenced by the frequency extent. Pure calcium sulfate cement exhibited some transient cytotoxicity on human-derived bone mesenchymal stem cells and it was compensated by introducing BG phase. Moreover, BG improved the cell proliferation and mineralization of extracellular matrix as shown by calcein measurements. The results indicate the injectable composite cement comprising 70 wt.% CSH and 30 wt.% Mg, Sr-doped BG has better setting, mechanical and cellular behaviors and hence, is a potential candidate for bone repair, however more animal and human clinical evaluations are essential.
Collapse
Affiliation(s)
- Nazanin Moazeni
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj 31779-83634, Alborz, Iran; (N.M.); (A.B.)
| | - Saeed Hesaraki
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj 31779-83634, Alborz, Iran; (N.M.); (A.B.)
| | - Aliasghar Behnamghader
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj 31779-83634, Alborz, Iran; (N.M.); (A.B.)
| | - Javad Esmaeilzadeh
- Department of Materials and Chemical Engineering, Esfarayen University of Technology, Esfarayen 96619-98195, North Khorasan, Iran;
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain;
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | | | - Shokoufeh Borhan
- Department of Materials, Chemical and Polymer Engineering, Buein Zahra Technical University, Buein Zahra 34518-66391, Qazvin, Iran;
| |
Collapse
|
48
|
Alonso-Fernández I, Haugen HJ, López-Peña M, González-Cantalapiedra A, Muñoz F. Use of 3D-printed polylactic acid/bioceramic composite scaffolds for bone tissue engineering in preclinical in vivo studies: A systematic review. Acta Biomater 2023; 168:1-21. [PMID: 37454707 DOI: 10.1016/j.actbio.2023.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
3D-printed composite scaffolds have emerged as an alternative to deal with existing limitations when facing bone reconstruction. The aim of the study was to systematically review the feasibility of using PLA/bioceramic composite scaffolds manufactured by 3D-printing technologies as bone grafting materials in preclinical in vivo studies. Electronic databases were searched using specific search terms, and thirteen manuscripts were selected after screening. The synthesis of the scaffolds was carried out using mainly extrusion-based techniques. Likewise, hydroxyapatite was the most used bioceramic for synthesizing composites with a PLA matrix. Among the selected studies, seven were conducted in rats and six in rabbits, but the high variability that exists regarding the experimental process made it difficult to compare them. Regarding the results, PLA/Bioceramic composite scaffolds have shown to be biocompatible and mechanically resistant. Preclinical studies elucidated the ability of the scaffolds to be used as bone grafts, allowing bone growing without adverse reactions. In conclusion, PLA/Bioceramics scaffolds have been demonstrated to be a promising alternative for treating bone defects. Nevertheless, more care should be taken when designing and performing in vivo trials, since the lack of standardization of the processes, which prevents the comparison of the results and reduces the quality of the information. STATEMENT OF SIGNIFICANCE: 3D-printed polylactic acid/bioceramic composite scaffolds have emerged as an alternative to deal with existing limitations when facing bone reconstruction. Since preclinical in vivo studies with animal models represent a mandatory step for clinical translation, the present manuscript analyzed and discussed not only those aspects related to the selection of the bioceramic material, the synthesis of the implants and their characterization. But provides a new approach to understand how the design and perform of clinical trials, as well as the selection of the analysis methods, may affect the obtained results, by covering authors' knowledgebase from veterinary medicine to biomaterial science. Thus, this study aims to systematically review the feasibility of using polylactic acid/bioceramic scaffolds as grafting materials in preclinical trials.
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.
