1
|
Li T, Shu M, Zhu C, Liu Q, Li Y, Wang R, Chen L, Shi W, Sun Z, Hou Z, Fang B, Xia L. Triple-Combination Therapy with a Multifunctional Yolk-Shell Nanozyme Au@CeO 2 Loaded with Dimethyl Fumarate for Periodontitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413891. [PMID: 39716921 PMCID: PMC11831482 DOI: 10.1002/advs.202413891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/03/2024] [Indexed: 12/25/2024]
Abstract
Periodontitis, a chronic inflammatory disease, is the leading cause of tooth loss in adults and is one of the most prevalent and complex oral conditions. Oxidative stress induced by the excessive generation of reactive oxygen species (ROS) leads to periodontitis, which is closely associated with pathological processes, including mitochondrial dysfunction of periodontal cells and local immune dysregulation. However, current treatment modalities that target single pathological processes have limited long-term therapeutic effects. Herein, a multifunctional Yolk-Shell nanozyme, Au@CeO2-dimethyl fumarate (DMF), which comprehensively addresses the oxidative stress-induced pathophysiological processes of periodontitis through antioxidant activity, mitochondrial maintenance, and immune modulation mechanisms, is described. For material design logic, functionally complementary Au and CeO2 formed an excellent photothermally regulated high-efficiency nanozyme, which also provided an ideal drug carrier for DMF. As for the therapeutic logic, Au@CeO2-DMF restores mitochondrial dysfunction and immune dysregulation, which also contributes to endogenous ROS elimination, thereby achieving long-term stable therapeutic effects. In a rat model, local Au@CeO2-DMF photothermal therapy effectively alleviated ROS-induced tissue damage and restored periodontal homeostasis. Altogether, this study presents a novel antioxidant nanozyme for managing alveolar bone loss under prolonged oxidative stress and demonstrates the importance of comprehensive intervention in key pathological processes in periodontitis treatment design.
Collapse
Affiliation(s)
- Tiancheng Li
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Mengmeng Shu
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Cheng Zhu
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Qicheng Liu
- School of Food Science and Pharmaceutical EngineeringNanjing Normal UniversityNanjing210023China
| | - Yixin Li
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Ruike Wang
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Lihan Chen
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Wenxiao Shi
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Zhaoxuan Sun
- ZhuHai Campus of Zunyi Medical UniversityZhuhai519041China
| | - Zhiyao Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Bing Fang
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Lunguo Xia
- Department of OrthodonticsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| |
Collapse
|
2
|
Ma Y, Wang X, Huang X, He Y, Su T, Niu X, Gao J, Lu F, Chang Q. Radial Egg White Hydrogel Releasing Extracellular Vesicles for Cell Fate Guidance and Accelerated Diabetic Skin Regeneration. Adv Healthc Mater 2024; 13:e2400016. [PMID: 39285803 DOI: 10.1002/adhm.202400016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 08/01/2024] [Indexed: 12/18/2024]
Abstract
Topology and bioactive molecules are crucial for stimulating cellular and tissue functions. To regulate the chronic wound microenvironment, mono-assembly technology is employed to fabricate a radial egg white hydrogel loaded with lyophilized adipose tissue-extracellular vesicles (radial EWH@L-EVs). The radial architecture not only significantly modified the gene expression of functional cells, but also achieved directional and controlled release kinetics of L-EVs. Through the synergy of topographical and inherent bioactive cues, radial EWH@L-EVs effectively reduced intracellular oxidative stress and promoted the polarization of macrophages toward an anti-inflammatory phenotype during the inflammatory phase. Afterward, radial EWH@L-EVs facilitated the centripetal migration and proliferation of fibroblasts and endothelial cells as the wound transitioned to the proliferative phase. During the latter remodeling phase, radial EWH@L-EVs accelerated the regeneration of granulation tissue, angiogenesis, and collagen deposition, thereby promoting the reorganization chronic wound. Compared with the gold standard collagen scaffold, radial EWH@L-EVs actively accommodated the microenvironment via various functions throughout all stages of diabetic wound healing. This can be attributed to the orientation of topological structures and bioactive molecules, which should be considered of utmost importance in tissue engineering.
Collapse
Affiliation(s)
- Yuan Ma
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Xinhui Wang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Xiaoqi Huang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Yu He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Ting Su
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Xingtang Niu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
3
|
de Souza Araújo I, Perkins RS, Ibrahim MM, Huang GTJ, Zhang W. Bioprinting PDLSC-Laden Collagen Scaffolds for Periodontal Ligament Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59979-59990. [PMID: 39467547 PMCID: PMC11551894 DOI: 10.1021/acsami.4c13830] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024]
Abstract
Periodontitis and severe trauma are major causes of damage to the periodontal ligament (PDL). Repairing the native conditions of the PDL is essential for the stability of the tissue and its interfaces. Bioprinting periodontal ligament stem cells (PDLSCs) is an interesting approach to guide the regeneration of PDL and interfacial integration. Herein, a collagen-based bioink mimicking the native extracellular matrix conditions and carrying PDLSCs was tested to guide the periodontal ligament organization. The bioink was tested at two different concentrations (10 and 15 mg/mL) and characterized by swelling and degradation, microstructural organization, and rheological properties. The biological properties were assessed after loading PDLSCs into bioinks for bioprinting. The characterization was performed through cell viability, alizarin red assay, and expression for ALP, COL1A1, RUNX2, and OCN. The in vivo biocompatibility of the PDLSC-laden bioinks was verified using subcutaneous implantation in mice. Later, the ability of the bioprinted PDLSC-laden bioinks on dental root fragments to form PDL was also investigated in vivo in mice for 4 and 10 weeks. The bioinks demonstrated typical shear-thinning behavior, a porous microstructure, and stable swelling and degradation characteristics. Both concentrations were printable and provided suitable conditions for a high cell survival, proliferation, and differentiation. PDLSC-laden bioinks demonstrated biocompatibility in vivo, and the bioprinted scaffolds on the root surface evidenced PDLSC alignment, organization, and PDLSC migration to the root surface. The versatility of collagen-based bioinks provides native ECM conditions for PDLSC proliferation, alignment, organization, and differentiation, with translational applications in bioprinting scaffolds for PDL regeneration.
