1
|
Khosronejad A, Arabion H, Iraji A, Mokhtarzadegan M, Daneshi SS, Asadi-Yousefabad SL, Zare S, Nowzari F, Abbaspour S, Akbarizadeh F, Aliabadi E, Amiri MA, Zarei M, Ebrahimi R, Mussin NM, Kurmanalina MA, Tanideh N, Tamadon A. Mandibular bone defect healing using polylactic acid-nano-hydroxyapatite-gelatin scaffold loaded with hesperidin and dental pulp stem cells in rat. Tissue Cell 2024; 93:102700. [PMID: 39724839 DOI: 10.1016/j.tice.2024.102700] [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: 09/11/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Addressing mandibular defects poses a significant challenge in maxillofacial surgery. Recent advancements have led to the development of various biomimetic composite scaffolds aimed at facilitating mandibular defect reconstruction. This study aimed to assess the regenerative potential of a novel composite scaffold consisting of polylactic acid (PLA), hydroxyapatite nanoparticles (n-HA), gelatin, hesperidin, and human dental pulp stem cells (DPSCs) in a rat model of mandibular bone defect. The PLA-HA-GLA composite was synthesized using solvent casting-leaching and freeze-drying methods and subsequently treated with 11 mg of hesperidin. The physicochemical properties of the PLA-HA-GLA and PLA-HA-GLA-HIS composites were analyzed by scanning electron microscopy (SEM), X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR), and thermal gravimetric analysis (TGA). Additionally, the mechanical properties and cytotoxicity of DPSCs were assessed. Subsequently, PLA-HA-GLA and PLA-HA-GLA-HIS scaffolds with or without DPSCs were implanted into mandibular bone defects in rats, followed by histopathological, histomorphometric, and cone-beam computed tomography (CBCT) evaluations after eight weeks. SEM analysis revealed the porous structure of the fabricated PLA-HA-GLA and PLA-HA-GLA-HIS composites without aggregation. FTIR and XRD analyses confirmed the presence of functional groups and elements associated with PLA, HA, GLA, and hesperidin in the composites. Although the PLA-HA-GLA-HIS composite exhibited good thermal stability, its mechanical properties decreased after the addition of hesperidin. The cell viability of DPSCs on the surface of the PLA-HA-GLA-HIS scaffolds was statistically significant compared to that of the control group. Furthermore, histopathological, histomorphometric, and radiological evaluations demonstrated that the implantation of the DPSC-loaded PLA-HA-GLA-HIS scaffold had a beneficial effect on bone tissue reconstruction in rats with mandibular defects. These findings highlight the potential of DPSC-loaded PLA-HA-GLA-HIS composite scaffolds for spongy bone tissue engineering and mandibular defect repair.
Collapse
Affiliation(s)
- Arya Khosronejad
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Science, Shiraz, Iran
| | - Hamidreza Arabion
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Science, Shiraz, Iran.
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Central Research laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohamad Mokhtarzadegan
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Seyyed Sajad Daneshi
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fariborz Nowzari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shekofeh Abbaspour
- Department of Chemical and Polymer Engineering, Faculty of Engineering, Yazd University, Yazd, Iran
| | - Fatemeh Akbarizadeh
- Department of Oral & Maxillofacial Radiology, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ehsan Aliabadi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Science, Shiraz, Iran.
| | | | - Moein Zarei
- Department of Polymer and Biomaterials Science, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology, Szczecin, Al. Piastow 45, Szczecin 71-311, Poland.
| | - Reyhaneh Ebrahimi
- Department of Periodontics, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nadiar M Mussin
- Department of Surgery No. 2, West Kazakhstan Medical University, Aktobe, Kazakhstan.
| | - Madina A Kurmanalina
- Department of Therapeutic and Prosthetic Dentistry, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amin Tamadon
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan.
| |
Collapse
|
2
|
Cheng QS, Xu PY, Luo SC, Chen AZ. Advances in Adhesive Materials for Oral and Maxillofacial Soft Tissue Diseases. Macromol Biosci 2024:e2400494. [PMID: 39588806 DOI: 10.1002/mabi.202400494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Oral diseases represent a prevalent global health burden, profoundly affecting patients' quality of life. Given the involvement of oral mucosa and muscles in diverse physiological functions, coupled with clinical aesthetics considerations, repairing oral and maxillofacial soft tissue defects poses a formidable challenge. Wet-adhesive materials are regarded as promising oral repair materials due to their unique advantages in easily overcoming physical and biological barriers in the oral cavity. This review first introduces the intricate wet-state environment prevalent in the oral cavity, meticulously explaining the fundamental physical and chemical adhesion mechanisms that underpin adhesive materials. It then comprehensively summarizes the diverse types of adhesives utilized in stomatology, encompassing polysaccharide, protein, and synthetic polymer adhesive materials. The review further evaluates the latest research advancements in utilizing these materials to treat various oral and maxillofacial soft tissue diseases, including oral mucosal diseases, periodontitis, peri-implantitis, oral and maxillofacial skin defects, and maxillofacial tumors. Finally, it also highlights the promising future prospects and pivotal challenges related to stomatology application of multifunctional adhesive materials.
Collapse
Affiliation(s)
- Qiu-Shuang Cheng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian, 361021, P. R. China
| | - Pei-Yao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian, 361021, P. R. China
| | - Sheng-Chang Luo
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian, 361021, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian, 361021, P. R. China
| |
Collapse
|
3
|
Villani C, Murugan P, George A. Exosome-Laden Hydrogels as Promising Carriers for Oral and Bone Tissue Engineering: Insight into Cell-Free Drug Delivery. Int J Mol Sci 2024; 25:11092. [PMID: 39456873 PMCID: PMC11508290 DOI: 10.3390/ijms252011092] [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: 09/07/2024] [Revised: 10/08/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Mineralization is a key biological process that is required for the development and repair of tissues such as teeth, bone and cartilage. Exosomes (Exo) are a subset of extracellular vesicles (~50-150 nm) that are secreted by cells and contain genetic material, proteins, lipids, nucleic acids, and other biological substances that have been extensively researched for bone and oral tissue regeneration. However, Exo-free biomaterials or exosome treatments exhibit poor bioavailability and lack controlled release mechanisms at the target site during tissue regeneration. By encapsulating the Exos into biomaterials like hydrogels, these disadvantages can be mitigated. Several tissue engineering approaches, such as those for wound healing processes in diabetes mellitus, treatment of osteoarthritis (OA) and cartilage degeneration, repair of intervertebral disc degeneration, and cardiovascular diseases, etc., have been exploited to deliver exosomes containing a variety of therapeutic and diagnostic cargos to target tissues. Despite the significant efficacy of Exo-laden hydrogels, their use in mineralized tissues, such as oral and bone tissue, is very sparse. This review aims to explore and summarize the literature related to the therapeutic potential of hydrogel-encapsulated exosomes for bone and oral tissue engineering and provides insight and practical procedures for the development of future clinical techniques.
Collapse
Affiliation(s)
| | | | - Anne George
- Brodie Tooth Development Genetics & Regenerative Medicine Research Laboratory, Department of Oral Biology, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.V.); (P.M.)
| |
Collapse
|
4
|
Sexton B, Han Y, Dal-Fabbro R, Xu J, Kaigler D, Bottino MC. The role of fibroblast growth factor-2 in modulating the differentiation of periodontal ligament and alveolar bone-derived stem cells. Arch Oral Biol 2024; 165:106027. [PMID: 38870610 DOI: 10.1016/j.archoralbio.2024.106027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE This study examined how range concentrations of Fibroblast Growth Factor-2 (FGF-2) influence the differentiation and activity of human-derived periodontal ligament (hPDLSCs) and alveolar bone-derived stem cells (haBMSCs). DESIGN hPDLSCs and haBMSCs were cultured with varying concentrations of FGF-2 (0, 1, 2.5, 5, 10, 20 ng/mL) and monitored for osteogenic differentiation through alkaline phosphatase (ALP) activity and quantification of gene expression (qRT-PCR) for osteogenesis markers. Additionally, alizarin red staining and a hydroxyproline colorimetric assay evaluated and quantified osteogenic matrix mineralization and collagen deposition. Statistical analyses were performed using one-way ANOVA or two-way ANOVA for multiple comparisons between groups. RESULTS At low FGF-2 concentrations, hPDLSCs differentiated toward an osteogenic lineage, whereas higher concentrations of FGF-2 inhibited osteogenesis and promoted fibroblastic differentiation. The effect of FGF-2 at the lowest concentration tested (1 ng/mL) led to significantly higher ALP activity than osteogenically induced positive controls at early time points and equivalent RUNX2 expression at early and later time points. FGF-2 supplementation of haBMSC cultures was sufficient, at all concentrations, to increase ALP activity at an earlier time point. Mineralization of haBMSC cultures increased significantly within 5-20 ng/mL FGF-2 concentrations under basal growth media conditions (α-minimal essential medium supplemented with 15 % fetal bovine serum and 1 % penicillin/streptomycin). CONCLUSIONS FGF-2 has a dual capacity in promoting osteogenic and fibroblastic differentiation within hPDLSCs contingent upon the dosage and timing of administration, alongside supporting osteogenic differentiation in haBMSCs. These findings underscore the need for precision growth factors dosing when considering the design of biomaterials for periodontal regeneration.
Collapse
Affiliation(s)
- Benjamin Sexton
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Yuanyuan Han
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Jinping Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Darnell Kaigler
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
5
|
Thonglam J, Nuntanaranont T, Kong X, Meesane J. Tissue scaffolds mimicking hierarchical bone morphology as biomaterials for oral maxillofacial surgery with augmentation: structure, properties, and performance evaluation for in vitrotesting. Biomed Mater 2024; 19:055035. [PMID: 39094618 DOI: 10.1088/1748-605x/ad6ac4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/02/2024] [Indexed: 08/04/2024]
Abstract
In this study, tissue scaffolds mimicking hierarchical morphology are constructed and proposed for bone augmentation. The scaffolds are fabricated using lyophilization, before coating them with collagen (Col). Subsequently, the Col-coated scaffolds undergo a second lyophilization, followed by silk fibroin (SF) coating, and a third lyophilization. Thereafter, the scaffolds are divided into six groups with varying ratios of Col to SF: Col/SF = 7:3, 5:5, 3:7, 10:0, and 0:10, with an SF scaffold serving as the control group. The scaffold morphology is examined using a scanning electron microscope, while molecular and structural formations are characterized by Fourier transform infrared spectrometer and differential scanning calorimeter, respectively. Physical and mechanical properties including swelling and compression are tested. Biological functions are assessed throughin vitroosteoblast cell culturing. Biomarkers indicative of bone formation-cell viability and proliferation, alkaline phosphatase activity, and calcium content-are analyzed. Results demonstrate that scaffolds coated with Col and SF exhibit sub-porous formations within the main pore. The molecular formation reveals interactions between the hydrophilic groups of Col and SF. The scaffold structure contains bound water and SF formation gets disrupted by Col. Physical and mechanical properties are influenced by the Col/SF ratio and morphology due to coating. The biological functions of scaffolds with Col and SF coating show enhanced potential for promoting bone tissue formation, particularly the Col/SF (7:3) ratio, which is most suitable for bone augmentation in small defect areas.
