1
|
Wu H, Li Y, Shi L, Liu Y, Shen J. New Advances in Periodontal Functional Materials Based on Antibacterial, Anti-Inflammatory, and Tissue Regeneration Strategies. Adv Healthc Mater 2025:e2403206. [PMID: 39895157 DOI: 10.1002/adhm.202403206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/17/2025] [Indexed: 02/04/2025]
Abstract
With the global population aging, awareness of oral health is rising. Periodontitis, a widespread bacterial infectious disease, is gaining attention. Current novel biomaterials address key clinical issues like bacterial infection, gum inflammation, tooth loosening, and loss, focusing on antibacterial, anti-inflammatory, and tissue regeneration properties. However, strategies that integrate the advantages of these biomaterials to achieve synergistic therapeutic effects by clearing oral biofilms, inhibiting inflammation activation, and restoring periodontal soft and hard tissue functions remain very limited. Recent studies highlight the link between periodontitis and systemic diseases, underscoring the complexity of the periodontal disease. There is an urgent need to find comprehensive treatment plans that address clinical requirements. Whether by integrating new biomaterials to enhance existing periodontal treatments or by developing novel approaches to replace traditional therapies, these efforts will drive advancements in periodontitis treatment. Therefore, this review compares novel biomaterials with traditional treatments. It highlights the design concepts and mechanisms of these functional materials, focusing on their antibacterial, anti-inflammatory, and tissue regeneration properties, and discusses the importance of developing comprehensive treatment strategies. This review aims to provide guidance for emerging periodontitis research and to promote the development of precise and efficient treatment strategies.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| |
Collapse
|
2
|
Jeong H, Subramanian K, Lee JB, Byun H, Shin H, Yun JH. Anti-inflammatory and osteoconductive multi-functional nanoparticles for the regeneration of an inflamed alveolar bone defect. Biomater Sci 2025; 13:810-825. [PMID: 39749408 DOI: 10.1039/d4bm01280a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Infected alveolar bone defects pose challenging clinical issues due to disrupted intrinsic healing mechanisms. Thus, the employment of advanced biomaterials enabling the modulation of several aspects of bone regeneration is necessary. This study investigated the effect of multi-functional nanoparticles on anti-inflammatory/osteoconductive characteristics and bone repair in the context of inflamed bone abnormalities. Tannic-acid mineral nanoparticles (TMPs) were prepared by the supramolecular assembly of tannic acid with bioactive calcium and phosphate ions, which were subsequently incorporated into collagen plugs via molecular interactions. Under physiological conditions, in vitro analysis confirmed that tannic acid was dissociated and released, which significantly reduced the expression of pro-inflammatory genes in lipopolysaccharide (LPS)-activated RAW264.7 cells. Meanwhile, the bioactive ions of Ca2+ and PO43- synergistically increased the gene and protein expressions of osteogenic markers of bone marrow-derived stem cells. For in vivo studies, combined endodontic-periodontal lesions were induced in beagle dogs where the plugs were readily implanted. After 2 months of the implantation, analysis of micro-computed tomography and histomorphometry revealed that groups of dogs implanted with the plug incorporating TMPs exhibited a significant decrease in bone surface density as well as structural model index, and significant increase in the mineralized bone content, respectively, with positive OPN expression being observed in reversal lines. Notably, the profound improvement in bone regeneration relied on the concentration of TMPs in the implants, underscoring the promise of multi-functional nanoparticles for treating infected alveolar bones.
Collapse
Affiliation(s)
- Hyewoo Jeong
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Keerthana Subramanian
- Department of Periodontology, College of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea.
| | - Jong-Bin Lee
- Department of Periodontology, College of Dentistry and Research Institute of Oral Sciences, Gangneung-Wonju National University, Gangneung, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, Republic of Korea
| | - Jeong-Ho Yun
- Department of Periodontology, College of Dentistry and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, Republic of Korea.