Collapse
Affiliation(s)
- Isaac
J. de Souza Araújo
- Department
of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Rachel S. Perkins
- Department
of Orthopaedic Surgery and Biomedical Engineering, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Mohamed Moustafa Ibrahim
- Department
of Ophthalmology, Hamilton Eye Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Pharmaceutics, Faculty of Pharmacy, Mansoura
University, Mansoura 35516, Egypt
| | - George T.-J. Huang
- Department
of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Physiology, College of Medicine, University
of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Endodontics, The University of Tennessee
Health Science Center, Memphis, Tennessee 38163, United States
| | - Wenjing Zhang
- Department
of Genetics, Genomics & Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
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
|
Wang L, Liu Y, Lin Z, Chen H, Liu B, Yan X, Zhu T, Zhang Q, Zhao J. Durable immunomodulatory hierarchical patch for rotator cuff repairing. Bioact Mater 2024; 37:477-492. [PMID: 38698919 PMCID: PMC11063994 DOI: 10.1016/j.bioactmat.2024.03.029] [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: 12/24/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Degradable rotator cuff patches, followed over five years, have been observed to exhibit high re-tear rates exceeding 50%, which is attributed to the inability of degradable polymers alone to restore the post-rotator cuff tear (RCT) inflammatory niche. Herein, poly(ester-ferulic acid-urethane)urea (PEFUU) was developed, featuring prolonged anti-inflammatory functionality, achieved by the integration of ferulic acid (FA) into the polyurethane repeating units. PEFUU stably releases FA in vitro, reversing the inflammatory niche produced by M1 macrophages and restoring the directed differentiation of stem cells. Utilizing PEFUU, hierarchical composite nanofiber patch (HCNP) was fabricated, simulating the natural microstructure of the tendon-to-bone interface with an aligned-random alignment. The incorporation of enzymatic hydrolysate derived from decellularized Wharton jelly tissue into the random layer could further enhance cartilage regeneration at the tendon-to-bone interface. Via rat RCT repairing model, HCNP possessing prolonged anti-inflammatory properties uniquely facilitated physiological healing at the tendon-to-bone interface's microstructure. The alignment of fibers was restored, and histologically, the characteristic tripartite distribution of collagen I - collagen II - collagen I was achieved. This study offers a universal approach to the functionalization of degradable polymers and provides a foundational reference for their future applications in promoting the in vivo regeneration of musculoskeletal tissues.
Collapse
Affiliation(s)
- Liren Wang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Yonghang Liu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, China
| | - Zhiqi Lin
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Huiang Chen
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Bowen Liu
- Bioarticure Medical Technology (Shanghai) Co., Ltd, Shanghai, China
| | - Xiaoyu Yan
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, 99 Shangda Rd., Shanghai, 200444, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
6
|
Peng S, Fu H, Li R, Li H, Wang S, Li B, Sun J. A new direction in periodontitis treatment: biomaterial-mediated macrophage immunotherapy. J Nanobiotechnology 2024; 22:359. [PMID: 38907216 PMCID: PMC11193307 DOI: 10.1186/s12951-024-02592-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/28/2024] [Indexed: 06/23/2024] Open
Abstract
Periodontitis is a chronic inflammation caused by a bacterial infection and is intimately associated with an overactive immune response. Biomaterials are being utilized more frequently in periodontal therapy due to their designability and unique drug delivery system. However, local and systemic immune response reactions driven by the implantation of biomaterials could result in inflammation, tissue damage, and fibrosis, which could end up with the failure of the implantation. Therefore, immunological adjustment of biomaterials through precise design can reduce the host reaction while eliminating the periodontal tissue's long-term chronic inflammation response. It is important to note that macrophages are an active immune system component that can participate in the progression of periodontal disease through intricate polarization mechanisms. And modulating macrophage polarization by designing biomaterials has emerged as a new periodontal therapy technique. In this review, we discuss the role of macrophages in periodontitis and typical strategies for polarizing macrophages with biomaterials. Subsequently, we discuss the challenges and potential opportunities of using biomaterials to manipulate periodontal macrophages to facilitate periodontal regeneration.
Collapse
Affiliation(s)
- Shumin Peng
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Haojie Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Rui Li
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
| | - Hui Li
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100069, China
| | - Shuyuan Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Bingyan Li
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Jingjing Sun
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China.
| |
Collapse
|
7
|
Sun H, Luan J, Dong S. Hydrogels promote periodontal regeneration. Front Bioeng Biotechnol 2024; 12:1411494. [PMID: 38827033 PMCID: PMC11140061 DOI: 10.3389/fbioe.2024.1411494] [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: 04/03/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
Periodontal defects involve the damage and loss of periodontal tissue, primarily caused by periodontitis. This inflammatory disease, resulting from various factors, can lead to irreversible harm to the tissues supporting the teeth if not treated effectively, potentially resulting in tooth loss or loosening. Such outcomes significantly impact a patient's facial appearance and their ability to eat and speak. Current clinical treatments for periodontitis, including surgery, root planing, and various types of curettage, as well as local antibiotic injections, aim to mitigate symptoms and halt disease progression. However, these methods fall short of fully restoring the original structure and functionality of the affected tissue, due to the complex and deep structure of periodontal pockets and the intricate nature of the supporting tissue. To overcome these limitations, numerous biomaterials have been explored for periodontal tissue regeneration, with hydrogels being particularly noteworthy. Hydrogels are favored in research for their exceptional absorption capacity, biodegradability, and tunable mechanical properties. They have shown promise as barrier membranes, scaffolds, carriers for cell transplantation and drug delivery systems in periodontal regeneration therapy. The review concludes by discussing the ongoing challenges and future prospects for hydrogel applications in periodontal treatment.