Collapse
Affiliation(s)
- Jutakan Thonglam
- Institute of Biomedical Engineering, Department of Biomedical Science and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Thongchai Nuntanaranont
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Jirut Meesane
- Institute of Biomedical Engineering, Department of Biomedical Science and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| |
Collapse
|
6
|
Visan AI, Negut I. Development and Applications of PLGA Hydrogels for Sustained Delivery of Therapeutic Agents. Gels 2024; 10:497. [PMID: 39195026 DOI: 10.3390/gels10080497] [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: 07/02/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) hydrogels are highly utilized in biomedical research due to their biocompatibility, biodegradability, and other versatile properties. This review comprehensively explores their synthesis, properties, sustained release mechanisms, and applications in drug delivery. The introduction underscores the significance of PLGA hydrogels in addressing challenges like short half-lives and systemic toxicity in conventional drug formulations. Synthesis methods, including emulsion solvent evaporation, solvent casting, electrospinning, thermal gelation, and photopolymerization, are described in detail and their role in tailoring hydrogel properties for specific applications is highlighted. Sustained release mechanisms-such as diffusion-controlled, degradation-controlled, swelling-controlled, and combined systems-are analyzed alongside key kinetic models (zero-order, first-order, Higuchi, and Peppas models) for designing controlled drug delivery systems. Applications of PLGA hydrogels in drug delivery are discussed, highlighting their effectiveness in localized and sustained chemotherapy for cancer, as well as in the delivery of antibiotics and antimicrobials to combat infections. Challenges and future prospects in PLGA hydrogel research are discussed, with a focus on improving drug loading efficiency, improving release control mechanisms, and promoting clinical translation. In summary, PLGA hydrogels provide a promising platform for the sustained delivery of therapeutic agents and meet diverse biomedical requirements. Future advancements in materials science and biomedical engineering are anticipated to further optimize their efficacy and applicability in clinical settings. This review consolidates the current understanding and outlines future research directions for PLGA hydrogels, emphasizing their potential to revolutionize therapeutic delivery and improve patient outcomes.
Collapse
Affiliation(s)
- Anita Ioana Visan
- National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania
| | - Irina Negut
- National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania
| |
Collapse
|
7
|
Shopova D, Yaneva A, Mihaylova A, Dinkova A, Bakova D. Unlocking the Future: Bioprinting Salivary Glands-From Possibility to Reality. J Funct Biomater 2024; 15:151. [PMID: 38921525 PMCID: PMC11204800 DOI: 10.3390/jfb15060151] [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: 05/08/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
Salivary gland biofabrication represents a promising avenue in regenerative medicine, aiming to address the challenges of salivary gland dysfunction caused by various factors such as autoimmune diseases and radiotherapy. This review examines the current state of bioprinting technology, biomaterials, and tissue engineering strategies in the context of creating functional, implantable salivary gland constructs. Key considerations include achieving vascularization for proper nutrient supply, maintaining cell viability and functionality during printing, and promoting tissue maturation and integration with surrounding tissues. Despite the existing challenges, recent advancements offer significant potential for the development of personalized therapeutic options to treat salivary gland disorders. Continued research and innovation in this field hold the potential to revolutionize the management of salivary gland conditions, improving patient outcomes and quality of life. This systematic review covers publications from 2018 to April 2024 and was conducted on four databases: Google Scholar, PubMed, EBSCOhost, and Web of Science. The key features necessary for the successful creation, implantation and functioning of bioprinted salivary glands are addressed.
Collapse
Affiliation(s)
- Dobromira Shopova
- Department of Prosthetic Dentistry, Faculty of Dental Medicine, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria
| | - Antoniya Yaneva
- Department of Medical Informatics, Biostatistics and eLearning, Faculty of Public Health, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Anna Mihaylova
- Department of Healthcare Management, Faculty of Public Health, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria; (A.M.); (D.B.)
| | - Atanaska Dinkova
- Department of Oral Surgery, Faculty of Dental Medicine, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Desislava Bakova
- Department of Healthcare Management, Faculty of Public Health, Medical University-Plovdiv, 4000 Plovdiv, Bulgaria; (A.M.); (D.B.)
| |
Collapse
|
8
|
El-Haddad K, El-Zainy MA, Nagy M, Fathy I. De novo regeneration of dentin pulp complex mediated by Adipose derived stem cells in an immunodeficient albino rat model (Histological, histochemical and scanning electron microscopic Study). Saudi Dent J 2024; 36:899-904. [PMID: 38883895 PMCID: PMC11178961 DOI: 10.1016/j.sdentj.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 06/18/2024] Open
Abstract
Background Dental tissue engineering is an alternative procedure for restoring damaged dental tissues. Adipose-derived stem cells are a new source of cells for regenerative endodontics in combination with scaffold materials. The descriptive data about this regenerative process is still insufficient. Objective To evaluate the regenerative potential of Adipose-derived stem cells using a self-assembling polypeptide scaffold for the dentin-pulp complex in an emptied root canal space. Material and Methods 40 root segments of human single-rooted teeth were transplanted into the albino rats' dorsal subcutaneous tissue. Root segments were divided into two groups: group I contained only a self-assembling polypeptide scaffold, and group II contained fluorescent-labeled Adipose-derived stem cells embedded in a self-assembling polypeptide scaffold. The newly formed tissues were assessed on the 60th and 90th days post-transplantation using routine histological examination, Masson trichrome staining, and scanning electron microscopy. Results Group I showed granulation tissue without any signs of predentin formation or odontoblast-like cells. Group II revealed the presence of predentin tissue along the dentin margin, with arranged odontoblast-like cells. An organized connective tissue with abundant vasculature and calcific masses was observed in the pulp space. Conclusion Adipose-derived stem cells can be considered as alternative stem cells for regenerating the dentin-pulp complex. Dentin pulp complex regeneration utilizing a self-assembling polypeptide scaffold alone would not yield successful results.
Collapse
Affiliation(s)
- Khaled El-Haddad
- Department of Basic Oral Medical Sciences, College of Dentistry, Qassim University, Kingdom of Saudi Arabia, Qassim Region, Al-Mulida, Saudi Arabia
- Department of Oral Biology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Medhat A El-Zainy
- Department of Oral Biology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Mohamed Nagy
- Department of Endodontics, Faculty of Dentistry, Ain-Shams University, Cairo, Egypt
| | - Iman Fathy
- Department of Oral Biology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Angjelova A, Jovanova E, Polizzi A, Annunziata M, Laganà L, Santonocito S, Isola G. Insights and Advancements in Periodontal Tissue Engineering and Bone Regeneration. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:773. [PMID: 38792956 PMCID: PMC11123221 DOI: 10.3390/medicina60050773] [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: 03/14/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
The regeneration of periodontal bone defects continues to be an essential therapeutic concern in dental biomaterials. Numerous biomaterials have been utilized in this sector so far. However, the immune response and vascularity in defect regions may be disregarded when evaluating the effectiveness of biomaterials for bone repair. Among several regenerative treatments, the most recent technique of in situ tissue engineering stands out for its ability to replicate endogenous restorative processes by combining scaffold with particular growth factors. Regenerative medicine solutions that combine biomaterials/scaffolds, cells, and bioactive substances have attracted significant interest, particularly for bone repair and regeneration. Dental stem cells (DSCs) share the same progenitor and immunomodulatory properties as other types of MSCs, and because they are easily isolable, they are regarded as desirable therapeutic agents in regenerative dentistry. Recent research has demonstrated that DSCs sown on newly designed synthetic bio-material scaffolds preserve their proliferative capacity while exhibiting increased differentiation and immuno-suppressive capabilities. As researchers discovered how short peptide sequences modify the adhesion and proliferative capacities of scaffolds by activating or inhibiting conventional osteogenic pathways, the scaffolds became more effective at priming MSCs. In this review, the many components of tissue engineering applied to bone engineering will be examined, and the impact of biomaterials on periodontal regeneration and bone cellular biology/molecular genetics will be addressed and updated.
Collapse
Affiliation(s)
- Angela Angjelova
- University Dental Clinical Center St. Pantelejmon, Skopje, Faculty of Dentistry, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia; (A.A.); (E.J.)
| | - Elena Jovanova
- University Dental Clinical Center St. Pantelejmon, Skopje, Faculty of Dentistry, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia; (A.A.); (E.J.)
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy; (L.L.); (S.S.)
| | - Marco Annunziata
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via L. De Crecchio, 6, 80138 Naples, Italy;
| | - Ludovica Laganà
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy; (L.L.); (S.S.)
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy; (L.L.); (S.S.)
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy; (L.L.); (S.S.)
| |
Collapse
|
10
|
Theodosaki AM, Tzemi M, Galanis N, Bakopoulou A, Kotsiomiti E, Aggelidou E, Kritis A. Bone Regeneration with Mesenchymal Stem Cells in Scaffolds: Systematic Review of Human Clinical Trials. Stem Cell Rev Rep 2024; 20:938-966. [PMID: 38407793 PMCID: PMC11087324 DOI: 10.1007/s12015-024-10696-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
The aim of the study is to determine the effectiveness of stem cells in scaffolds in the treatment of bone deficits, in regard of bone regeneration, safety, rehabilitation and quality of life in humans. The systematic review was conducted in accordance with PRISMA 2020. A systematic search was conducted in three search engines and two registries lastly in 29-9-2022.for studies of the last 15 years. The risk of bias was assessed with RoB-2, ROBINS- I and NIH Quality of Before-After (Pre-Post) Studies with no Control group. The certainty of the results was assessed with the GRADE assessment tool. Due to heterogeneity, the results were reported in tables, graphs and narratively. The study protocol was published in PROSPERO with registration number CRD42022359049. Of the 10,091 studies retrieved, 14 were meeting the inclusion criteria, and were qualitatively analyzed. 138 patients were treated with mesenchymal stem cells in scaffolds, showing bone healing in all cases, and even with better results than the standard care. The adverse events were mild in most cases and in accordance with the surgery received. When assessed, there was a rehabilitation of the deficit and a gain in quality of life was detected. Although the heterogeneity between the studies and the small number of patients, the administration of mesenchymal stem cells in scaffolds seems safe and effective in the regeneration of bone defects. These results pave the way for the conduction of more clinical trials, with greater number of participants, with more standardized procedures.
Collapse
Affiliation(s)
- Astero Maria Theodosaki
- Research Methodology in Medicine and Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
- Regenerative Medicine Center, Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece.