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| |
Collapse
|
3
|
Deng J, Yao Z, Wang S, Zhang X, Zhan L, Wang T, Yu W, Zeng J, Wu J, Fu S, Wu S, Ouyang Y, Huang C. Uni-directional release of ibuprofen from an asymmetric fibrous membrane enables effective peritendinous anti-adhesion. J Control Release 2024; 372:251-264. [PMID: 38908755 DOI: 10.1016/j.jconrel.2024.06.046] [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: 12/16/2023] [Revised: 05/31/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Drug-loaded porous membranes have been deemed to be effective physicochemical barriers to separate postoperative adhesion-prone tissues in tendon healing. However, cell viability and subsequent tissue regeneration might be severely interfered with the unrestricted release and the locally excessive concentration of anti-inflammatory drugs. Herein, we report a double-layered membrane with sustained and uni-directional drug delivery features to prevent peritendinous adhesion without hampering the healing outcome. A vortex-assisted electrospinning system in combination with ibuprofen (IBU)-in-water emulsion was utilized to fabricate IBU-loaded poly-ʟ-lactic-acid (PLLA) fiber bundle membrane (PFB-IBU) as the anti-adhesion layer. The resultant highly porous structure, oleophilic and hydrophobic nature of PLLA fibers enabled in situ loading of IBU with a concentration gradient across the membrane thickness. Aligned collagen nanofibers were further deposited at the low IBU concentration side of the membrane for regulating cell growth and achieving uni-directional release of IBU. Drug release kinetics showed that the release amount of IBU from the high concentration side reached 79.32% at 14 d, while it was only 0.35% at the collagen side. Therefore, fibroblast proliferation at the high concentration side was successfully inhibited without affecting the oriented growth of tendon-derived stem cells at the other side. In vivo evaluation of the rat Achilles adhesion model confirmed the successful peritendinous anti-adhesion of our double-layered membrane, in that the macrophage recruitment, the inflammatory factor secretion and the deposition of pathological adhesion markers such as α-SMA and COL-III were all inhibited, which greatly improved the peritendinous fibrosis and restored the motor function of tendon.
Collapse
Affiliation(s)
- Jixia Deng
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Zhixiao Yao
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Shikun Wang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Xinyu Zhang
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Lei Zhan
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Tongyu Wang
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Wenhua Yu
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Jiamei Zeng
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Jinglei Wu
- Biomaterials and Tissue Engineering Laboratory, College of Chemistry and Chemical Engineering and Biological Engineering, Donghua University, Shanghai 201620, China
| | - Shaoju Fu
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Shihao Wu
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, China.
| | - Yuanming Ouyang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| | - Chen Huang
- Shanghai Frontiers Science Center of Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China.
| |
Collapse
|
4
|
Sarfi S, Azaryan E, Hanafi-Bojd MY, Emadian Razavi F, Naseri M. Green synthesis of nanohydroxyapatite with Elaeagnus angustifolia L. extract as a metronidazole nanocarrier for in vitro pulpitis model treatment. Sci Rep 2024; 14:14702. [PMID: 38926433 PMCID: PMC11208562 DOI: 10.1038/s41598-024-65582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
The aim of this study is to introduce a dental capping agent for the treatment of pulp inflammation (pulpitis). Nanohydroxyapatite with Elaeagnus angustifolia L. extract (nHAEA) loaded with metronidazole (nHAEA@MTZ) was synthesized and evaluated using a lipopolysaccharide (LPS) in vitro model of pulpitis. nHAEA was synthesized through sol-gel method and analyzed using Scanning Electron Microscopy, Transmission Electron Microscopy, and Brunauer Emmett Teller. Inflammation in human dental pulp stem cells (HDPSCs) induced by LPS. A scratch test assessed cell migration, RT PCR measured cytokines levels, and Alizarin red staining quantified odontogenesis. The nHAEA nanorods were 17-23 nm wide and 93-146 nm length, with an average pore diameter of 27/312 nm, and a surface area of 210.89 m2/g. MTZ loading content with controlled release, suggesting suitability for therapeutic applications. nHAEA@MTZ did not affect the odontogenic abilities of HDPSCs more than nHAEA. However, it was observed that nHAEA@MTZ demonstrated a more pronounced anti-inflammatory effect. HDPSCs treated with nanoparticles exhibited improved migration compared to other groups. These findings demonstrated that nHAEA@MTZ could be an effective material for pulp capping and may be more effective than nHAEA in reducing inflammation and activating HDPSCs to enhance pulp repair after pulp damage.