Collapse
Affiliation(s)
- Huiying Sun
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Jiayi Luan
- Foshan Stomatology Hospital and School of Medicine, Foshan, Guangdong, China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
8
|
Staples R, Ivanovski S, Vaswani K, Vaquette C. Melt electrowriting scaffolds with fibre-guiding features for periodontal attachment. Acta Biomater 2024; 180:337-357. [PMID: 38583749 DOI: 10.1016/j.actbio.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
Periodontal regeneration requires the re-attachment of oblique and perpendicular periodontal ligament (PDL) fibres to newly formed cementum and alveolar bone, which has proven elusive with existing approaches. In this study, multiple fibre-guiding biphasic tissue engineered constructs were fabricated by melt electrowriting. The biphasic scaffolds were 95 % porous and consisted of a pore size gradient bone compartment and periodontal compartment made of fibre-guiding channels with micro-architectural features ranging from 100 to 60 µm aimed to direct PDL fibre alignment and attachment. In vitro evaluations over 3 and 7 days demonstrated a marked improvement in collagen fibre orientation (over 60 % fully aligned) for scaffolds with micro-architecture ≤100 µm. The biphasic scaffolds were placed on a dentine slice and implanted ectopically, and this demonstrated that all micro-channels groups facilitated oblique and perpendicular alignment and attachment on the dentine with a mean nuclei angle and mean collagen fibre angle of approximately 60° resembling the native periodontal ligament attachment. A further in vivo testing using a surgically created rodent periodontal model highlighted the 80 µm micro-channel group's effectiveness, showing a significant increase in oblique PDL fibre attachment (72 %) and periodontal regeneration (56 %) when compared to all other groups onto the tooth root compared to control groups. Further to this, immunohistochemistry demonstrated the presence of periostin in the newly formed ligament indicating that functional regeneration occurred These findings suggest that scaffold micro-architectures of 100 µm or below can play a crucial role in directing periodontal tissue regeneration, potentially addressing a critical gap in periodontal therapy. STATEMENT OF SIGNIFICANCE: Periodontal regeneration remains a significant clinical challenge. Essential to restoring dental health and function is the proper attachment of the periodontal ligament, which is functionally oriented, to regenerated bone and cementum. Our research presents an innovative biphasic scaffold, utilizing Melt Electrowriting to systematically guide tissue growth. Distinct from existing methods, our scaffold is highly porous, adaptable, and precisely guides periodontal ligament fibre attachment to the opposing tooth root and alveolar bone interfaces, a critical step for achieving periodontal functional regeneration. Our findings not only bridge a significant gap in biomaterial driven tissue guidance but also promise more predictable outcomes for patients, marking a transformative advancement in the field.
Collapse
Affiliation(s)
- Reuben Staples
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Sašo Ivanovski
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Kanchan Vaswani
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Cedryck Vaquette
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia.
| |
Collapse
|
9
|
Ming P, Liu Y, Yu P, Jiang X, Yuan L, Cai S, Rao P, Cai R, Lan X, Tao G, Xiao J. A Biomimetic Se-nHA/PC Composite Microsphere with Synergistic Immunomodulatory and Osteogenic Ability to Activate Bone Regeneration in Periodontitis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305490. [PMID: 37852940 DOI: 10.1002/smll.202305490] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/21/2023] [Indexed: 10/20/2023]
Abstract
Accumulation of reactive oxygen species (ROS) in periodontitis exacerbates the destruction of alveolar bone. Therefore, scavenging ROS to reshape the periodontal microenvironment, alleviate the inflammatory response and promote endogenous stem cell osteogenic differentiation may be an effective strategy for treating bone resorption in periodontitis. In this study, sericin-hydroxyapatite nanoparticles (Se-nHA NPs) are synthesized using a biomimetic mineralization method. Se-nHA NPs and proanthocyanidins (PC) are then encapsulated in sericin/sodium alginate (Se/SA) using an electrostatic injection technique to prepare Se-nHA/PC microspheres. Microspheres are effective in scavenging ROS, inhibiting the polarization of macrophages toward the M1 type, and inducing the polarization of macrophages toward the M2 type. In normal or macrophage-conditioned media, the Se-nHA/PC microspheres effectively promoted the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs). Furthermore, the Se-nHA/PC microspheres demonstrated anti-inflammatory effects in a periodontitis rat model by scavenging ROS and suppressing pro-inflammatory cytokines. The Se-nHA/PC microspheres are also distinguished by their capacity to decrease alveolar bone loss, reduce osteoclast activity, and boost osteogenic factor expression. Therefore, the biomimetic Se-nHA/PC composite microspheres have efficient ROS-scavenging, anti-inflammatory, and osteogenic abilities and can be used as a multifunctional filling material for inflammatory periodontal tissue regeneration.
Collapse
Affiliation(s)
- Piaoye Ming
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Yunfei Liu
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Peiyang Yu
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Xueyu Jiang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Linlin Yuan
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Shuyu Cai
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Pengcheng Rao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Rui Cai
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Gang Tao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Jingang Xiao
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
10
|
Zhao F, Zhang Z, Guo W. The 3-dimensional printing for dental tissue regeneration: the state of the art and future challenges. Front Bioeng Biotechnol 2024; 12:1356580. [PMID: 38456006 PMCID: PMC10917914 DOI: 10.3389/fbioe.2024.1356580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Tooth loss or damage poses great threaten to oral and general health. While contemporary clinical treatments have enabled tooth restoration to a certain extent, achieving functional tooth regeneration remains a challenging task due to the intricate and hierarchically organized architecture of teeth. The past few decades have seen a rapid development of three-dimensional (3D) printing technology, which has provided new breakthroughs in the field of tissue engineering and regenerative dentistry. This review outlined the bioactive materials and stem/progenitor cells used in dental regeneration, summarized recent advancements in the application of 3D printing technology for tooth and tooth-supporting tissue regeneration, including dental pulp, dentin, periodontal ligament, alveolar bone and so on. It also discussed current obstacles and potential future directions, aiming to inspire innovative ideas and encourage further development in regenerative medicine.
Collapse
Affiliation(s)
- Fengxiao Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhijun Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, China
- Yunnan Key Laboratory of Stomatology, The Affiliated Hospital of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
| |
Collapse
|
11
|
Shi Y, Tao W, Yang W, Wang L, Qiu Z, Qu X, Dang J, He J, Fan H. Calcium phosphate coating enhances osteointegration of melt electrowritten scaffold by regulating macrophage polarization. J Nanobiotechnology 2024; 22:47. [PMID: 38297240 PMCID: PMC10829397 DOI: 10.1186/s12951-024-02310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Abstract
The osteoimmune microenvironment induced by implants plays a significant role in bone regeneration. It is essential to efficiently and timely switch the macrophage phenotype from M1 to M2 for optimal bone healing. This study examined the impact of a calcium phosphate (CaP) coating on the physiochemical properties of highly ordered polycaprolactone (PCL) scaffolds fabricated using melt electrowritten (MEW). Additionally, it investigated the influence of these scaffolds on macrophage polarization and their immunomodulation on osteogenesis. The results revealed that the CaP coated PCL scaffold exhibited a rougher surface topography and higher hydrophilicity in comparison to the PCL scaffold without coating. Besides, the surface morphology of the coating and the release of Ca2+ from the CaP coating were crucial in regulating the transition of macrophages from M1 to M2 phenotypes. They might activate the PI3K/AKT and cAMP-PKA pathways, respectively, to facilitate M2 polarization. In addition, the osteoimmune microenvironment induced by CaP coated PCL could not only enhance the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) in vitro but also promote the bone regeneration in vivo. Taken together, the CaP coating can be employed to control the phenotypic switching of macrophages, thereby creating a beneficial immunomodulatory microenvironment that promotes bone regeneration.