- Postgraduate program of Research Methodology in Medicine and Health Sciences, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
- , Thessaloniki, Greece.
| | - Maria Tzemi
- Research Methodology in Medicine and Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Postgraduate program of Research Methodology in Medicine and Health Sciences, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikiforos Galanis
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- 1st Orthopaedic Department, George Papanikolaou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, Faculty of Dentistry, Aristotle University of Thessaloniki, University Campus, Dentistry Building, 54124, Thessaloniki, Greece
- Regenerative Medicine Center, Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| | - Eleni Kotsiomiti
- Department of Prosthodontics, Faculty of Dentistry, Aristotle University of Thessaloniki, University Campus, Dentistry Building, 54124, Thessaloniki, Greece
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, University Campus, 54006, Thessaloniki, Greece
- Regenerative Medicine Center, Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, University Campus, 54006, Thessaloniki, Greece
- Regenerative Medicine Center, Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| |
Collapse
|
11
|
Alnasser M, Alshammari AH, Siddiqui AY, Alothmani OS, Issrani R, Iqbal A, Khattak O, Prabhu N. Tissue Regeneration on Rise: Dental Hard Tissue Regeneration and Challenges-A Narrative Review. SCIENTIFICA 2024; 2024:9990562. [PMID: 38690100 PMCID: PMC11057954 DOI: 10.1155/2024/9990562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 05/02/2024]
Abstract
Background As people live longer, there is an increasing need for hard tissue regeneration and whole-tooth regeneration. Despite the advancements in the field of medicine, the field of regenerative dentistry is still challenging due to the complexity of dental hard tissues. Cross-disciplinary collaboration among material scientists, cellular biologists, and odontologists aimed at developing strategies and uncovering solutions related to dental tissue regeneration. Methodology. A search of the literature was done for pertinent research. Consistent with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) 2020 Statement, the electronic databases looked at were PubMed, Science Direct, Scopus, and Google Scholar, with the keyword search "hard dental tissue regeneration." Results Database analysis yielded a total of 476 articles. 222 duplicate articles have been removed in total. Articles that have no connection to the directed regeneration of hard dental tissue were disregarded. The review concluded with the inclusion of four studies that were relevant to our research objective. Conclusion Current molecular signaling network investigations and novel viewpoints on cellular heterogeneity have made advancements in understanding of the kinetics of dental hard tissue regeneration possible. Here, we outline the fundamentals of stem hard dental tissue maintenance, regeneration, and repair, as well as recent advancements in the field of hard tissue regeneration. These intriguing findings help establish a framework that will eventually enable basic research findings to be utilized towards oral health-improving medicines.
Collapse
Affiliation(s)
- Muhsen Alnasser
- Department of Restorative Dental Sciences, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
| | | | - Amna Yusuf Siddiqui
- Department of Endodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Osama Shujaa Alothmani
- Department of Endodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rakhi Issrani
- Department of Preventive Dentistry, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Azhar Iqbal
- Department of Restorative Dental Sciences, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
| | - Osama Khattak
- Department of Restorative Dental Sciences, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
| | - Namdeo Prabhu
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
12
|
Sharif H, Ziaei H, Rezaei N. Stem Cell-Based Regenerative Approaches for the Treatment of Cleft Lip and Palate: A Comprehensive Review. Stem Cell Rev Rep 2024; 20:637-655. [PMID: 38270744 DOI: 10.1007/s12015-024-10676-9] [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] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
Cleft lip and/or palate (CLP) is a prevalent congenital craniofacial abnormality that can lead to difficulties in eating, speaking, hearing, and psychological distress. The traditional approach for treating CLP involves bone graft surgery, which has limitations, post-surgical complications, and donor site morbidity. However, regenerative medicine has emerged as a promising alternative, employing a combination of stem cells, growth factors, and scaffolds to promote tissue regeneration. This review aims to provide a comprehensive overview of stem cell-based regenerative approaches in the management of CLP. A thorough search was conducted in the Medline/PubMed and Scopus databases, including cohort studies, randomized controlled trials, case series, case controls, case reports, and animal studies. The identified studies were categorized into two main groups: clinical studies involving human subjects and in vivo studies using animal models. While there are only a limited number of studies investigating the combined use of stem cells and scaffolds for CLP treatment, they have shown promising results. Various types of stem cells have been utilized in conjunction with scaffolds. Importantly, regenerative methods have been successfully applied to patients across a broad range of age groups. The collective findings derived from the reviewed studies consistently support the notion that regenerative medicine holds potential advantages over conventional bone grafting and represents a promising therapeutic option for CLP. However, future well-designed clinical trials, encompassing diverse combinations of stem cells and scaffolds, are warranted to establish the clinical efficacy of these interventions with a larger number of patients.
Collapse
Affiliation(s)
- Helia Sharif
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Dental Society, Faculty of Dentistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Heliya Ziaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, US
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
| |
Collapse
|
13
|
Wang J, Zhao Z, Yang K, Bai Y. Research progress in cell therapy for oral diseases: focus on cell sources and strategies to optimize cell function. Front Bioeng Biotechnol 2024; 12:1340728. [PMID: 38515628 PMCID: PMC10955105 DOI: 10.3389/fbioe.2024.1340728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024] Open
Abstract
In recent years, cell therapy has come to play an important therapeutic role in oral diseases. This paper reviews the active role of mesenchymal stem cells, immune cell sources, and other cells in oral disorders, and presents data supporting the role of cell therapy in oral disorders, including bone and tooth regeneration, oral mucosal disorders, oral soft tissue defects, salivary gland dysfunction, and orthodontic tooth movement. The paper will first review the progress of cell optimization strategies for oral diseases, including the use of hormones in combination with stem cells, gene-modified regulatory cells, epigenetic regulation of cells, drug regulation of cells, cell sheets/aggregates, cell-binding scaffold materials and hydrogels, nanotechnology, and 3D bioprinting of cells. In summary, we will focus on the therapeutic exploration of these different cell sources in oral diseases and the active application of the latest cell optimization strategies.
Collapse
Affiliation(s)
| | | | | | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Salehi Abar E, Vandghanooni S, Torab A, Jaymand M, Eskandani M. A comprehensive review on nanocomposite biomaterials based on gelatin for bone tissue engineering. Int J Biol Macromol 2024; 254:127556. [PMID: 37884249 DOI: 10.1016/j.ijbiomac.2023.127556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
The creation of a suitable scaffold is a crucial step in the process of bone tissue engineering (BTE). The scaffold, acting as an artificial extracellular matrix, plays a significant role in determining the fate of cells by affecting their proliferation and differentiation in BTE. Therefore, careful consideration should be given to the fabrication approach and materials used for scaffold preparation. Natural polypeptides such as gelatin and collagen have been widely used for this purpose. The unique properties of nanoparticles, which vary depending on their size, charge, and physicochemical properties, have demonstrated potential in solving various challenges encountered in BTE. Therefore, nanocomposite biomaterials consisting of polymers and nanoparticles have been extensively used for BTE. Gelatin has also been utilized in combination with other nanomaterials to apply for this purpose. Composites of gelatin with various types of nanoparticles are particularly promising for creating scaffolds with superior biological and physicochemical properties. This review explores the use of nanocomposite biomaterials based on gelatin and various types of nanoparticles together for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Elaheh Salehi Abar
- Department of Prosthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Torab
- Department of Prosthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Urkande NK, Mankar N, Nikhade PP, Chandak M. Understanding the Complexities of Cast Post Retention: A Comprehensive Review of Influential Factors. Cureus 2023; 15:e51258. [PMID: 38288201 PMCID: PMC10823198 DOI: 10.7759/cureus.51258] [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: 11/18/2023] [Accepted: 12/27/2023] [Indexed: 01/31/2024] Open
Abstract
This comprehensive review delves into the intricate landscape of cast post retention in restorative dentistry, encompassing historical perspectives, contemporary techniques, and future directions. Examining factors ranging from tooth-related considerations to prosthesis-related dynamics, the review provides a detailed analysis of clinical techniques, including step-by-step procedures, common challenges, and innovative advancements. Technological breakthroughs, such as digital impressions, computer-aided design and computer-aided manufacturing (CAD/CAM) technology, three-dimensional (3D) printing, and finite element analysis, are explored for their transformative impact on precision and customization. The discussion extends to the promising future of cast post retention, emphasising emerging materials, the integration of artificial intelligence in treatment planning, and patient-specific approaches. Implications for clinical practice underscore the importance of individualised treatment planning and the adoption of advanced technologies. Recommendations for future research advocate for comprehensive long-term clinical studies, investigations into AI-driven treatment planning, and a focus on patient outcomes and satisfaction. This review consolidates existing knowledge and anticipates a future marked by enhanced precision, individualised care, and improved long-term success in cast post-retained restorations.
Collapse
Affiliation(s)
- Neha K Urkande
- Conservative Dentistry and Endodontics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Nikhil Mankar
- Conservative Dentistry and Endodontics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Pradnya P Nikhade
- Conservative Dentistry and Endodontics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Manoj Chandak
- Conservative Dentistry and Endodontics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
16
|
Shopova D, Mihaylova A, Yaneva A, Bakova D. Advancing Dentistry through Bioprinting: Personalization of Oral Tissues. J Funct Biomater 2023; 14:530. [PMID: 37888196 PMCID: PMC10607235 DOI: 10.3390/jfb14100530] [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: 09/16/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Despite significant advancements in dental tissue restoration and the use of prostheses for addressing tooth loss, the prevailing clinical approaches remain somewhat inadequate for replicating native dental tissue characteristics. The emergence of three-dimensional (3D) bioprinting offers a promising innovation within the fields of regenerative medicine and tissue engineering. This technology offers notable precision and efficiency, thereby introducing a fresh avenue for tissue regeneration. Unlike the traditional framework encompassing scaffolds, cells, and signaling factors, 3D bioprinting constitutes a contemporary addition to the arsenal of tissue engineering tools. The ongoing shift from conventional dentistry to a more personalized paradigm, principally under the guidance of bioprinting, is poised to exert a significant influence in the foreseeable future. This systematic review undertakes the task of aggregating and analyzing insights related to the application of bioprinting in the context of regenerative dentistry. Adhering to PRISMA guidelines, an exhaustive literature survey spanning the years 2019 to 2023 was performed across prominent databases including PubMed, Scopus, Google Scholar, and ScienceDirect. The landscape of regenerative dentistry has ushered in novel prospects for dentoalveolar treatments and personalized interventions. This review expounds on contemporary accomplishments and avenues for the regeneration of pulp-dentin, bone, periodontal tissues, and gingival tissues. The progressive strides achieved in the realm of bioprinting hold the potential to not only enhance the quality of life but also to catalyze transformative shifts within the domains of medical and dental practices.
Collapse
Affiliation(s)
- Dobromira Shopova
- Department of Prosthetic Dentistry, Faculty of Dental Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | - Anna Mihaylova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria (D.B.)
| | - Antoniya Yaneva
- Department of Medical Informatics, Biostatistics and eLearning, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Desislava Bakova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria (D.B.)
| |
Collapse
|
17
|
Lyu S, Dong Z, Xu X, Bei HP, Yuen HY, James Cheung CW, Wong MS, He Y, Zhao X. Going below and beyond the surface: Microneedle structure, materials, drugs, fabrication, and applications for wound healing and tissue regeneration. Bioact Mater 2023; 27:303-326. [PMID: 37122902 PMCID: PMC10140753 DOI: 10.1016/j.bioactmat.2023.04.003] [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: 10/11/2022] [Revised: 03/11/2023] [Accepted: 04/02/2023] [Indexed: 05/02/2023] Open
Abstract
Microneedle, as a novel drug delivery system, has attracted widespread attention due to its non-invasiveness, painless and simple administration, controllable drug delivery, and diverse cargo loading capacity. Although microneedles are initially designed to penetrate stratum corneum of skin for transdermal drug delivery, they, recently, have been used to promote wound healing and regeneration of diverse tissues and organs and the results are promising. Despite there are reviews about microneedles, few of them focus on wound healing and tissue regeneration. Here, we review the recent advances of microneedles in this field. We first give an overview of microneedle system in terms of its potential cargos (e.g., small molecules, macromolecules, nucleic acids, nanoparticles, extracellular vesicle, cells), structural designs (e.g., multidrug structures, adhesive structures), material selection, and drug release mechanisms. Then we briefly summarize different microneedle fabrication methods, including their advantages and limitations. We finally summarize the recent progress of microneedle-assisted wound healing and tissue regeneration (e.g., skin, cardiac, bone, tendon, ocular, vascular, oral, hair, spinal cord, and uterine tissues). We expect that our article would serve as a guideline for readers to design their microneedle systems according to different applications, including material selection, drug selection, and structure design, for achieving better healing and regeneration efficacy.