Collapse
Affiliation(s)
- Sepideh Sarfi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Ehsaneh Azaryan
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Yahya Hanafi-Bojd
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Fariba Emadian Razavi
- Dental Research Center, Faculty of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Naseri
- Cellular and Molecular Research Center, Department of Molecular Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
5
|
Muresan GC, Boca S, Lucaciu O, Hedesiu M. The Applicability of Nanostructured Materials in Regenerating Soft and Bone Tissue in the Oral Cavity-A Review. Biomimetics (Basel) 2024; 9:348. [PMID: 38921228 PMCID: PMC11201588 DOI: 10.3390/biomimetics9060348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Two of the most exciting new technologies are biotechnology and nanotechnology. The science of nanostructures, or nanotechnology, is concerned with the development, testing, and use of structures and molecules with nanoscale dimensions ranging from 1 to 100 nm. The development of materials and tools with high specificity that interact directly at the subcellular level is what makes nanotechnology valuable in the medical sciences. At the cellular or tissue level, this might be converted into focused clinical applications with the greatest possible therapeutic benefits and the fewest possible side effects. The purpose of the present study was to review the literature and explore the applicability of the nanostructured materials in the process of the regeneration of the soft and hard tissues of the oral cavity. MATERIALS AND METHODS An electronic search of articles was conducted in several databases, such as PubMed, Embase, and Web of Science, to conduct this study, and the 183 articles that were discovered were chosen and examined, and only 22 articles met the inclusion criteria in this review. RESULTS The findings of this study demonstrate that using nanoparticles can improve the mechanical properties, biocompatibility, and osteoinductivity of biomaterials. CONCLUSIONS Most recently, breakthroughs in tissue engineering and nanotechnology have led to significant advancements in the design and production of bone graft substitutes and hold tremendous promise for the treatment of bone abnormalities. The creation of intelligent nanostructured materials is essential for various applications and therapies, as it allows for the precise and long-term delivery of medication, which yields better results.
Collapse
Affiliation(s)
- Giorgiana Corina Muresan
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Sanda Boca
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babes-Bolyai University, 400271 Cluj-Napoca, Romania;
| | - Ondine Lucaciu
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Mihaela Hedesiu
- Department of Oral Radiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| |
Collapse
|
6
|
Gao H, Chen N, Sun L, Sheng D, Zhong Y, Huang M, Yu C, Yang X, Hao Y, Chen S, Shao Z, Chen J. Time-programmed release of curcumin and Zn 2+ from multi-layered RSF coating modified PET graft for improvement of graft-host integration. Int J Biol Macromol 2024; 272:132830. [PMID: 38825264 DOI: 10.1016/j.ijbiomac.2024.132830] [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: 10/11/2023] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Artificial graft serves as the primary grafts used in the clinical management of sports-related injuries. Until now, optimizing its graft-host integration remains a great challenge due to the excessive inflammatory response during the inflammatory phase, coupled with an absence of tissue-inductive capacity during the regeneration phase. Here, a multi-layered regenerated silk fibroin (RSF) coating loaded with curcumin (Cur) and Zn2+ on the surface of the PET grafts (Cur@Zn2+@PET) was designed and fabricated for providing time-matched regulation specifically tailored to address issues arising at both inflammatory and regeneration phases, respectively. The release of Cur and Zn2+ from the Cur@Zn2+@PET followed a time-programmed pattern in vitro. Specifically, cellular assays revealed that Cur@Zn2+@PET initially released Cur during the inflammatory phase, thereby markedly inhibit the expression of inflammatory cytokines TNF-a and IL-1β. Meanwhile, a significant release of Zn2+ was major part during the regeneration phase, serving to induce the osteogenic differentiation of rBMSC. Furthermore, rat model of anterior cruciate ligament reconstruction (ACLR) showed that through time-programmed drug release, Cur@Zn2+@PET could suppress the formation of fibrous interface (FI) caused by inflammatory response, combined with significant new bone (NB) formation during regeneration phase. Consequently, the implementation of the Cur@Zn2+@PET characterized by its time-programmed release patterns hold considerable promise for improving graft-host integration for sports-related injuries.