Collapse
Affiliation(s)
- Yubo Shi
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weidong Tao
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenjing Yang
- Xijing 986 Hospital Department, The Fourth Military Medical University, Xi'an, China
| | - Lei Wang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
12
|
Wang J, Zhang H, Wang Y, Liu X, Zhu W, Jiang F, Li S, Liu L. AuNP-Loaded Electrospinning Membrane Cooperated with CDs for Periodontal Tissue Engineering. Tissue Eng Regen Med 2023; 20:1091-1108. [PMID: 37823990 PMCID: PMC10646012 DOI: 10.1007/s13770-023-00583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/12/2023] [Accepted: 08/04/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Guided bone regeneration (GBR) is commonly used to regenerate periodontal tissue. However, the bone inductivity and antibacterial properties of the GBR membranes currently in use are severely limited. This issue can be resolved by loading growth factors and antibiotics. Bioactive substitutes, such as Au nanoparticles (AuNPs) and carbon quantum dots (CDs), were proposed to prevent the denaturation of osteogenic growth factors and the induction of antibacterial drug resistance. METHODS Ornidazole was initially used as the raw material to prepare the CDs, followed by the incorporation of an optimal ratio of nanoparticles to produce the electrospun membrane doped with AuNPs and novel traceable antibacterial CDs. The morphology of the membrane was characterized. The adhesion, proliferation, and osteogenic differentiation of cells on the membrane were evaluated in vitro. The antimicrobial characteristics of the membrane were also investigated. The electrospun membrane was implanted into a rat skull defect model in vivo to investigate its osteogenic potential. RESULTS The blending of nanomaterials did not affect the micro morphology of the fiber, resulting in enhanced mechanical properties. Membranes doped with AuNPs and CDs exhibited excellent biocompatibility, increased ALP activity, improved calcified nodules, and increased expression of osteogenic-associated proteins, in addition to pronounced antibacterial effects. The membrane also demonstrated excellent osteogenic characteristics in rat models. CONCLUSION The synergistic effect of loaded AuNPs electrospun fiber membrane with CDs can promote periodontal bone regeneration and exert antibacterial activity.
Collapse
Affiliation(s)
- Jie Wang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
- Department of General of Dentistry, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hang Zhang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
| | - Yan Wang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
| | - Xiang Liu
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
| | - Weiwen Zhu
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
| | - Fei Jiang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
- Department of General of Dentistry, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Size Li
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
| | - Laikui Liu
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China.
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
13
|
Liu Y, Li X, Liu S, Du J, Xu J, Liu Y, Guo L. The changes and potential effects of zinc homeostasis in periodontitis microenvironment. Oral Dis 2023; 29:3063-3077. [PMID: 35996971 DOI: 10.1111/odi.14354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/28/2022] [Accepted: 08/14/2022] [Indexed: 11/29/2022]
Abstract
Zinc is a very important and ubiquitous element, which is present in oral environment, daily diet, oral health products, dental restorative materials, and so on. However, there is a lack of attention to the role of both extracellular or intracellular zinc in the progression of periodontitis and periodontal regeneration. This review summarizes the characteristics of immunological microenvironment and host cells function in several key stages of periodontitis progression, and explores the regulatory effect of zinc during this process. We find multiple evidence indicate that zinc may be involved and play a key role in the stages of immune defense, inflammatory response and bone remodeling. Zinc supplementation in an appropriate dose range or regulation of zinc transport proteins can promote periodontal regeneration by either enhancing immune defense or up-regulating local cells proliferation and differentiation functions. Therefore, zinc homeostasis is essential in periodontal remodeling and regeneration. More attention is suggested to be focused on zinc homeostasis regulation and consider it as a potential strategy in the studies on periodontitis treatment, periodontal-guided tissue regeneration, implant material transformation, and so on.
Collapse
Affiliation(s)
- Yitong Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Siyan Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Junji Xu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lijia Guo
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
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
|
15
|
Lin J, He Y, He Y, Feng Y, Wang X, Yuan L, Wang Y, Chen J, Luo F, Li Z, Li J, Tan H. Janus functional electrospun polyurethane fibrous membranes for periodontal tissue regeneration. J Mater Chem B 2023; 11:9223-9236. [PMID: 37700625 DOI: 10.1039/d3tb01407j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
The guided tissue regeneration (GTR) technique with GTR membranes is an efficient method for repairing periodontal defects. Conventional periodontal membranes act as physical barriers that resist the growth of fibroblasts, epithelial cells, and connective tissue. However, they cannot facilitate the regeneration of periodontal tissue. To address this issue, the exploitation of novel GTR membranes with bioactive functions based on therapeutic requirements is critical. Herein, we exploited a biodegradable bilayer polyurethane fibrous membrane by uniaxial electrostatic spinning to construct two sides with Janus properties by integrating the bioactive molecule dopamine (DA) and antimicrobial Gemini quaternary ammonium salt (QAS). The DA-containing side, located inside the injury, can effectively promote cell adhesion and mesenchymal stem cell growth as well as support mineralization and antioxidant properties, which are beneficial for bone regeneration. The QAS-containing side, located on the outer surface of the injury, endows antibacterial properties and limits fibroblast adhesion and growth on its surface owing to its strong hydrophilicity. An in vivo study demonstrates that the Janus polyurethane fibrous membrane can significantly promote the regeneration of periodontal defects in rats. Owing to its superior mechanical properties and biocompatibility, this polyurethane fibrous membrane has potential applications in the field of periodontal regeneration.
Collapse
Affiliation(s)
- Jingjing Lin
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Yushui He
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Yuanyuan He
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Yuan Feng
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Xiao Wang
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Lei Yuan
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Yanchao Wang
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, China
| | - Jie Chen
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610065, China
| | - Feng Luo
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Zhen Li
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Jiehua Li
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| | - Hong Tan
- College of Polymer Science and Engineering, State Kedy Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
16
|
Atia GA, Shalaby HK, Roomi AB, Ghobashy MM, Attia HA, Mohamed SZ, Abdeen A, Abdo M, Fericean L, Bănățean Dunea I, Atwa AM, Hasan T, Mady W, Abdelkader A, Ali SA, Habotta OA, Azouz RA, Malhat F, Shukry M, Foda T, Dinu S. Macro, Micro, and Nano-Inspired Bioactive Polymeric Biomaterials in Therapeutic, and Regenerative Orofacial Applications. Drug Des Devel Ther 2023; 17:2985-3021. [PMID: 37789970 PMCID: PMC10543943 DOI: 10.2147/dddt.s419361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/12/2023] [Indexed: 10/05/2023] Open
Abstract
Introducing dental polymers has accelerated biotechnological research, advancing tissue engineering, biomaterials development, and drug delivery. Polymers have been utilized effectively in dentistry to build dentures and orthodontic equipment and are key components in the composition of numerous restorative materials. Furthermore, dental polymers have the potential to be employed for medication administration and tissue regeneration. To analyze the influence of polymer-based investigations on practical medical trials, it is required to evaluate the research undertaken in this sector. The present review aims to gather evidence on polymer applications in dental, oral, and maxillofacial reconstruction.