Collapse
Affiliation(s)
- Shang Lyu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, PR China
| | - Zhifei Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
- Faculty of Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Xiaoxiao Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
- Faculty of Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
| | - Ho-Yin Yuen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
| | - Chung-Wai James Cheung
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
| | - Man-Sang Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
- Corresponding author.
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, PR China
- Corresponding author.
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, PR China
- Corresponding author.
| |
Collapse
|
18
|
Lau CS, Chua J, Prasadh S, Lim J, Saigo L, Goh BT. Alveolar Ridge Augmentation with a Novel Combination of 3D-Printed Scaffolds and Adipose-Derived Mesenchymal Stem Cells-A Pilot Study in Pigs. Biomedicines 2023; 11:2274. [PMID: 37626770 PMCID: PMC10452669 DOI: 10.3390/biomedicines11082274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Alveolar ridge augmentation is an important dental procedure to increase the volume of bone tissue in the alveolar ridge before the installation of a dental implant. To meet the high demand for bone grafts for alveolar ridge augmentation and to overcome the limitations of autogenous bone, allografts, and xenografts, researchers are developing bone grafts from synthetic materials using novel fabrication techniques such as 3D printing. To improve the clinical performance of synthetic bone grafts, stem cells with osteogenic differentiation capability can be loaded into the grafts. In this pilot study, we propose a novel bone graft which combines a 3D-printed polycaprolactone-tricalcium phosphate (PCL-TCP) scaffold with adipose-derived mesenchymal stem cells (AD-MSCs) that can be harvested, processed and implanted within the alveolar ridge augmentation surgery. We evaluated the novel bone graft in a porcine lateral alveolar defect model. Radiographic analysis revealed that the addition of AD-MSCs to the PCL-TCP scaffold improved the bone volume in the defect from 18.6% to 28.7% after 3 months of healing. Histological analysis showed the presence of AD-MSCs in the PCL-TCP scaffold led to better formation of new bone and less likelihood of fibrous encapsulation of the scaffold. Our pilot study demonstrated that the loading of AD-MSCs improved the bone regeneration capability of PCL-TCP scaffolds, and our novel bone graft is suitable for alveolar ridge augmentation.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore 168938, Singapore; (C.S.L.); (L.S.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jasper Chua
- Duke-NUS Medical School, Singapore 169857, Singapore;
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Jing Lim
- Osteopore International Pte Ltd., Singapore 618305, Singapore;
| | - Leonardo Saigo
- National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore 168938, Singapore; (C.S.L.); (L.S.)
| | - Bee Tin Goh
- National Dental Research Institute Singapore, National Dental Centre Singapore, Singapore 168938, Singapore; (C.S.L.); (L.S.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
19
|
Kim S, Hwangbo H, Chae S, Lee H. Biopolymers and Their Application in Bioprinting Processes for Dental Tissue Engineering. Pharmaceutics 2023; 15:2118. [PMID: 37631331 PMCID: PMC10457894 DOI: 10.3390/pharmaceutics15082118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Dental tissues are composed of multiple tissues with complex organization, such as dentin, gingiva, periodontal ligament, and alveolar bone. These tissues have different mechanical and biological properties that are essential for their functions. Therefore, dental diseases and injuries pose significant challenges for restorative dentistry, as they require innovative strategies to regenerate damaged or missing dental tissues. Biomimetic bioconstructs that can effectively integrate with native tissues and restore their functionalities are desirable for dental tissue regeneration. However, fabricating such bioconstructs is challenging due to the diversity and complexity of dental tissues. This review provides a comprehensive overview of the recent developments in polymer-based tissue engineering and three-dimensional (3D) printing technologies for dental tissue regeneration. It also discusses the current state-of-the-art, focusing on key techniques, such as polymeric biomaterials and 3D printing with or without cells, used in tissue engineering for dental tissues. Moreover, the final section of this paper identifies the challenges and future directions of this promising research field.
Collapse
Affiliation(s)
- Suhon Kim
- Barun Plant Orthodontics and Dental Clinic, Seongnam 13312, Republic of Korea;
| | - Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; (H.H.); (S.C.)
| | - SooJung Chae
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; (H.H.); (S.C.)
| | - Hyeongjin Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; (H.H.); (S.C.)
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
20
|
Cabaña-Muñoz ME, Pelaz Fernández MJ, Parmigiani-Cabaña JM, Parmigiani-Izquierdo JM, Merino JJ. Adult Mesenchymal Stem Cells from Oral Cavity and Surrounding Areas: Types and Biomedical Applications. Pharmaceutics 2023; 15:2109. [PMID: 37631323 PMCID: PMC10459416 DOI: 10.3390/pharmaceutics15082109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Adult mesenchymal stem cells are those obtained from the conformation of dental structures (DMSC), such as deciduous and permanent teeth and other surrounding tissues. Background: The self-renewal and differentiation capacities of these adult stem cells allow for great clinical potential. Because DMSC are cells of ectomesenchymal origin, they reveal a high capacity for complete regeneration of dental pulp, periodontal tissue, and other biomedical applications; their differentiation into other types of cells promotes repair in muscle tissue, cardiac, pancreatic, nervous, bone, cartilage, skin, and corneal tissues, among others, with a high predictability of success. Therefore, stem and progenitor cells, with their exosomes of dental origin and surrounding areas in the oral cavity due to their plasticity, are considered a fundamental pillar in medicine and regenerative dentistry. Tissue engineering (MSCs, scaffolds, and bioactive molecules) sustains and induces its multipotent and immunomodulatory effects. It is of vital importance to guarantee the safety and efficacy of the procedures designed for patients, and for this purpose, more clinical trials are needed to increase the efficacy of several pathologies. Conclusion: From a bioethical and transcendental anthropological point of view, the human person as a unique being facilitates better clinical and personalized therapy, given the higher prevalence of dental and chronic systemic diseases.
Collapse
Affiliation(s)
- María Eugenia Cabaña-Muñoz
- CIROM—Centro de Rehabilitación Oral Multidisciplinaria, 30001 Murcia, Spain; (M.E.C.-M.); (J.M.P.-C.); (J.M.P.-I.)
| | | | - José María Parmigiani-Cabaña
- CIROM—Centro de Rehabilitación Oral Multidisciplinaria, 30001 Murcia, Spain; (M.E.C.-M.); (J.M.P.-C.); (J.M.P.-I.)
| | | | - José Joaquín Merino
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (U.C.M), 28040 Madrid, Spain
| |
Collapse
|
21
|
Hao M, Wang D, Duan M, Kan S, Li S, Wu H, Xiang J, Liu W. Functional drug-delivery hydrogels for oral and maxillofacial wound healing. Front Bioeng Biotechnol 2023; 11:1241660. [PMID: 37600316 PMCID: PMC10434880 DOI: 10.3389/fbioe.2023.1241660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
The repair process for oral and maxillofacial injuries involves hemostasis, inflammation, proliferation, and remodeling. Injury repair involves a variety of cells, including platelets, immune cells, fibroblasts, and various cytokines. Rapid and adequate healing of oral and maxillofacial trauma is a major concern to patients. Functional drug-delivery hydrogels play an active role in promoting wound healing and have shown unique advantages in wound dressings. Functional hydrogels promote wound healing through their adhesive, anti-inflammatory, antioxidant, antibacterial, hemostatic, angiogenic, and re-epithelialization-promoting properties, effectively sealing wounds and reducing inflammation. In addition, functional hydrogels can respond to changes in temperature, light, magnetic fields, pH, and reactive oxygen species to release drugs, enabling precise treatment. Furthermore, hydrogels can deliver various cargos that promote healing, including nucleic acids, cytokines, small-molecule drugs, stem cells, exosomes, and nanomaterials. Therefore, functional drug-delivery hydrogels have a positive impact on the healing of oral and maxillofacial injuries. This review describes the oral mucosal structure and healing process and summarizes the currently available responsive hydrogels used to promote wound healing.
Collapse
Affiliation(s)
- Ming Hao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Mengna Duan
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shaoning Kan
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shuangji Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Han Wu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jingcheng Xiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
22
|
Naguib GH, Abd El-Aziz GS, Almehmadi A, Bayoumi A, Mira AI, Hassan AH, Hamed MT. Evaluation of the time-dependent osteogenic activity of glycerol incorporated magnesium oxide nanoparticles in induced calvarial defects. Heliyon 2023; 9:e18757. [PMID: 37593643 PMCID: PMC10432181 DOI: 10.1016/j.heliyon.2023.e18757] [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: 05/05/2023] [Revised: 07/06/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Magnesium-based biomaterials have been explored for their potential as bone healing materials, as a result of their outstanding biodegradability and biocompatibility. These characteristics make magnesium oxide nanoparticles (MgO NPs) a promising material for treating bone disorders. The purpose of this investigation is to assess the osteogenic activity of newly-developed locally administered glycerol-incorporated MgO NPs (GIMgO NPs) in rabbits' calvarial defects. Materials and methods Characterization of GIMgO was done by X-ray Diffraction (XRD) and Fourier Transform Infrared Spectroscopy (FTIR). Bilateral calvarial defects were created in eighteen New Zealand Rabbits, of which they were divided into 3 groups with time points corresponding to 2, 4, and 6 weeks postoperatively (n = 6). One defect was implanted with absorbable gel foam impregnated with GIMgO NPs while the other was implanted with gel foam soaked with glycerol (the control). The defects were assessed using histological, Micro-Computed Tomography (Micro-CT), and histometric evaluation. Results The characterization of the GIMgO nanogel revealed the presence of MgO NPs and glycerol as well as the formation of the crystalline phase of the MgO NPs within the nanogel sample. The histological and micro-CT analysis showed time-dependent improvement of healing activity in the calvarial defects implanted with GIMgO NPs when compared to the control. Furthermore, the histometric analysis demonstrated a marked increase in the total area of new bone, connective tissue, new bone area and volume in the GIMgO NPs implanted site. Statistically, the amount of new bone formation was more significant at 6 weeks than at 2 and 4 weeks postoperatively in the calvarial defects implanted with GIMgO NPs as compared to the control. Conclusion The locally applied GIMgO NPs demonstrated efficacy in promoting bone formation, with more significant effects observed over an extended period. These findings suggest its suitability for clinical use as a therapeutic alternative to enhance bone healing.