Collapse
Affiliation(s)
- Han Gao
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Ni Chen
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials and Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Luyi Sun
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Dandan Sheng
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Yuting Zhong
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Mingru Huang
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Chengxuan Yu
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Xing Yang
- Department of orthopedics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215500, Jiangsu, China
| | - Yuefeng Hao
- Department of orthopedics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215500, Jiangsu, China
| | - Shiyi Chen
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China.
| | - Zhengzhong Shao
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials and Department of Macromolecular Science, Fudan University, Shanghai 200433, China.
| | - Jun Chen
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China.
| |
Collapse
|
7
|
Garg U, Dua T, Kaul S, Jain N, Pandey M, Nagaich U. Enhancing periodontal defences with nanofiber treatment: recent advances and future prospects. J Drug Target 2024; 32:470-484. [PMID: 38404239 DOI: 10.1080/1061186x.2024.2321372] [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: 12/02/2023] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
The term periodontal disease is used to define diseases characterised by inflammation and regeneration of the gums, cementum, supporting bone, and periodontal ligament. The conventional treatment involves the combination of scaling, root planning, and surgical approaches which are invasive and can pose certain challenges. Intrapocket administration of nanofibers can be used for overcoming challenges which can help in speeding up the wound repair process and can also be used to promote osteogenesis. To help make drug delivery more effective, nanofibers are an interesting solution. Nanofibers are nanosized 3D structures that can fill the pockets and have excellent mucoadhesion which prolongs their retention time on the target site. Moreover, their structure mimics the natural extracellular matrix which enables nanomaterials to sense local biological conditions and start cellular-level reprogramming to produce the necessary therapeutic efficacy. In this review, the significance of intrapocket administration of nanofibers using recent research for the management of periodontitis has been discussed in detail. Furthermore, we have discussed polymers used for the preparation of nanofibers, nanofiber production methods, and the patents associated with these developments. This comprehensive compilation of data serves as a valuable resource, consolidating recent developments in nanofiber applications for periodontitis management into one accessible platform.
Collapse
Affiliation(s)
- Unnati Garg
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, UP, India
| | - Tanya Dua
- Department of Periodontology, Inderprastha Dental College and Hospital, Atal Bihari Vajpayee Medical University, Lucknow, UP, India
| | - Shreya Kaul
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, UP, India
| | - Neha Jain
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, UP, India
| | - Manisha Pandey
- Department of Pharmaceutical Sciences, Central University of Haryana, India
| | - Upendra Nagaich
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, UP, India
| |
Collapse
|
8
|
Kandaswamy K, Subramanian R, Giri J, Guru A, Arockiaraj J. A Robust Strategy Against Multi-Resistant Pathogens in Oral Health: Harnessing the Potency of Antimicrobial Peptides in Nanofiber-Mediated Therapies. Int J Pept Res Ther 2024; 30:35. [DOI: 10.1007/s10989-024-10613-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2024] [Indexed: 01/12/2025]
|
9
|
Huang X, Lou Y, Duan Y, Liu H, Tian J, Shen Y, Wei X. Biomaterial scaffolds in maxillofacial bone tissue engineering: A review of recent advances. Bioact Mater 2024; 33:129-156. [PMID: 38024227 PMCID: PMC10665588 DOI: 10.1016/j.bioactmat.2023.10.031] [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: 08/28/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Maxillofacial bone defects caused by congenital malformations, trauma, tumors, and inflammation can severely affect functions and aesthetics of maxillofacial region. Despite certain successful clinical applications of biomaterial scaffolds, ideal bone regeneration remains a challenge in maxillofacial region due to its irregular shape, complex structure, and unique biological functions. Scaffolds that address multiple needs of maxillofacial bone regeneration are under development to optimize bone regeneration capacity, costs, operational convenience. etc. In this review, we first highlight the special considerations of bone regeneration in maxillofacial region and provide an overview of the biomaterial scaffolds for maxillofacial bone regeneration under clinical examination and their efficacy, which provide basis and directions for future scaffold design. Latest advances of these scaffolds are then discussed, as well as future perspectives and challenges. Deepening our understanding of these scaffolds will help foster better innovations to improve the outcome of maxillofacial bone tissue engineering.