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - 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
| | - 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
| | - Fernando Muñoz
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain
| |
Collapse
|
49
|
Hatt LP, Wirth S, Ristaniemi A, Ciric DJ, Thompson K, Eglin D, Stoddart MJ, Armiento AR. Micro-porous PLGA/ β-TCP/TPU scaffolds prepared by solvent-based 3D printing for bone tissue engineering purposes. Regen Biomater 2023; 10:rbad084. [PMID: 37936893 PMCID: PMC10627288 DOI: 10.1093/rb/rbad084] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 11/09/2023] Open
Abstract
The 3D printing process of fused deposition modelling is an attractive fabrication approach to create tissue-engineered bone substitutes to regenerate large mandibular bone defects, but often lacks desired surface porosity for enhanced protein adsorption and cell adhesion. Solvent-based printing leads to the spontaneous formation of micropores on the scaffold's surface upon solvent removal, without the need for further post processing. Our aim is to create and characterize porous scaffolds using a new formulation composed of mechanically stable poly(lactic-co-glycol acid) and osteoconductive β-tricalcium phosphate with and without the addition of elastic thermoplastic polyurethane prepared by solvent-based 3D-printing technique. Large-scale regenerative scaffolds can be 3D-printed with adequate fidelity and show porosity at multiple levels analysed via micro-computer tomography, scanning electron microscopy and N2 sorption. Superior mechanical properties compared to a commercially available calcium phosphate ink are demonstrated in compression and screw pull out tests. Biological assessments including cell activity assay and live-dead staining prove the scaffold's cytocompatibility. Osteoconductive properties are demonstrated by performing an osteogenic differentiation assay with primary human bone marrow mesenchymal stromal cells. We propose a versatile fabrication process to create porous 3D-printed scaffolds with adequate mechanical stability and osteoconductivity, both important characteristics for segmental mandibular bone reconstruction.
Collapse
Affiliation(s)
- Luan P Hatt
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
- Institute for Biomechanics, ETH Zürich, 8093 Zürich, Switzerland
| | - Sylvie Wirth
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
- Institute for Biomechanics, ETH Zürich, 8093 Zürich, Switzerland
| | | | - Daniel J Ciric
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - Keith Thompson
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
- UCB Pharma, SL1 3WE Slough, UK
| | - David Eglin
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
- Mines Saint-Étienne, Université de Lyon, Université Jean Monnet, INSERM, U1059, 42023 Sainbiose, Saint-Étienne, France
| | - Martin J Stoddart
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
- Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
| | - Angela R Armiento
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
- UCB Pharma, SL1 3WE Slough, UK
| |
Collapse
|
50
|
Zarei M, Sayedain SS, Askarinya A, Sabbaghi M, Alizadeh R. Improving physio-mechanical and biological properties of 3D-printed PLA scaffolds via in-situ argon cold plasma treatment. Sci Rep 2023; 13:14120. [PMID: 37644122 PMCID: PMC10465552 DOI: 10.1038/s41598-023-41226-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
As a bone tissue engineering material, polylactic acid (PLA) has received significant attention and interest due to its ease of processing and biocompatibility. However, its insufficient mechanical properties and poor wettability are two major drawbacks that limit its extensive use. For this purpose, the present study uses in-situ cold argon plasma treatment coupled with a fused deposition modeling printer to enhance the physio-mechanical and biological behavior of 3D-printed PLA scaffolds. Following plasma treatment, field emission scanning electron microscopy images indicated that the surface of the modified scaffold became rough, and the interlayer bonding was enhanced. This resulted in an improvement in the tensile properties of samples printed in the X, Y, and Z directions, with the enhancement being more significant in the Z direction. Additionally, the root mean square value of PLA scaffolds increased (up to 70-fold) after plasma treatment. X-ray photoelectron spectroscopy analysis demonstrated that the plasma technique increased the intensity of oxygen-containing bonds, thereby reducing the water contact angle from 92.5° to 42.1°. The in-vitro degradation study also demonstrated that argon plasma treatment resulted in a 77% increase in PLA scaffold degradation rate. Furthermore, the modified scaffold improved the viability, attachment, and proliferation of human adipose-derived stem cells. These findings suggest that in-situ argon plasma treatment may be a facile and effective method for improving the properties of 3D-printed parts for bone tissue engineering and other applications.
Collapse
Affiliation(s)
- Masoud Zarei
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Sayed Shahab Sayedain
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Amirhossein Askarinya
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Mobina Sabbaghi
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Reza Alizadeh
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran.
| |
Collapse
|