Collapse
Affiliation(s)
- Ghada H. Naguib
- Department of Restorative Dentistry, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Oral Biology, Cairo University School of Dentistry, Cairo, Egypt
| | - Gamal S. Abd El-Aziz
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Almehmadi
- Department of Oral Biology, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amr Bayoumi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulghani I. Mira
- Department of Restorative Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mohamed T. Hamed
- Department of Oral and Maxillofacial Prosthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Fixed Prosthodontics, Cairo University School of Dentistry, Cairo, Egypt
| |
Collapse
|
23
|
Kaczmarek-Szczepańska B, Polkowska I, Małek M, Kluczyński J, Paździor-Czapula K, Wekwejt M, Michno A, Ronowska A, Pałubicka A, Nowicka B, Otrocka-Domagała I. The characterization of collagen-based scaffolds modified with phenolic acids for tissue engineering application. Sci Rep 2023; 13:9966. [PMID: 37340023 DOI: 10.1038/s41598-023-37161-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/16/2023] [Indexed: 06/22/2023] Open
Abstract
The aim of the experiment was to study the morphology of collagen-based scaffolds modified by caffeic acid, ferulic acid, and gallic acid, their swelling, and degradation rate, as well as the biological properties of scaffolds, such as antioxidant activity, hemo- and cytocompatibility, histological observation, and antibacterial properties. Scaffolds based on collagen with phenolic acid showed higher swelling rate and enzymatic stability compared to scaffolds based on pure collagen, and the radical scavenging activity was in the range 85-91%. All scaffolds were non-hemolytic and compatible with surrounding tissues. Collagen modified by ferulic acid showed potentially negative effects on hFOB cells as a significantly increased LDH release was found, but all of the studied materials had antimicrobial activity against Staphylococcus aureus and Escherichia coli. It may be assumed that phenolic acids, such as caffeic, ferulic, and gallic acid, are modifiers and provide novel biological properties of collagen-based scaffolds. This paper provides the summarization and comparison of the biological properties of scaffolds based on collagen modified with three different phenolic acids.
Collapse
Affiliation(s)
- Beata Kaczmarek-Szczepańska
- Department of Biomaterials and Cosmetics Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100, Toruń, Poland.
| | - Izabela Polkowska
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| | - Marcin Małek
- Faculty of Civil Engineering and Geodesy, Military University of Technology, ul. Gen. Sylwestra Kaliskiego 2, 00-908, Warsaw, Poland
| | - Janusz Kluczyński
- Faculty of Mechanical Engineering, Military University of Technology, ul. Gen. Sylwestra Kaliskiego 2, 00-908, Warsaw, Poland
| | - Katarzyna Paździor-Czapula
- Department of Pathological Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-719, Olsztyn, Poland
| | - Marcin Wekwejt
- Department of Biomaterials Technology, Faculty of Mechanical Engineering and Ship Technology, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-229, Gdańsk, Poland
| | - Anna Michno
- Department of Laboratory Medicine, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland
| | - Anna Ronowska
- Department of Laboratory Medicine, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland
| | - Anna Pałubicka
- Department of Laboratory Diagnostics and Microbiology With Blood Bank, Specialist Hospital in Kościerzyna, Alojzego Piechowskiego 36, 83-400, Kościerzyna, Poland
| | - Beata Nowicka
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| | - Iwona Otrocka-Domagała
- Department of Pathological Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-719, Olsztyn, Poland
| |
Collapse
|
24
|
Zong C, Bronckaers A, Willems G, He H, Cadenas de Llano-Pérula M. Nanomaterials for Periodontal Tissue Regeneration: Progress, Challenges and Future Perspectives. J Funct Biomater 2023; 14:290. [PMID: 37367254 DOI: 10.3390/jfb14060290] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Bioactive nanomaterials are increasingly being applied in oral health research. Specifically, they have shown great potential for periodontal tissue regeneration and have substantially improved oral health in translational and clinical applications. However, their limitations and side effects still need to be explored and elucidated. This article aims to review the recent advancements in nanomaterials applied for periodontal tissue regeneration and to discuss future research directions in this field, especially focusing on research using nanomaterials to improve oral health. The biomimetic and physiochemical properties of nanomaterials such as metals and polymer composites are described in detail, including their effects on the regeneration of alveolar bone, periodontal ligament, cementum and gingiva. Finally, the biomedical safety issues of their application as regenerative materials are updated, with a discussion about their complications and future perspectives. Although the applications of bioactive nanomaterials in the oral cavity are still at an initial stage, and pose numerous challenges, recent research suggests that they are a promising alternative in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Chen Zong
- Department of Oral Health Sciences-Orthodontics, University of Leuven (KU Leuven) and Dentistry, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute, Faculty of Life Sciences, University of Hasselt, 3590 Diepenbeek, Belgium
| | - Guy Willems
- Department of Oral Health Sciences-Orthodontics, University of Leuven (KU Leuven) and Dentistry, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Hong He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Orthodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Center for Dentofacial Development and Sleep Medicine, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Maria Cadenas de Llano-Pérula
- Department of Oral Health Sciences-Orthodontics, University of Leuven (KU Leuven) and Dentistry, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
25
|
Cell–scaffold interactions in tissue engineering for oral and craniofacial reconstruction. Bioact Mater 2023; 23:16-44. [DOI: 10.1016/j.bioactmat.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
|
26
|
Zhao Z, Attanasio C, Pedano MS, Cadenas de Llano-Pérula M. Comparison of human dental tissue RNA extraction methods for RNA sequencing. Arch Oral Biol 2023; 148:105646. [PMID: 36812743 DOI: 10.1016/j.archoralbio.2023.105646] [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: 01/02/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/13/2023]
Abstract
OBJECTIVE The purpose of this study was to identify an efficient RNA extraction method for periodontal ligament (PDL) and dental pulp (DP) tissues to be used in RNA sequencing studies, given the increased use of these techniques in dental research and the lack of standard protocols. DESIGN PDL and DP were harvested from extracted third molars. Total RNA was extracted with four RNA extraction kits. RNA concentration, purity and integrity were assessed by means of NanoDrop and Bioanalyzer and statistically compared. RESULTS RNA from PDL was more likely to be degraded than that of DP. The TRIzol method yielded the highest RNA concentration from both tissues. All methods harvested RNA with A260/A280 close to 2.0 and with A260/A230 above 1.5, except for the A260/A230 from PDL obtained with the RNeasy Mini kit. For RNA integrity, the RNeasy Fibrous Tissue Mini kit yielded the highest RIN values and 28 S/18 S from PDL, while the RNeasy Mini kit obtained relatively high RIN values with an appropriate 28 S/18 S for DP. CONCLUSION Significantly different results were obtained for PDL and DP when using the RNeasy Mini kit. The RNeasy Mini kit provided the highest RNA yields and quality for DP, while the RNeasy Fibrous Tissue Mini kit obtained the highest quality RNA from PDL.
Collapse
Affiliation(s)
- Zuodong Zhao
- Department of Oral Health Sciences-Orthodontics, KU Leuven and Dentistry, University Hospitals Leuven, Kapucijnenvoer 7, 3000 Leuven, Belgium.
| | - Catia Attanasio
- Laboratory of Gene Regulation and Disease, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Mariano Simón Pedano
- Department of Oral Health Sciences-Endodontics and BIOMAT - Biomaterials Research group, KU Leuven and Dentistry, University Hospitals Leuven, Kapucijnenvoer 7, 3000 Leuven, Belgium
| | - Maria Cadenas de Llano-Pérula
- Department of Oral Health Sciences-Orthodontics, KU Leuven and Dentistry, University Hospitals Leuven, Kapucijnenvoer 7, 3000 Leuven, Belgium
| |
Collapse
|
27
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Xu T, Xie K, Wang C, Ivanovski S, Zhou Y. Immunomodulatory nanotherapeutic approaches for periodontal tissue regeneration. NANOSCALE 2023; 15:5992-6008. [PMID: 36896757 DOI: 10.1039/d2nr06149j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Periodontitis is an infection-induced inflammatory disease characterized by progressive destruction of tooth supporting tissues, which, if left untreated, can result in tooth loss. The destruction of periodontal tissues is primarily caused by an imbalance between the host immune protection and immune destruction mechanisms. The ultimate goal of periodontal therapy is to eliminate inflammation and promote the repair and regeneration of both hard and soft tissues, so as to restore the physiological structure and function of periodontium. Advancement in nanotechnologies has enabled the development of nanomaterials with immunomodulatory properties for regenerative dentistry. This review discusses the immune mechanisms of the major effector cells in the innate and adaptive immune systems, the physicochemical and biological properties of nanomaterials, and the research advancements in immunomodulatory nanotherapeutic approaches for the management of periodontitis and the regeneration of periodontal tissues. The current challenges, and prospects for future applications of nanomaterials are then discussed so that researchers at the intersections of osteoimmunology, regenerative dentistry and materiobiology will continue to advance the development of nanomaterials for improved periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tian Xu
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Kunke Xie
- Clinical Laboratory, Bo'Ai Hospital of Zhongshan, 6 Chenggui Road, East District, Zhongshan 528403, Guangdong, China
| | - Cong Wang
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Yinghong Zhou
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| |
Collapse
|
29
|
Guo J, Yao H, Li X, Chang L, Wang Z, Zhu W, Su Y, Qin L, Xu J. Advanced Hydrogel systems for mandibular reconstruction. Bioact Mater 2023; 21:175-193. [PMID: 36093328 PMCID: PMC9413641 DOI: 10.1016/j.bioactmat.2022.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/16/2022] [Accepted: 08/02/2022] [Indexed: 12/23/2022] Open
Abstract
Mandibular defect becomes a prevalent maxillofacial disease resulting in mandibular dysfunctions and huge psychological burdens to the patients. Considering the routine presence of oral contaminations and aesthetic restoration of facial structures, the current clinical treatments are however limited, incapable to reconstruct the structural integrity and regeneration, spurring the need for cost-effective mandibular tissue engineering. Hydrogel systems possess great merit for mandibular reconstruction with precise involvement of cells and bioactive factors. In this review, current clinical treatments and distinct mode(s) of mandible formation and pathological resorption are summarized, followed by a review of hydrogel-related mandibular tissue engineering, and an update on the advanced fabrication of hydrogels with improved mechanical property, antibacterial ability, injectable form, and 3D bioprinted hydrogel constructs. The exploration of advanced hydrogel systems will lay down a solid foundation for a bright future with more biocompatible, effective, and personalized treatment in mandibular reconstruction.
Collapse
Affiliation(s)
- Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zixuan Wang
- Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Wangyong Zhu
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yuxiong Su
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Corresponding author. Director of Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Corresponding author. Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
30
|
Fu Y, Huang S, Feng Z, Huang L, Zhang X, Lin H, Mo A. MXene-Functionalized Ferroelectric Nanocomposite Membranes with Modulating Surface Potential Enhance Bone Regeneration. ACS Biomater Sci Eng 2023; 9:900-917. [PMID: 36715700 DOI: 10.1021/acsbiomaterials.2c01174] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rapid and effective bone defect repair remains a challenging issue for clinical treatment. Applying biomaterials with endogenous surface potential has been widely studied to enhance bone regeneration, but how to regulate the electric potential and surface morphology of the implanted materials precisely to achieve an optimal bioelectric microenvironment is still a major challenge. The aim of this study is to develop electroactive biomaterials that better mimic the extracellular microenvironment for bone regeneration. Hence, MXene/polyvinylidene fluoride (MXene/PVDF) ferroelectric nanocomposite membranes were prepared by electrospinning. Physicochemical characterization demonstrated that Ti3C2Tx MXene nanosheets were wrapped in PVDF shell layer and the surface morphology and potential were modulated by altering the content of MXene, where uniform distribution of fibers and enhanced electric potential can be obtained and precisely assembled into a natural extracellular matrix (ECM) in bone tissue. Consequently, the MXene/PVDF membranes facilitated cell adhesion, stretching, and growth, showing good biocompatibility; meanwhile, their intrinsic electric potential promoted the recruitment of osteogenic cells and accelerated the differentiation of osteoblast. Furthermore, 1 wt % MXene/PVDF membrane with a suitable surface potential and better topographical structure for bone regeneration qualitatively and quantitatively promoted bone tissue formation in a rat calvarial bone defect after 4 and 8 weeks of healing. The fabricated MXene/PVDF ferroelectric nanocomposite membranes show a biomimetic microenvironment with a sustainable electric potential and optimal 3D topographical structure, providing an innovative and well-suited strategy for application in bone regeneration.