Collapse
Affiliation(s)
- Xiangya Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yaxin Lou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yihong Duan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - He Liu
- Division of Endodontics, Department of Oral Biological and Medical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jun Tian
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Ya Shen
- Division of Endodontics, Department of Oral Biological and Medical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
10
|
Li P, Xu T, Dang X, Shao L, Yan L, Yang X, Lin L, Ren L, Song R. Improving astaxanthin-loaded chitosan/polyvinyl alcohol/graphene oxide nanofiber membranes and their application in periodontitis. Int J Biol Macromol 2024; 258:128980. [PMID: 38151084 DOI: 10.1016/j.ijbiomac.2023.128980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/02/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
Periodontitis is a chronic inflammatory disease primarily driven by host inflammation and plaque-induced immune responses. Controlling the host inflammatory response and improving the periodontal inflammatory microenvironment are crucial to promoting periodontal tissue regeneration. In this study, the blended nanofiber membranes previously prepared by our research group were improved, and we developed multifunctional chitosan/polyvinyl alcohol/graphene oxide/astaxanthin coaxial nanofiber membranes. Scanning electron microscopy showed that the prepared nanofibers had a smooth surface and a uniform diameter distribution. The mechanical property test results showed that the coaxial nanofiber membranes exhibited higher tensile strength compared to the blended nanofiber membranes, which increased from 4.50 ± 0.32 and 3.70 ± 0.45 MPa to 7.12 ± 0.22 and 5.62 ± 0.79 MPa respectively. Drug release studies indicated that the "shell-core" structure of coaxial nanofibers significantly reduced the initial burst release of astaxanthin (ASTA), with only 13.49 % and 10.71 % release in the first 24 h, and drug release lasted for over a week. Animal experiments confirmed that the coaxial nanofiber membranes loaded with ASTA promoted periodontal bone defect repair while inhibiting periodontal inflammation. In conclusion, the prepared coaxial nanofiber membranes are a promising sustained-release drug system for treating periodontitis.
Collapse
Affiliation(s)
- Pei Li
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, No. 143 Yiman Street, Nangang District, Harbin 150001, China
| | - Tao Xu
- School of Medicine Huaqiao University, No. 269 Chenghua North Road, Quanzhou 362000, China
| | - Xuan Dang
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, No. 143 Yiman Street, Nangang District, Harbin 150001, China
| | - Lu Shao
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Linlin Yan
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Xiaobin Yang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Lexun Lin
- Department of Pathogenic Microbiology, School of Basic Medical Sciences, Harbin Medical University, No. 157 Baojian Street, Nangang District, Harbin 150081, China
| | - Liping Ren
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, No. 143 Yiman Street, Nangang District, Harbin 150001, China
| | - Rong Song
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, No. 143 Yiman Street, Nangang District, Harbin 150001, China.