Collapse
Affiliation(s)
- Gamal A Atia
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Hany K Shalaby
- Department of Oral Medicine, Periodontology and Oral Diagnosis, Faculty of Dentistry, Suez University, Suez, Egypt
| | - Ali B Roomi
- Department of Quality Assurance, University of Thi-Qar, Thi-Qar, Iraq
- Department of Medical Laboratory, College of Health and Medical Technology, National University of Science and Technology, Thi-Qar, Iraq
| | - Mohamed M Ghobashy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Hager A Attia
- Department of Molecular Biology and Chemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Sara Z Mohamed
- Department of Removable Prosthodontics, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Mohamed Abdo
- Department of Animal Histology and Anatomy, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Egypt
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, University of Sadat City, Sadat, Egypt
| | - Liana Fericean
- Department of Biology and Plant Protection, Faculty of Agriculture. University of Life Sciences “King Michael I” from Timișoara, Timișoara, Romania
| | - Ioan Bănățean Dunea
- Department of Biology and Plant Protection, Faculty of Agriculture. University of Life Sciences “King Michael I” from Timișoara, Timișoara, Romania
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Tabinda Hasan
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Wessam Mady
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Afaf Abdelkader
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Susan A Ali
- Department of Radiodiagnosis, Faculty of Medicine, Ain Shams University, Abbassia, 1181, Egypt
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Rehab A Azouz
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Farag Malhat
- Department of Pesticide Residues and Environmental Pollution, Central Agricultural Pesticide Laboratory, Agricultural Research Center, Giza, Egypt
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Tarek Foda
- Oral Health Sciences Department, Temple University’s Kornberg School of Dentistry, Philadelphia, PA, USA
| | - Stefania Dinu
- Department of Pedodontics, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy Timisoara, Timisoara, 300041, Romania
| |
Collapse
|
17
|
Blaudez F, Ivanovski S, Vaquette C. Harnessing the Native Extracellular Matrix for Periodontal Regeneration Using a Melt Electrowritten Biphasic Scaffold. J Funct Biomater 2023; 14:479. [PMID: 37754893 PMCID: PMC10531993 DOI: 10.3390/jfb14090479] [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: 08/07/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Scaffolds have been used to promote periodontal regeneration by providing control over the spacio-temporal healing of the periodontium (cementum, periodontal ligament (PDL) and alveolar bone). This study proposes to enhance the biofunctionality of a biphasic scaffold for periodontal regeneration by means of cell-laid extracellular matrix (ECM) decoration. To this end, a melt electrowritten scaffold was cultured with human osteoblasts for the deposition of bone-specific ECM. In parallel, periodontal ligament cells were used to form a cell sheet, which was later combined with the bone ECM scaffold to form a biphasic PDL-bone construct. The resulting biphasic construct was decellularised to remove all cellular components while preserving the deposited matrix. Decellularisation efficacy was confirmed in vitro, before the regenerative performance of freshly decellularised constructs was compared to that of 3-months stored freeze-dried scaffolds in a rodent periodontal defect model. Four weeks post-surgery, microCT revealed similar bone formation in all groups. Histology showed higher amounts of newly formed cementum and periodontal attachment in the fresh and freeze-dried ECM functionalised scaffolds, although it did not reach statistical significance. This study demonstrated that the positive effect of ECM decoration was preserved after freeze-drying and storing the construct for 3 months, which has important implications for clinical translation.
Collapse
Affiliation(s)
- Fanny Blaudez
- School of Dentistry, Centre for Oral Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Herston, QLD 4006, Australia; (F.B.); (S.I.)
- School of Dentistry and Oral Health, Griffith University, Southport, QLD 4222, Australia
| | - Saso Ivanovski
- School of Dentistry, Centre for Oral Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Herston, QLD 4006, Australia; (F.B.); (S.I.)
| | - Cedryck Vaquette
- School of Dentistry and Oral Health, Griffith University, Southport, QLD 4222, Australia
| |
Collapse
|
18
|
Größbacher G, Bartolf-Kopp M, Gergely C, Bernal PN, Florczak S, de Ruijter M, Rodriguez NG, Groll J, Malda J, Jungst T, Levato R. Volumetric Printing Across Melt Electrowritten Scaffolds Fabricates Multi-Material Living Constructs with Tunable Architecture and Mechanics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300756. [PMID: 37099802 DOI: 10.1002/adma.202300756] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/17/2023] [Indexed: 06/19/2023]
Abstract
Major challenges in biofabrication revolve around capturing the complex, hierarchical composition of native tissues. However, individual 3D printing techniques have limited capacity to produce composite biomaterials with multi-scale resolution. Volumetric bioprinting recently emerged as a paradigm-shift in biofabrication. This ultrafast, light-based technique sculpts cell-laden hydrogel bioresins into 3D structures in a layerless fashion, providing enhanced design freedom over conventional bioprinting. However, it yields prints with low mechanical stability, since soft, cell-friendly hydrogels are used. Herein, the possibility to converge volumetric bioprinting with melt electrowriting, which excels at patterning microfibers, is shown for the fabrication of tubular hydrogel-based composites with enhanced mechanical behavior. Despite including non-transparent melt electrowritten scaffolds in the volumetric printing process, high-resolution bioprinted structures are successfully achieved. Tensile, burst, and bending mechanical properties of printed tubes are tuned altering the electrowritten mesh design, resulting in complex, multi-material tubular constructs with customizable, anisotropic geometries that better mimic intricate biological tubular structures. As a proof-of-concept, engineered tubular structures are obtained by building trilayered cell-laden vessels, and features (valves, branches, fenestrations) that can be rapidly printed using this hybrid approach. This multi-technology convergence offers a new toolbox for manufacturing hierarchical and mechanically tunable multi-material living structures.