Collapse
Affiliation(s)
- Yu Fu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| | - Si Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| | - Zeru Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| | - Lirong Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| | - Xiaoqing Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| | - Hua Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| | - Anchun Mo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, 14th 3 sect of Renmin South Road, Chengdu610041, China
| |
Collapse
|
31
|
Kwan JC, Dondani J, Iyer J, Muaddi HA, Nguyen TT, Tran SD. Biomimicry and 3D-Printing of Mussel Adhesive Proteins for Regeneration of the Periodontium-A Review. Biomimetics (Basel) 2023; 8:biomimetics8010078. [PMID: 36810409 PMCID: PMC9944831 DOI: 10.3390/biomimetics8010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Innovation in the healthcare profession to solve complex human problems has always been emulated and based on solutions proven by nature. The conception of different biomimetic materials has allowed for extensive research that spans several fields, including biomechanics, material sciences, and microbiology. Due to the atypical characteristics of these biomaterials, dentistry can benefit from these applications in tissue engineering, regeneration, and replacement. This review highlights an overview of the application of different biomimetic biomaterials in dentistry and discusses the key biomaterials (hydroxyapatite, collagen, polymers) and biomimetic approaches (3D scaffolds, guided bone and tissue regeneration, bioadhesive gels) that have been researched to treat periodontal and peri-implant diseases in both natural dentition and dental implants. Following this, we focus on the recent novel application of mussel adhesive proteins (MAPs) and their appealing adhesive properties, in addition to their key chemical and structural properties that relate to the engineering, regeneration, and replacement of important anatomical structures in the periodontium, such as the periodontal ligament (PDL). We also outline the potential challenges in employing MAPs as a biomimetic biomaterial in dentistry based on the current evidence in the literature. This provides insight into the possible increased functional longevity of natural dentition that can be translated to implant dentistry in the near future. These strategies, paired with 3D printing and its clinical application in natural dentition and implant dentistry, develop the potential of a biomimetic approach to overcoming clinical problems in dentistry.
Collapse
Affiliation(s)
- Jan C. Kwan
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Jay Dondani
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Janaki Iyer
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Hasan A. Muaddi
- Department of Oral and Maxillofacial Surgery, King Khalid University, Abha 62529, Saudi Arabia
| | - Thomas T. Nguyen
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Division of Periodontics, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Correspondence:
| |
Collapse
|
32
|
Liu C, Sharpe P, Volponi AA. Applications of regenerative techniques in adult orthodontics. FRONTIERS IN DENTAL MEDICINE 2023. [DOI: 10.3389/fdmed.2022.1100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Management of the growing adult orthodontic patient population must contend with challenges particular to orthodontic treatment in adults. These include a limited rate of tooth movement, increased incidence of periodontal complications, higher risk of iatrogenic root resorption and pulp devitalisation, resorbed edentulous ridges, and lack of growth potential. The field of regenerative dentistry has evolved numerous methods of manipulating cellular and molecular processes to rebuild functional oral and dental tissues, and research continues to advance our understanding of stem cells, signalling factors that stimulate repair and extracellular scaffold interactions for the purposes of tissue engineering. We discuss recent findings in the literature to synthesise our understanding of current and prospective approaches based on biological repair that have the potential to improve orthodontic treatment outcomes in adult patients. Methods such as mesenchymal stem cell transplantation, biomimetic scaffold manipulation, and growth factor control may be employed to overcome the challenges described above, thereby reducing adverse sequelae and improving orthodontic treatment outcomes in adult patients. The overarching goal of such research is to eventually translate these regenerative techniques into clinical practice, and establish a new gold standard of safe, effective, autologous therapies.
Collapse
|
33
|
Ghosh S, Qiao W, Yang Z, Orrego S, Neelakantan P. Engineering Dental Tissues Using Biomaterials with Piezoelectric Effect: Current Progress and Future Perspectives. J Funct Biomater 2022; 14:8. [PMID: 36662055 PMCID: PMC9867283 DOI: 10.3390/jfb14010008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Dental caries and traumatic injuries to teeth may cause irreversible inflammation and eventual death of the dental pulp. Nevertheless, predictably, repair and regeneration of the dentin-pulp complex remain a formidable challenge. In recent years, smart multifunctional materials with antimicrobial, anti-inflammatory, and pro-regenerative properties have emerged as promising approaches to meet this critical clinical need. As a unique class of smart materials, piezoelectric materials have an unprecedented advantage over other stimuli-responsive materials due to their inherent capability to generate electric charges, which have been shown to facilitate both antimicrobial action and tissue regeneration. Nonetheless, studies on piezoelectric biomaterials in the repair and regeneration of the dentin-pulp complex remain limited. In this review, we summarize the biomedical applications of piezoelectric biomaterials in dental applications and elucidate the underlying molecular mechanisms contributing to the biological effect of piezoelectricity. Moreover, we highlight how this state-of-the-art can be further exploited in the future for dental tissue engineering.
Collapse
Affiliation(s)
- Sumanta Ghosh
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Wei Qiao
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengbao Yang
- Department of Mechanical Engineering & Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, China
| | - Santiago Orrego
- Oral Health Sciences Department, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19140, USA
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA 19140, USA
| | | |
Collapse
|
34
|
Lyu H, Zhou X, Qian Y, Liu X, Gopinathan G, Pandya M, Qin C, Luan X, Diekwisch TGH. Long-acting PFI-2 small molecule release and multilayer scaffold design achieve extensive new formation of complex periodontal tissues with unprecedented fidelity. Biomaterials 2022; 290:121819. [PMID: 36209579 DOI: 10.1016/j.biomaterials.2022.121819] [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: 05/01/2022] [Revised: 08/20/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
The faithful engineering of complex human tissues such as the bone/soft tissue/mineralized tissue interface in periodontal tissues requires innovative molecular cues in conjunction with tailored scaffolds. To address the loss of periodontal bone and connective tissues following periodontal disease, we have generated a polydopamine and collagen coated electrospun PLGA-PCL (PP) scaffold enriched with the small molecule mediator PFI-2 (PP-PFI-pDA-COL-PFI). In vitro 3D studies using PDL progenitors revealed that the PP-PFI-pDA-COL-PFI scaffold substantially enhanced Alizarin Red staining, increased Ca/P ratios 4-fold, and stimulated cell proliferation more than 12-fold compared to PP-controls, suggestive of its potential for mineralized tissue engineering. When applied in our experimental periodontitis model, the PP-PFI-pDA-COL-PFI scaffold resulted in a substantial 34% reduction in alveolar bone defect height, a 25% root-length gain in periodontal attachment, and the formation of highly ordered regenerated acellular cementum twice as thick as in controls. Explaining the mechanism of PFI-2 mineralized tissue regeneration in periodontal tissues, PFI-2 inhibited SETD7-mediated β-Catenin protein methylation and increased β-Catenin nuclear localization. Together, dual-level PFI-2 incorporation into a degradable, dopamine/collagen coated PLGA/PCL scaffold backbone resulted in the regeneration of the tripartite periodontal complex with unprecedented fidelity, including periodontal attachment and new formation of mineralized tissues in inflamed periodontal environments.
Collapse
Affiliation(s)
- Huling Lyu
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Xuefeng Zhou
- UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA; State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yunzhu Qian
- UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA; Center for Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Gokul Gopinathan
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Mirali Pandya
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Chunlin Qin
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Xianghong Luan
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA; UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA
| | - Thomas G H Diekwisch
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, TX, USA; UIC College of Dentistry, Department of Oral Biology, Chicago, IL, USA.
| |
Collapse
|
35
|
Deng S, Lei T, Chen H, Zheng H, Xiao Z, Cai S, Hang Z, Xiong W, Yu Y, Zhang X, Yang Y, Bi W, Du H. Metformin pre-treatment of stem cells from human exfoliated deciduous teeth promotes migration and angiogenesis of human umbilical vein endothelial cells for tissue engineering. Cytotherapy 2022; 24:1095-1104. [PMID: 36064533 DOI: 10.1016/j.jcyt.2022.07.003] [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: 10/04/2021] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AIMS Stem cells from human exfoliated deciduous teeth (SHED) play a significant role in tissue engineering and regenerative medicine. Angiogenesis is crucial in tissue regeneration and a primary target of regenerative medicine. As a first-line anti-diabetic drug, metformin demonstrates numerous valuable impacts on stem cells. This study aimed to explore metformin's impact and mechanism of action on SHED-mediated angiogenesis. METHODS First, cell proliferation; flow cytometry; osteogenic, adipogenic and chondrogenic induction; and proteomics analyses were conducted to explore the role of metformin in SHED. Subsequently, migration and tube formation assays were used to evaluate chemotaxis and angiogenesis enhancement by SHED pre-treated with metformin under co-culture conditions in vitro, and relative messenger RNA expression levels were determined by quantitative reverse transcription polymerase chain reaction. Finally, nude mice were used for in vivo tube formation assay, and sections were analyzed through immunohistochemistry staining with anti-human CD31 antibody. RESULTS Metformin significantly promoted SHED proliferation as well as osteogenic, adipogenic and chondrogenic differentiation. Proteomics showed that metformin significantly upregulated 124 differentially abundant proteins involved in intracellular processes, including various proteins involved in cell migration and angiogenesis, such as MAPK1. The co-culture system demonstrated that SHED pre-treated with metformin significantly improved the migration and angiogenesis of human umbilical vein endothelial cells. In addition, SHED pre-treated with metformin possessed greater ability to promote angiogenesis in vivo. CONCLUSIONS In summary, the authors' findings illustrate metformin's mechanism of action on SHED and confirm that SHED pre-treated with metformin exhibits a strong capacity for promoting angiogenesis. This helps in promoting the application of dental pulp-derived stem cells pre-treated with metformin in regeneration engineering.