| |
Collapse
|
11
|
Mirhaj M, Varshosaz J, Labbaf S, Emadi R, Seifalian AM, Sharifianjazi F, Tavakoli M. Mupirocin loaded core-shell pluronic-pectin-keratin nanofibers improve human keratinocytes behavior, angiogenic activity and wound healing. Int J Biol Macromol 2023; 253:126700. [PMID: 37673152 DOI: 10.1016/j.ijbiomac.2023.126700] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 08/11/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
In the current study, a core-shell nanofibrous wound dressing based on Pluronic-F127 (F127) containing 2 wt% mupirocin (Mup) core and pectin (Pec)-keratin (Kr) shell was fabricated through coaxial electrospinning technique, and the blended nanofibers were also fabricated from the same materials. The fiber diameter and specific surface area of the blended nanofibers were about 101.56 nm and 20.16 m2/g, while for core-shell nanofibers they were about 97.32 nm and 25.26 m2/g, respectively. The resultant blended and core-shell nanofibers experienced a degradation of 27.65 % and 32.28 % during 7 days, respectively. The drug release profile of core-shell nanofibers revealed a sustained release of Mup over 7 days (87.66 %), while the blended F127-Pec-Kr-Mup nanofibers had a burst release within the first few hours (89.38 % up to 48 h) and a cumulative release of 91.36 % after 7 days. Due to the controlled release of Mup, the core-shell structure significantly improved the human keratinocytes behavior, angiogenic potential and wound healing in a rat model compared to the blended structure. In conclusion, the F127-Mup/Pec-Kr core-shell nanofibrous wound dressing appears to be a promising candidate for the prevention of infection, and can potentially accelerate the recovery and healing of chronic and ischemic wounds.
Collapse
Affiliation(s)
- Marjan Mirhaj
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Centre, Department of Pharmaceutics, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Rahmatollah Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Alexander Marcus Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd, Nanoloom Ltd, Liberum Health Ltd), London BioScience Innovation Centre, London, United Kingdom
| | - Fariborz Sharifianjazi
- Department of Natural Sciences, School of Science and Technology, University of Georgia, Tbilisi 0171, Georgia.
| | - Mohamadreza Tavakoli
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
12
|
Wu L, Li J, Wang Y, Zhao X, He Y, Mao H, Tang W, Liu R, Luo K, Gu Z. Engineered Hierarchical Microdevices Enable Pre-Programmed Controlled Release for Postsurgical and Unresectable Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305529. [PMID: 37549042 DOI: 10.1002/adma.202305529] [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: 06/09/2023] [Revised: 07/24/2023] [Indexed: 08/09/2023]
Abstract
Drug treatment is required for both resectable and unresectable cancers to strive for any meaningful improvement in patient outcomes. However, the clinical benefit of receiving conventional systemic administrations is often less than satisfactory. Drug delivery systems are preferable substitutes but still fail to meet diverse clinical demands due to the difficulty in programming drug release profiles. Herein, a microfabrication concept, termed "Hierarchical Multiple Polymers Immobilization" (HMPI), is introduced and biodegradable-polymer-based hierarchical microdevices (HMDs) that can pre-program any desired controlled release profiles are engineered. Based on the first-line medication of pancreatic and breast cancer, controlled release of single gemcitabine and the doxorubicin/paclitaxel combination in situ for multiple courses is implemented, respectively. Preclinical models of postsurgical pancreatic, postsurgical breast, and unresectable breast cancer are established, and the designed HMDs are demonstrated as well-tolerable and effective treatments for inhibiting tumor growth, recurrence, and metastasis. The proposed HMPI strategy allows the creation of tailorable and high-resolution hierarchical microstructures for pre-programming controlled release according to clinical medication schedules, which may provide promising alternative treatments for postsurgical and unresectable tumor control.