Collapse
Affiliation(s)
- Gabriel Größbacher
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Michael Bartolf-Kopp
- Department of Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Csaba Gergely
- Department of Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Paulina Núñez Bernal
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Sammy Florczak
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Mylène de Ruijter
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Núria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, The Netherlands
| | - Tomasz Jungst
- Department of Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, The Netherlands
| |
Collapse
|
19
|
Namazi SS, Mahmoud AH, Dal-Fabbro R, Han Y, Xu J, Sasaki H, Fenno JC, Bottino MC. Multifunctional and biodegradable methacrylated gelatin/Aloe vera nanofibers for endodontic disinfection and immunomodulation. BIOMATERIALS ADVANCES 2023; 150:213427. [PMID: 37075551 PMCID: PMC11027083 DOI: 10.1016/j.bioadv.2023.213427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/21/2023] [Accepted: 04/08/2023] [Indexed: 04/21/2023]
Abstract
Currently employed approaches and materials used for vital pulp therapies (VPTs) and regenerative endodontic procedures (REPs) lack the efficacy to predictably achieve successful outcomes due to their inability to achieve adequate disinfection and/or lack of desired immune modulatory effects. Natural polymers and medicinal herbs are biocompatible, biodegradable, and present several therapeutic benefits and immune-modulatory properties; thus, standing out as a clinically viable approach capable of establishing a conducive environment devoid of bacteria and inflammation to support continued root development, dentinal bridge formation, and dental pulp tissue regeneration. However, the low stability and poor mechanical properties of the natural compounds have limited their application as potential biomaterials for endodontic procedures. In this study, Aloe vera (AV), as a natural antimicrobial and anti-inflammatory agent, was incorporated into photocrosslinkable Gelatin methacrylate (GelMA) nanofibers with the purpose of developing a highly biocompatible biomaterial capable of eradicating endodontic infection and modulating inflammation. Stable GelMA/AV nanofibers with optimal properties were obtained at the ratio of (70:30) by electrospinning. In addition to the pronounced antibacterial effect against Enterococcus faecalis, the GelMA/AV (70:30) nanofibers also exhibited a sustained antibacterial activity over 14 days and significant biofilm reduction with minimal cytotoxicity, as well as anti-inflammatory properties and immunomodulatory effects favoring healing. Our results indicate that the novel GelMA/AV (70:30) nanofibers hold great potential as a biomaterial strategy for endodontic infection eradication and enhanced healing.
Collapse
Affiliation(s)
- Sharon S Namazi
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Abdel H Mahmoud
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Han
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Jinping Xu
- 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
| | - J Christopher Fenno
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, 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
|
20
|
Toledo PTA, Anselmi C, Dal-Fabbro R, Mahmoud AH, Abel AK, Becker ML, Delbem ACB, Bottino MC. Calcium Trimetaphosphate-Loaded Electrospun Poly(Ester Urea) Nanofibers for Periodontal Tissue Engineering. J Funct Biomater 2023; 14:350. [PMID: 37504845 PMCID: PMC10381820 DOI: 10.3390/jfb14070350] [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: 05/24/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
The objective of this research was to create and appraise biodegradable polymer-based nanofibers containing distinct concentrations of calcium trimetaphosphate (Ca-TMP) for periodontal tissue engineering. Poly(ester urea) (PEU) (5% w/v) solutions containing Ca-TMP (15%, 30%, 45% w/w) were electrospun into fibrous scaffolds. The fibers were evaluated using SEM, EDS, TGA, FTIR, XRD, and mechanical tests. Degradation rate, swelling ratio, and calcium release were also evaluated. Cell/Ca-TMP and cell/scaffold interaction were assessed using stem cells from human exfoliated deciduous teeth (SHEDs) for cell viability, adhesion, and alkaline phosphatase (ALP) activity. Analysis of variance (ANOVA) and post-hoc tests were used (α = 0.05). The PEU and PEU/Ca-TMP-based membranes presented fiber diameters at 469 nm and 414-672 nm, respectively. Chemical characterization attested to the Ca-TMP incorporation into the fibers. Adding Ca-TMP led to higher degradation stability and lower dimensional variation than the pure PEU fibers; however, similar mechanical characteristics were observed. Minimal calcium was released after 21 days of incubation in a lipase-enriched solution. Ca-TMP extracts enhanced cell viability and ALP activity, although no differences were found between the scaffold groups. Overall, Ca-TMP was effectively incorporated into the PEU fibers without compromising the morphological properties but did not promote significant cell function.
Collapse
Affiliation(s)
- Priscila T. A. Toledo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; (P.T.A.T.); (C.A.); (R.D.-F.); (A.H.M.)
- Department of Preventive and Restorative Dentistry, School of Dentistry, São Paulo State University (UNESP), Araçatuba 16015-050, SP, Brazil;
| | - Caroline Anselmi
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; (P.T.A.T.); (C.A.); (R.D.-F.); (A.H.M.)
- Department of Morphology and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-385, SP, Brazil
| | - Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; (P.T.A.T.); (C.A.); (R.D.-F.); (A.H.M.)
| | - Abdel H. Mahmoud
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; (P.T.A.T.); (C.A.); (R.D.-F.); (A.H.M.)
| | - Alexandra K. Abel
- Departments of Chemistry, Mechanical Engineering and Material Science, Orthopaedic Surgery, Duke University, Durham, NC 27708, USA; (A.K.A.); (M.L.B.)
| | - Matthew L. Becker
- Departments of Chemistry, Mechanical Engineering and Material Science, Orthopaedic Surgery, Duke University, Durham, NC 27708, USA; (A.K.A.); (M.L.B.)
| | - Alberto C. B. Delbem
- Department of Preventive and Restorative Dentistry, School of Dentistry, São Paulo State University (UNESP), Araçatuba 16015-050, SP, Brazil;
| | - Marco C. Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; (P.T.A.T.); (C.A.); (R.D.-F.); (A.H.M.)