Collapse
Affiliation(s)
- Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China; School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Hongyu Chen
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Huiting Zheng
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Zhuangzhuang Xiao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Shanglin Cai
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Zhongci Hang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Weini Xiong
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yanqing Yu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Xiaoshuang Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Yanjie Yang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Wangyu Bi
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, China.
| |
Collapse
|
36
|
Kirilova J, Kirov D, Yovchev D, Topalova-Pirinska S, Deliverska E. Endodontic and surgical treatment of chronic apical periodontitis: a randomized clinical study. BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2022.2108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Affiliation(s)
- Janet Kirilova
- Department of Conservative Dentistry, Faculty of Dental Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Dimitar Kirov
- Department of Prosthetic Dental Medicine, Faculty of Dental Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Dimitar Yovchev
- Department of Imaging and Oral Diagnostic, Faculty of Dental Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Snezhanka Topalova-Pirinska
- Department of Conservative Dentistry, Faculty of Dental Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Elitsa Deliverska
- Department of Maxillofacial Surgery, Faculty of Dental Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
37
|
Chimerad M, Barazesh A, Zandi M, Zarkesh I, Moghaddam A, Borjian P, Chimehrad R, Asghari A, Akbarnejad Z, Khonakdar HA, Bagher Z. Tissue engineered scaffold fabrication methods for medical applications. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2101112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Mohammadreza Chimerad
- Department of Mechanical & Aerospace Engineering, College of Engineering & Computer Science, University of Central Florida, Orlando, Florida, USA
| | - Alireza Barazesh
- Tissue Engineering and Biological Systems Research Laboratory, School of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Mojgan Zandi
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Ibrahim Zarkesh
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Armaghan Moghaddam
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Pouya Borjian
- Department of Mechanical & Aerospace Engineering, College of Engineering & Computer Science, University of Central Florida, Orlando, Florida, USA
| | - Rojan Chimehrad
- Department of Biological Sciences, Islamic Azad University Tehran Medical Branch, Tehran, Iran
| | - Alimohamad Asghari
- Skull Base Research Center, School of Medicine, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Akbarnejad
- ENT and Head and Neck Research Center and Department, School of Medicine, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Ali Khonakdar
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Zohreh Bagher
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- ENT and Head and Neck Research Center and Department, School of Medicine, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Rusu LC, Ardelean LC. Advanced Materials for Oral Application. MATERIALS 2022; 15:ma15144749. [PMID: 35888216 PMCID: PMC9315927 DOI: 10.3390/ma15144749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Laura-Cristina Rusu
- Department of Oral Pathology, Multidisciplinary Center for Research, Evaluation, Diagnosis and Therapies in Oral Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Lavinia Cosmina Ardelean
- Department of Technology of Materials and Devices in Dental Medicine, Multidisciplinary Center for Research, Evaluation, Diagnosis and Therapies in Oral Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Correspondence:
| |
Collapse
|
39
|
Novel In Situ-Cross-Linked Electrospun Gelatin/Hydroxyapatite Nonwoven Scaffolds Prove Suitable for Periodontal Tissue Engineering. Pharmaceutics 2022; 14:pharmaceutics14061286. [PMID: 35745858 PMCID: PMC9230656 DOI: 10.3390/pharmaceutics14061286] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
Periodontal diseases affect millions of people worldwide and can result in tooth loss. Regenerative treatment options for clinical use are thus needed. We aimed at developing new nonwoven-based scaffolds for periodontal tissue engineering. Nonwovens of 16% gelatin/5% hydroxyapatite were produced by electrospinning and in situ glyoxal cross-linking. In a subset of scaffolds, additional porosity was incorporated via extractable polyethylene glycol fibers. Cell colonization and penetration by human mesenchymal stem cells (hMSCs), periodontal ligament fibroblasts (PDLFs), or cocultures of both were visualized by scanning electron microscopy and 4′,6-diamidin-2-phenylindole (DAPI) staining. Metabolic activity was assessed via Alamar Blue® staining. Cell type and differentiation were analyzed by immunocytochemical staining of Oct4, osteopontin, and periostin. The electrospun nonwovens were efficiently populated by both hMSCs and PDLFs, while scaffolds with additional porosity harbored significantly more cells. The metabolic activity was higher for cocultures of hMSCs and PDLFs, or for PDLF-seeded scaffolds. Periostin and osteopontin expression was more pronounced in cocultures of hMSCs and PDLFs, whereas Oct4 staining was limited to hMSCs. These novel in situ-cross-linked electrospun nonwoven scaffolds allow for efficient adhesion and survival of hMSCs and PDLFs. Coordinated expression of differentiation markers was observed, which rendered this platform an interesting candidate for periodontal tissue engineering.
Collapse
|
40
|
Advanced Biomaterials, Coatings, and Techniques: Applications in Medicine and Dentistry. COATINGS 2022. [DOI: 10.3390/coatings12060797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The field of biomaterials is very extensive, encompassing both the materials themselves and the manufacturing methods, which are constantly developing [...]
Collapse
|
41
|
A Molecular View on Biomaterials and Dental Stem Cells Interactions: Literature Review. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biomaterials and stem cells are essential components in the field of regenerative medicine. Various biomaterials have been designed that have appropriate biochemical and biophysical characteristics to mimic the microenvironment of an extracellular matrix. Dental stem cells (DT-MSCs) represent a novel source for the development of autologous therapies due to their easy availability. Although research on biomaterials and DT-MSCs has progressed, there are still challenges in the characteristics of biomaterials and the molecular mechanisms involved in regulating the behavior of DT-MSCs. In this review, the characteristics of biomaterials are summarized, and their classification according to their source, bioactivity, and different biological effects on the expansion and differentiation of DT-MSCs is summarized. Finally, advances in research on the interaction of biomaterials and the molecular components involved (mechanosensors and mechanotransduction) in DT-MSCs during their proliferation and differentiation are analyzed. Understanding the molecular dynamics of DT-MSCs and biomaterials can contribute to research in regenerative medicine and the development of autologous stem cell therapies.
Collapse
|
42
|
Liu H, Lu J, Jiang Q, Haapasalo M, Qian J, Tay FR, Shen Y. Biomaterial scaffolds for clinical procedures in endodontic regeneration. Bioact Mater 2022; 12:257-277. [PMID: 35310382 PMCID: PMC8897058 DOI: 10.1016/j.bioactmat.2021.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022] Open
Abstract
Regenerative endodontic procedures have been rapidly evolving over the past two decades and are employed extensively in clinical endodontics. These procedures have been perceived as valuable adjuvants to conventional strategies in the treatment of necrotic immature permanent teeth that were deemed to have poor prognosis. As a component biological triad of tissue engineering (i.e., stem cells, growth factors and scaffolds), biomaterial scaffolds have demonstrated clinical potential as an armamentarium in regenerative endodontic procedures and achieved remarkable advancements. The aim of the present review is to provide a broad overview of biomaterials employed for scaffolding in regenerative endodontics. The favorable properties and limitations of biomaterials organized in naturally derived, host-derived and synthetic material categories were discussed. Preclinical and clinical studies published over the past five years on the performance of biomaterial scaffolds, as well as current challenges and future perspectives for the application of biomaterials for scaffolding and clinical evaluation of biomaterial scaffolds in regenerative endodontic procedures were addressed in depth. Overview of biomaterials for scaffolding in regenerative endodontics are presented. Findings of preclinical and clinical studies on the performance of biomaterial scaffolds are summarized. Challenges and future prospects in biomaterial scaffolds are discussed.
Collapse
|
43
|
Thomas SM, Ackert-Bicknell CL, Zuscik MJ, Payne KA. Understanding the Transcriptomic Landscape to Drive New Innovations in Musculoskeletal Regenerative Medicine. Curr Osteoporos Rep 2022; 20:141-152. [PMID: 35156183 DOI: 10.1007/s11914-022-00726-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW RNA-sequencing (RNA-seq) is a novel and highly sought-after tool in the field of musculoskeletal regenerative medicine. The technology is being used to better understand pathological processes, as well as elucidate mechanisms governing development and regeneration. It has allowed in-depth characterization of stem cell populations and discovery of molecular mechanisms that regulate stem cell development, maintenance, and differentiation in a way that was not possible with previous technology. This review introduces RNA-seq technology and how it has paved the way for advances in musculoskeletal regenerative medicine. RECENT FINDINGS Recent studies in regenerative medicine have utilized RNA-seq to decipher mechanisms of pathophysiology and identify novel targets for regenerative medicine. The technology has also advanced stem cell biology through in-depth characterization of stem cells, identifying differentiation trajectories and optimizing cell culture conditions. It has also provided new knowledge that has led to improved growth factor use and scaffold design for musculoskeletal regenerative medicine. This article reviews recent studies utilizing RNA-seq in the field of musculoskeletal regenerative medicine. It demonstrates how transcriptomic analysis can be used to provide insights that can aid in formulating a regenerative strategy.
Collapse
Affiliation(s)
- Stacey M Thomas
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Mail Stop 8343, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Cheryl L Ackert-Bicknell
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Mail Stop 8343, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Michael J Zuscik
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Mail Stop 8343, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Karin A Payne
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Mail Stop 8343, 12800 East 19th Avenue, Aurora, CO, 80045, USA.
- Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
44
|
Zhang P, Zuo D, Mou S, Zhong Y, Yuan X, Zeng J. Effect of inward rectifier potassium 2.1 channel on the osteogenic differentiation of human dental follicle cells and its mechanism. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2022; 40:139-147. [PMID: 38597045 PMCID: PMC9002191 DOI: 10.7518/hxkq.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 01/30/2022] [Indexed: 01/24/2023]
Abstract
OBJECTIVES This study aims to explore the effect of inward rectifier potassium (Kir) 2.1 channel on the osteogenic differentiation of human dental follicle cells (hDFCs) and its mechanism. METHODS hDFCs were isolated and cultured, and their source was verified by flow cytometry. Osteogenic differentiation ability of hDFCs was evaluated by osteogenic induction. Reverse-transcription polymerase chain reaction (RT-PCR) was performed to detect the gene expression of Kir2.1 gene (KCNJ2) in hDFCs. Real-time quantitative PCR (RT-qPCR) was performed to detect the expression of the Kir2.1 gene (KCNJ2) in hDFCs before and after osteogenic induction. Patch clamp technique was conducted to record the membrane potential changes of hDFCs before and after osteogenic induction. Moreover, the effect on the osteogenic differentiation of hDFCs was confirmed by increasing the concentration of extracellular potassium ions (50 mmol·L-1). Kir2.1 channel blockers cesium chloride (CsCl) and C19H20CINO (ML133) were applied to determine the effect of the Kir2.1 potassium channel on the osteogenic differentiation of hDFCs. At the same time, RT-qPCR was used to observe the expression changes of osteogenic differentiation related genes Runx related transcription factor 2 (Runx2) and osteocalcin (OCN) before and after the two intervention measures. Calcium imaging was performed to observe the effect of membrane potential hyperpolarization caused by decreased extracellular potassium level (2 mmol·L-1) on intracellular calcium concentration. RESULTS RT-PCR results showed that hDFCs expressed the Kir2.1 channel gene (KCNJ2). The RT-qPCR results showed that the KCNJ2 gene expression in hDFCs was upregulated 7 days after osteogenic induction. The patch clamp results showed that the membrane potential of hDFCs hyperpolarized to (-47±5.2) mV from (-12±3.2) mV. Alizarin red and alkaline phosphatase staining results showed that increasing the concentration of the extracellular potassium or blocking the function of the Kir2.1 channel significantly inhibited the osteogenic mineralization ability of hDFCs. The membrane potential hyperpolarization increased the intracellular calcium concentration in hDFCs. CONCLUSIONS Membrane potential hyperpolarization mediated by the Kir2.1 channel plays an important role in the osteogenic differentiation of hDFCs.