Collapse
Affiliation(s)
- Lihuang Wu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Junhua Li
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Yuqi Wang
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Xinyue Zhao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, 210009, China
| | - Wenbo Tang
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Rong Liu
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwei Gu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
13
|
Xue Y, Zhang L, Liu F, Kong L, Ma D, Han Y. Fluoride releasing photothermal responsive TiO 2 matrices for antibiosis, biosealing and bone regeneration. J Control Release 2023; 363:657-669. [PMID: 37832724 DOI: 10.1016/j.jconrel.2023.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
Peri-implantitis induced by infection leads to gingival recession, alveolar resorption and eventual dental implant failure. So, antibiosis and biosealing of abutments as well as osseointegration of roots need to be projected seriously during the whole service lifespan of dental implants. In this work, a multipurpose photothermal therapy strategy based on Si/P/F doped TiO2 matrix is proposed to address the above issues. This TiO2 matrix not only has outstanding photothermal response, but also triggers the release of F ions under near-infrared (NIR) light irradiation. Local hyperthermia assisted with the released F ions reduces adenosine triphosphate (ATP) synthesis of staphylococcus aureus (S. aureus), increases bacterial membrane permeability, and induces abundant of reactive oxygen species, resulting in the oxidation of cellular components and eventual death of bacteria. Furthermore, the synergic action of mild photothermal stimulation and Si/P/F ions of TiO2 matrix up-regulates gingival epithelial cells behavior (e.g., hemidesmosome formation) and osteoblasts response in vitro. In an infected model, this TiO2 matrix obviously eliminates bacteria, reduces inflammatory response, improves epithelial sealing and osseointegration, and reduces alveolar resorption by regulating NIR irradiation.
Collapse
Affiliation(s)
- Yang Xue
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lan Zhang
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Fuwei Liu
- Fourth Military Medical University, Xi'an 710038, China
| | - Liang Kong
- Fourth Military Medical University, Xi'an 710038, China
| | - Dayan Ma
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yong Han
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China; Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
14
|
Karmakar R, Dey S, Alam A, Khandelwal M, Pati F, Rengan AK. Attributes of Nanomaterials and Nanotopographies for Improved Bone Tissue Engineering and Regeneration. ACS APPLIED BIO MATERIALS 2023; 6:4020-4041. [PMID: 37691480 DOI: 10.1021/acsabm.3c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Bone tissue engineering (BTE) is a multidisciplinary area that can solve the limitation of conventional grafting methods by developing viable and biocompatible bone replacements. The three essential components of BTE, i.e., Scaffold material and Cells and Growth factors altogether, facilitate support and guide for bone formation, differentiation of the bone tissues, and enhancement in the cellular activities and bone regeneration. However, there is a scarcity of the appropriate materials that can match the mechanical property as well as functional similarity to native tissue, considering the bone as hard tissue. In such scenarios, nanotechnology can be leveraged upon to achieve the desired aspects of BTE, and that is the key point of this review article. This review article examines the significant areas of nanotechnology research that have an impact on regeneration of bone: (a) scaffold with nanomaterials helps to enhance physicochemical interactions, biocompatibility, mechanical stability, and attachment; (b) nanoparticle-based approaches for delivering bioactive chemicals, growth factors, and genetic material. The article begins with the introduction of components and healing mechanisms of bone and the factors associated with them. The focus of this article is on the various nanotopographies that are now being used in scaffold formation, by describing how they are made, and how these nanotopographies affect the immune system and potential underlying mechanisms. The advantages of 4D bioprinting in BTE by using nanoink have also been mentioned. Additionally, we have investigated the importance of an in silico approach for finding the interaction between drugs and their related receptors, which can help to formulate suitable systems for delivery. This review emphasizes the role of nanoscale approach and how it helps to increase the efficacy of parameters of scaffold as well as drug delivery system for tissue engineering and bone regeneration.