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
21
|
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
|
22
|
Jin R, Ning X, Liu X, Zhao Y, Ye G. Porphyromonas gingivalis-induced periodontitis could contribute to cognitive impairment in Sprague–Dawley rats via the P38 MAPK signaling pathway. Front Cell Neurosci 2023; 17:1141339. [PMID: 37056710 PMCID: PMC10086325 DOI: 10.3389/fncel.2023.1141339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundPeriodontitis is one of the most common oral diseases and has been shown to be a risk factor for systemic diseases. Our aim was to investigate the relationship between periodontitis and cognitive impairment and to explore the role of the P38 MAPK signaling pathway in this process.MethodsWe established a periodontitis model by ligating the first molars of SD rats with silk thread and injecting Porphyromonas gingivalis (P. gingivalis) or P. gingivalis plus the P38 MAPK inhibitor SB203580 at the same time for ten weeks. We assessed alveolar bone resorption and spatial learning and memory using microcomputed tomography and the Morris water maze test, respectively. We used transcriptome sequencing to explore the genetic differences between the groups. The gingival tissue, peripheral blood and hippocampal tissue were assessed for the cytokines TNF-α, IL-1β, IL-6, IL-8 and C reactive protein (CRP) with enzyme-linked immunosorbent assay (ELISA) and reverse transcription polymerase chain reaction (RT–PCR). We observed the presence of P. gingivalis in the hippocampus of rats by paraffin-fluorescence in situ hybridization (FISH). We determined the activation of microglia by immunofluorescence. Finally, Western blot analysis was employed to determine the expression of amyloid precursor protein (APP), β-site APP-cleaving enzyme 1 (BACE1) and P38MAPK pathway activation.ResultsWe demonstrated that silk ligature-induced periodontitis plus injection of P. gingivalis into subgingival tissue could lead to memory and cognitive impairment. Transcriptome sequencing results suggested that there were neurodegenerative diseases in the P. gingivalis group, and the MWM test showed that periodontitis reduced the spatial learning and memory ability of mild cognitive impairment (MCI) model rats. We found high levels of inflammatory factors (TNF-α, IL-1β, IL-6, and IL-8) and CRP in the gingiva, peripheral blood and hippocampus, and the expression of APP and BACE1 was upregulated, as was the P38 MAPK pathway activation. Activated microglia and the presence of P. gingivalis were also found in the hippocampus. P38 MAPK inhibitors mitigated all of these changes.ConclusionOur findings strongly suggest that topical application of P. gingivalis increases the inflammatory burden in the peripheral and central nervous systems (CNS) and that neuroinflammation induced by activation of P38 MAPK leads to impaired learning and memory in SD rats. It can also modulate APP processing. Therefore, P38 MAPK may serve as a linking pathway between periodontitis and cognitive impairment.
Collapse
Affiliation(s)
- Ru Jin
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoqiao Ning
- The First People’s Hospital of Wanzhou, Chongqing, China
| | - Xiang Liu
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Guo Ye
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital, Chongqing Medical University, Chongqing, China
- *Correspondence: Guo Ye,
| |
Collapse
|
23
|
Golafshan N, Castilho M, Daghrery A, Alehosseini M, van de Kemp T, Krikonis K, de Ruijter M, Dal-Fabbro R, Dolatshahi-Pirouz A, Bhaduri SB, Bottino MC, Malda J. Composite Graded Melt Electrowritten Scaffolds for Regeneration of the Periodontal Ligament-to-Bone Interface. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12735-12749. [PMID: 36854044 PMCID: PMC11022588 DOI: 10.1021/acsami.2c21256] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Periodontitis is a ubiquitous chronic inflammatory, bacteria-triggered oral disease affecting the adult population. If left untreated, periodontitis can lead to severe tissue destruction, eventually resulting in tooth loss. Despite previous efforts in clinically managing the disease, therapeutic strategies are still lacking. Herein, melt electrowriting (MEW) is utilized to develop a compositionally and structurally tailored graded scaffold for regeneration of the periodontal ligament-to-bone interface. The composite scaffolds, consisting of fibers of polycaprolactone (PCL) and fibers of PCL-containing magnesium phosphate (MgP) were fabricated using MEW. To maximize the bond between bone (MgP) and ligament (PCL) regions, we evaluated two different fiber architectures in the interface area. These were a crosshatch pattern at a 0/90° angle and a random pattern. MgP fibrous scaffolds were able to promote in vitro bone formation even in culture media devoid of osteogenic supplements. Mechanical properties after MgP incorporation resulted in an increase of the elastic modulus and yield stress of the scaffolds, and fiber orientation in the interfacial zone affected the interfacial toughness. Composite graded MEW scaffolds enhanced bone fill when they were implanted in an in vivo periodontal fenestration defect model in rats. The presence of an interfacial zone allows coordinated regeneration of multitissues, as indicated by higher expression of bone, ligament, and cementoblastic markers compared to empty defects. Collectively, MEW-fabricated scaffolds having compositionally and structurally tailored zones exhibit a good mimicry of the periodontal complex, with excellent regenerative capacity and great potential as a defect-specific treatment strategy.
Collapse
Affiliation(s)
- Nasim Golafshan
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| | - Miguel Castilho
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, United States
| | - Morteza Alehosseini
- Technical University of Denmark, Department of Health Technology, Lyngby, Denmark
| | - Tom van de Kemp
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| | - Konstantinos Krikonis
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| | - Mylene de Ruijter
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| | - Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, United States
| | | | - Sarit B. Bhaduri
- Department of Mechanical, Industrial and Manufacturing Engineering, University of Toledo, Toledo, Ohio, United States
- EEC Division, Directorate of Engineering, The National Science Foundation, Alexandria, Virginia, United States
| | - Marco C. Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, United States
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan, United States
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
24
|
Yan Y, Ji Q, Fu R, Liu C, Yang J, Yin X, Li Q, Huang R. Biomaterials and tissue engineering strategies for posterior lamellar eyelid reconstruction: Replacement or regeneration? Bioeng Transl Med 2023. [DOI: 10.1002/btm2.10497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Affiliation(s)
- Yuxin Yan
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiumei Ji
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Rao Fu
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Chuanqi Liu
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jing Yang
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiya Yin
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Plastic and Burn Surgery West China Hospital, Sichuan University Chengdu China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ru‐Lin Huang
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
25
|
Immune microenvironment: novel perspectives on bone regeneration disorder in osteoradionecrosis of the jaws. Cell Tissue Res 2023; 392:413-430. [PMID: 36737519 DOI: 10.1007/s00441-023-03743-z] [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: 06/23/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023]
Abstract
Osteoradionecrosis of the jaws (ORNJ) is a severe complication that occurs after radiotherapy of head and neck malignancies. Clinically, conservative treatments and surgeries for ORNJ exhibited certain therapeutic effects, whereas the regenerative disorder of the post-radiation jaw remains a pending problem to be solved. In recent years, the recognition of the role of the immune microenvironment has led to a shift from an osteoblasts (OBs) or bone marrow mesenchymal stromal cells (BMSCs)-centered view of bone regeneration to the concept of a complicated microecosystem that supports bone regeneration. Current advances in osteoimmunology have uncovered novel targets within the immune microenvironment to help improve various regeneration therapies, notably therapies potentiating the interaction between BMSCs and immune cells. However, these researches lack a thorough understanding of the immune microenvironment and the interaction network of immune cells in the course of bone regeneration, especially for the post-operative defect of ORNJ. This review summarized the composition of the immune microenvironment during bone regeneration, how the immune microenvironment interacts with the skeletal system, and discussed existing and potential strategies aimed at targeting cellular and molecular immune microenvironment components.