Collapse
Affiliation(s)
- Peng Zhang
- Dept. of Orthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
- Oral and Facial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou 646000, China
| | - Dongchuan Zuo
- Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology of Ministry of Education, Southwest Medical University, Luzhou 646000, China
| | - Siyu Mou
- Oral and Facial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou 646000, China
| | - Yutong Zhong
- Oral and Facial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou 646000, China
| | - Xiaoping Yuan
- Dept. of Orthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
- Oral and Facial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou 646000, China
| | - Jin Zeng
- Dept. of Orthodontics, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
- Oral and Facial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
45
|
Peters K, Wiesmann N, Heimes D, Schwab R, Kämmerer PW, Al-Nawas B, Unger RE, Hasenburg A, Brenner W. Extracorporeal Shock Wave Therapy Improves In Vitro Formation of Multilayered Epithelium of Oral Mucosa Equivalents. Biomedicines 2022; 10:biomedicines10030700. [PMID: 35327502 PMCID: PMC8945876 DOI: 10.3390/biomedicines10030700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Oral mucosa is used in various surgical fields as a graft for the reconstruction of tissue defects. Tissue engineering of oral mucosa equivalents using autologous cells represents a suitable less burdensome alternative. The survival of the multilayered epithelium is essential for the functionality of the tissues in vivo. To ensure its functionality after transplantation, mucosa equivalents in vitro were subjected to extracorporeal shock wave therapy (ESWT) to determine whether this treatment stimulated the formation and differentiation of the epithelium. Mucosa equivalents treated with ESWT were examined for cellular metabolic activity using AlamarBlueTM assay. The formation of vascular structures, basement membrane, and multilayered epithelium were examined using confocal fluorescence microscopy and immunohistochemistry. The potential ingrowth in vivo was simulated using the chorioallantoic membrane model (CAM assay) in ovo. ESWT on culture day 19 of oral mucosa equivalents resulted in slightly increased cellular metabolic activity. The in vitro development of basement membrane and multilayer epithelium was stimulated by ESWT. Additionally, in the CAM assay, ESWT led to a more pronounced multilayered epithelium. Thus, ESWT stimulated the formation of a more distinct and differentiated multilayered epithelium of oral mucosa equivalents in vitro and might increase the chance of efficient ingrowth, survival, and functionality of tissue equivalents in vivo.
Collapse
Affiliation(s)
- Katharina Peters
- Department of Obstetrics and Gynecology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.P.); (R.S.); (A.H.)
| | - Nadine Wiesmann
- Department of Oral and Maxillofacial and Plastic Surgery, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (N.W.); (D.H.); (P.W.K.); (B.A.-N.)
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Diana Heimes
- Department of Oral and Maxillofacial and Plastic Surgery, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (N.W.); (D.H.); (P.W.K.); (B.A.-N.)
| | - Roxana Schwab
- Department of Obstetrics and Gynecology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.P.); (R.S.); (A.H.)
| | - Peer W. Kämmerer
- Department of Oral and Maxillofacial and Plastic Surgery, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (N.W.); (D.H.); (P.W.K.); (B.A.-N.)
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial and Plastic Surgery, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (N.W.); (D.H.); (P.W.K.); (B.A.-N.)
| | - Ronald E. Unger
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Annette Hasenburg
- Department of Obstetrics and Gynecology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.P.); (R.S.); (A.H.)
| | - Walburgis Brenner
- Department of Obstetrics and Gynecology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.P.); (R.S.); (A.H.)
- Correspondence: ; Tel.: +49-6131-17-2740
| |
Collapse
|
46
|
Standardized Human Platelet Lysates as Adequate Substitute to Fetal Calf Serum in Endothelial Cell Culture for Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3807314. [PMID: 35281595 PMCID: PMC8913112 DOI: 10.1155/2022/3807314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/06/2021] [Indexed: 11/17/2022]
Abstract
Fetal calf serum (FCS) is used for in vitro cell culture, as it provides the cells with various growth-promoting compounds. For applications in humans, FCS does not meet the required safety standards and should be replaced by an appropriate substitute. This study analyzed the suitability of using human platelet lysate (hPL) as a substitute for FCS in endothelial cell cultures for in vitro and in vivo tissue engineering applications. The focus was placed on standardized, commercially available hPLs (MultiPL'30, MultiPL'100), which are approved for applications in humans, and compared to laboratory-prepared hPLs (lp-hLP). Human umbilical vein endothelial cells (HUVEC) were cultured with FCS or with different hPLs. Cell morphology, proliferation, viability, apoptosis, and necrosis, as well as the organization of vascular structures, were assessed. No morphological changes were noticed when FCS was replaced by standardized hPLs in concentrations of 1-10%. In contrast, the use of lp-hLPs led to irregular cell shape and increased vacuolization of the cytoplasm. HUVEC proliferation and viability were not compromised by using media supplemented with standardized hPLs or pl-hPLs in concentrations of 1-10%, compared to cells grown in media supplemented with 20% FCS. The apoptosis rate using lp-hPLs was higher compared to the use of standardized hPLs. The necrosis rate tended to be lower when FCS was replaced by hPLs. HUVEC formed more pronounced capillary-like structures when the media were supplemented with hPLs instead of supplementation with FCS. Thus, compared to the use of FCS, the use of hPLs was beneficial for the growth and optimal expression of functional endothelial cell characteristics during in vitro experiments. Commercially available hPLs proved to be particularly suitable, as they led to reproducible results during in vitro experiments, while meeting the safety requirements for in vivo use.
Collapse
|
47
|
Duncan WJ, Coates DE. Meeting the challenges and clinical requirements for dental regeneration; the New Zealand experience. Bone 2022; 154:116181. [PMID: 34509689 DOI: 10.1016/j.bone.2021.116181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 11/02/2022]
Abstract
Disease and trauma leading to tooth loss and destruction of supporting bone is a significant oral handicap, which may be addressed through surgical therapies that aim to regenerate the lost tissue. Whilst complete regeneration of teeth is still aspirational, regeneration of supporting structures (dental pulp, cementum, periodontal ligament, bone) is becoming commonplace, both for teeth and for titanium dental implants that are used to replace teeth. Most grafting materials are essentially passive, however the next generation of oral regenerative devices will combine non-antibiotic antimicrobials and/or osteogenic or inductive factors and/or appropriate multipotential stem cells. The review gives an overview of the approaches taken, including fabrication of novel scaffolds, incorporation of growth factors and cell-based therapies, and discusses the preclinical animal models we employ in the development pathway.
Collapse
Affiliation(s)
- Warwick J Duncan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand.
| | - Dawn E Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
48
|
Abstract
Tissue engineering or tissue reconstruction/repair/regeneration may be considered as a guiding strategy in oral and maxillofacial surgery, as well as in endodontics, orthodontics, periodontics, and daily clinical practice. A wide range of techniques has been developed over the past years, from tissue grafts to the more recent and innovative regenerative procedures. Continuous research in the field of natural and artificial materials and biomaterials, as well as in advanced scaffold design strategies has been carried out. The focus has also been on various growth factors involved in dental tissue repair or reconstruction. Benefiting from the recent literature, this review paper illustrates current innovative strategies and technological approaches in oral and maxillofacial tissue engineering, trying to offer some information regarding the available scientific data and practical applications. After introducing tissue engineering aspects, an overview on additive manufacturing technologies will be provided, with a focus on the applications of superparamagnetic iron oxide nanoparticles in the biomedical field. The potential applications of magnetic fields and magnetic devices on the acceleration of orthodontic tooth movement will be analysed.
Collapse
|
49
|
Dziuba R, Kucharska M, Madej-Kiełbik L, Sulak K, Wiśniewska-Wrona M. Biopolymers and Biomaterials for Special Applications within the Context of the Circular Economy. MATERIALS (BASEL, SWITZERLAND) 2021; 14:7704. [PMID: 34947300 PMCID: PMC8708369 DOI: 10.3390/ma14247704] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022]
Abstract
The main challenge of the economy is counteracting the adverse effects of progressive industrialisation on the environment around the world. Economic development that accompanies this trend correlates to production increase in not only consumer articles but also special application articles that are difficult to remanufacture, such as medical supplies. For many researchers, discovering innovative materials for special applications that could become an essential element of circular economy production is important. Measures to reduce the production of industrial materials whose waste is difficult to recycle are more and more apparent to manufacturers, especially when faced with the new financial situation in European Union, as one of its priorities is to implement the principles of circular economy. The purpose of the article is to analyse the current state of research on special-application biomaterials within the context of the circular economy. Empirical analysis is conducted for Poland compared to the rest of the European Union (EU) within the time-frame of 2014-2020, which is the most recent financial timeframe of the EU. The submitted studies are based on secondary data obtained mainly from European databases, as well as primary data resulting from the research works at Łukasiewicz Research Network-Institute of Biopolymers and Chemical Fibres.
Collapse
Affiliation(s)
- Radosław Dziuba
- Department of World Economy and European Integration, University of Lodz, 41/43 Rewolucji 1905 Str., 90-214 Lodz, Poland;
| | - Magdalena Kucharska
- Łukasiewicz Research Network—Institute of Biopolymers and Chemical Fibres, 19/27 M. Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (M.K.); (L.M.-K.); (K.S.)
| | - Longina Madej-Kiełbik
- Łukasiewicz Research Network—Institute of Biopolymers and Chemical Fibres, 19/27 M. Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (M.K.); (L.M.-K.); (K.S.)
| | - Konrad Sulak
- Łukasiewicz Research Network—Institute of Biopolymers and Chemical Fibres, 19/27 M. Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (M.K.); (L.M.-K.); (K.S.)
| | - Maria Wiśniewska-Wrona
- Łukasiewicz Research Network—Institute of Biopolymers and Chemical Fibres, 19/27 M. Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (M.K.); (L.M.-K.); (K.S.)
| |
Collapse
|
50
|
Xu X, Xiao L, Xu Y, Zhuo J, Yang X, Li L, Xiao N, Tao J, Zhong Q, Li Y, Chen Y, Du Z, Luo K. Vascularized bone regeneration accelerated by 3D-printed nanosilicate-functionalized polycaprolactone scaffold. Regen Biomater 2021; 8:rbab061. [PMID: 34858634 PMCID: PMC8633727 DOI: 10.1093/rb/rbab061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/09/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Critical oral-maxillofacial bone defects, damaged by trauma and tumors, not only affect the physiological functions and mental health of patients but are also highly challenging to reconstruct. Personalized biomaterials customized by 3D printing technology have the potential to match oral-maxillofacial bone repair and regeneration requirements. Laponite (LAP) nanosilicates have been added to biomaterials to achieve biofunctional modification owing to their excellent biocompatibility and bioactivity. Herein, porous nanosilicate-functionalized polycaprolactone (PCL/LAP) was fabricated by 3D printing technology, and its bioactivities in bone regeneration were investigated in vitro and in vivo. In vitro experiments demonstrated that PCL/LAP exhibited good cytocompatibility and enhanced the viability of bone marrow mesenchymal stem cells (BMSCs). PCL/LAP functioned to stimulate osteogenic differentiation of BMSCs at the mRNA and protein levels and elevated angiogenic gene expression and cytokine secretion. Moreover, BMSCs cultured on PCL/LAP promoted the angiogenesis potential of endothelial cells by angiogenic cytokine secretion. Then, PCL/LAP scaffolds were implanted into the calvarial defect model. Toxicological safety of PCL/LAP was confirmed, and significant enhancement of vascularized bone formation was observed. Taken together, 3D-printed PCL/LAP scaffolds with brilliant osteogenesis to enhance bone regeneration could be envisaged as an outstanding bone substitute for a promising change in oral-maxillofacial bone defect reconstruction.
Collapse
Affiliation(s)
- Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Long Xiao
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Jin Zhuo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Xue Yang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Li Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Nianqi Xiao
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Jing Tao
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Quan Zhong
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Yanfen Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yuling Chen
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Zhibin Du
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| |
Collapse
|