Collapse
|
26
|
Cui H, You Y, Cheng GW, Lan Z, Zou KL, Mai QY, Han YH, Chen H, Zhao YY, Yu GT. Advanced materials and technologies for oral diseases. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2023; 24:2156257. [PMID: 36632346 PMCID: PMC9828859 DOI: 10.1080/14686996.2022.2156257] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/15/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Oral disease, as a class of diseases with very high morbidity, brings great physical and mental damage to people worldwide. The increasing burden and strain on individuals and society make oral diseases an urgent global health problem. Since the treatment of almost all oral diseases relies on materials, the rapid development of advanced materials and technologies has also promoted innovations in the treatment methods and strategies of oral diseases. In this review, we systematically summarized the application strategies in advanced materials and technologies for oral diseases according to the etiology of the diseases and the comparison of new and old materials. Finally, the challenges and directions of future development for advanced materials and technologies in the treatment of oral diseases were refined. This review will guide the fundamental research and clinical translation of oral diseases for practitioners of oral medicine.
Collapse
Affiliation(s)
- Hao Cui
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yan You
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Guo-Wang Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhou Lan
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Long Zou
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Qiu-Ying Mai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan-Hua Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yu-Yue Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Guang-Tao Yu
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Xuan Y, Li L, Zhang C, Zhang M, Cao J, Zhang Z. The 3D-Printed Ordered Bredigite Scaffold Promotes Pro-Healing of Critical-Sized Bone Defects by Regulating Macrophage Polarization. Int J Nanomedicine 2023; 18:917-932. [PMID: 36844434 PMCID: PMC9951604 DOI: 10.2147/ijn.s393080] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/29/2023] [Indexed: 02/22/2023] Open
Abstract
Background Repairing critical-sized bone defects secondary to traumatic or tumorous damage is a complex conundrum in clinical practice; in this case, artificial scaffolds exhibited preferable outcomes. Bredigite (BRT, Ca7MgSi4O16) bioceramic possesses excellent physicochemical properties and biological activity as a promising candidate for bone tissue engineering. Methods Structurally ordered BRT (BRT-O) scaffolds were fabricated by a three-dimensional (3D) printing technique, and the random BRT (BRT-R) scaffolds and clinically available β-tricalcium phosphate (β-TCP) scaffolds were compared as control groups. Their physicochemical properties were characterized, and RAW 264.7 cells, bone marrow mesenchymal stem cells (BMSCs), and rat cranial critical-sized bone defect models were utilized for evaluating macrophage polarization and bone regeneration. Results The BRT-O scaffolds exhibited regular morphology and homogeneous porosity. In addition, the BRT-O scaffolds released higher concentrations of ionic products based on coordinated biodegradability than the β-TCP scaffolds. In vitro, the BRT-O scaffolds facilitated RWA264.7 cells polarization to pro-healing M2 macrophage phenotype, whereas the BRT-R and β-TCP scaffolds stimulated more pro-inflammatory M1-type macrophages. A conditioned medium derived from macrophages seeding on the BRT-O scaffolds notably promoted the osteogenic lineage differentiation of BMSCs in vitro. The cell migration ability of BMSCs was significantly enhanced under the BRT-O-induced immune microenvironment. Moreover, in rat cranial critical-sized bone defect models, the BRT-O scaffolds group promoted new bone formation with a higher proportion of M2-type macrophage infiltration and expression of osteogenesis-related markers. Therefore, in vivo, BRT-O scaffolds play immunomodulatory roles in promoting critical-sized bone defects by enhancing the polarization of M2 macrophages. Conclusion 3D-printed BRT-O scaffolds can be a promising option for bone tissue engineering, at least partly through macrophage polarization and osteoimmunomodulation.
Collapse
Affiliation(s)
- Yaowei Xuan
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lin Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chenping Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Min Zhang
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Junkai Cao
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhen Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| |
Collapse
|
28
|
Chen P, Zhang C, He P, Pan S, Zhong W, Wang Y, Xiao Q, Wang X, Yu W, He Z, Gao X, Song J. A Biomimetic Smart Nanoplatform as “Inflammation Scavenger” for Regenerative Therapy of Periodontal Tissue. Int J Nanomedicine 2022; 17:5165-5186. [PMID: 36388874 PMCID: PMC9642321 DOI: 10.2147/ijn.s384481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction The functional reconstruction of periodontal tissue defects remains a clinical challenge due to excessive and prolonged host response to various endogenous and exogenous pro-inflammatory stimuli. Thus, a biomimetic nanoplatform with the capability of modulating inflammatory response in a microenvironment-responsive manner is attractive for regenerative therapy of periodontal tissue. Methods Herein, a facile and green design of engineered bone graft materials was developed by integrating a biomimetic apatite nanocomposite with a smart-release coating, which could realize inflammatory modulation by “on-demand” delivery of the anti-inflammatory agent through a pH-sensing mechanism. Results In vitro and in vivo experiments demonstrated that this biocompatible nanoplatform could facilitate the clearance of reactive oxygen species in human periodontal ligament stem cells under inflammatory conditions via inhibiting the production of endogenous proinflammatory mediators, in turn contributing to the enhanced healing efficacy of periodontal tissue. Moreover, this system exhibited effective antimicrobial activity against common pathogenic bacteria in the oral cavity, which is beneficial for the elimination of exogenous pro-inflammatory factors from bacterial infection during healing of periodontal tissue. Conclusion The proposed strategy provides a versatile apatite nanocomposite as a promising “inflammation scavenger” and propels the development of intelligent bone graft materials for periodontal and orthopedic applications.
Collapse
Affiliation(s)
- Poyu Chen
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Chuangwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Ping He
- Department of Stomatology, Dazhou Central Hospital, Dazhou, SiChuan, 635000, People’s Republic of China
| | - Shengyuan Pan
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Yue Wang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Qingyue Xiao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Xinyan Wang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Wenliang Yu
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Zhangmin He
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
- Correspondence: Xiang Gao; Jinlin Song, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China, Tel/Fax +86 23 88860105; Tel/Fax +86 23 88860026, Email ;
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, People’s Republic of China
| |
Collapse
|