Collapse
Affiliation(s)
- Rounik Karmakar
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Sreenath Dey
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Aszad Alam
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology, Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Mudrika Khandelwal
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology, Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| |
Collapse
|
15
|
Viglianisi G, Santonocito S, Lupi SM, Amato M, Spagnuolo G, Pesce P, Isola G. Impact of local drug delivery and natural agents as new target strategies against periodontitis: new challenges for personalized therapeutic approach. Ther Adv Chronic Dis 2023; 14:20406223231191043. [PMID: 37720593 PMCID: PMC10501082 DOI: 10.1177/20406223231191043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/07/2023] [Indexed: 09/19/2023] Open
Abstract
Periodontitis is a persistent inflammation of the soft tissue around the teeth that affects 60% of the population in the globe. The self-maintenance of the inflammatory process can cause periodontal damage from the alveolar bone resorption to tooth loss in order to contrast the effects of periodontitis, the main therapy used is scaling and root planing (SRP). At the same time, studying the physiopathology of periodontitis has shown the possibility of using a local drug delivery system as an adjunctive therapy. Using local drug delivery devices in conjunction with SRP therapy for periodontitis is a potential tool since it increases drug efficacy and minimizes negative effects by managing drug release. This review emphasized how the use of local drug delivery agents and natural agents could be promising adjuvants for the treatment of periodontitis patients affected or not by cardiovascular disease, diabetes, and other system problems. Moreover, the review evidences the current issues and new ideas that can inspire potential later study for both basic research and clinical practice for a tailored approach.
Collapse
Affiliation(s)
- Gaia Viglianisi
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Catania, Italy
| | - Saturnino Marco Lupi
- Department of Clinical Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Mariacristina Amato
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Catania, Italy
| | - Gianrico Spagnuolo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Naples, Italy
| | - Paolo Pesce
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialities, School of Dentistry, University of Catania, Via Santa Sofia 78, Catania 95123, Italy
| |
Collapse
|
16
|
Su BY, Chen ZJ, Lv JC, Wang ZG, Huang FW, Liu Y, Luo E, Wang J, Xu JZ, Li ZM. Scalable Fabrication of Polymeric Composite Microspheres to Inhibit Oral Pathogens and Promote Osteogenic Differentiation of Periodontal Membrane Stem Cells. ACS Biomater Sci Eng 2023; 9:4431-4441. [PMID: 37452570 DOI: 10.1021/acsbiomaterials.3c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Periodontitis is a worldwide bacterial infectious disease, resulting in the resorption of tooth-supporting structures. Biodegradable polymeric microspheres are emerging as an appealing local therapy candidate for periodontal defect regeneration but suffer from tedious procedures and low yields. Herein, we developed a facile yet scalable approach to prepare polylactide composite microspheres with outstanding drug-loading capability. It was realized by blending equimolar polylactide enantiomers at the temperature between the melting point of homocrystallites and stereocomplex (sc) crystallites, enabling the precipitation of sc crystallites in the form of microspheres. Meanwhile, epigallocatechin gallate (EGCG) and nano-hydroxyapatite were encapsulated in the microspheres in the designated amount. Such an assembly allowed the fast and sustained release of EGCG and Ca2+ ions. The resultant hybrid composite microspheres not only exhibited strong antimicrobial activity against typical oral pathogens (Porphyromonas gingivalis and Enterococcus faecalis), but also directly promoted osteogenic differentiation of periodontal ligament stem cells with good cytocompatibility. These dual-functional composite microspheres offer a desired drug delivery platform to address the practical needs for periodontitis treatment.
Collapse
Affiliation(s)
- Biao-Yao Su
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zi-Jian Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jia-Cheng Lv
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zhi-Guo Wang
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fu-Wen Huang
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Jing Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jia-Zhuang Xu
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
17
|
Zong C, Bronckaers A, Willems G, He H, Cadenas de Llano-Pérula M. Nanomaterials for Periodontal Tissue Regeneration: Progress, Challenges and Future Perspectives. J Funct Biomater 2023; 14:290. [PMID: 37367254 DOI: 10.3390/jfb14060290] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Bioactive nanomaterials are increasingly being applied in oral health research. Specifically, they have shown great potential for periodontal tissue regeneration and have substantially improved oral health in translational and clinical applications. However, their limitations and side effects still need to be explored and elucidated. This article aims to review the recent advancements in nanomaterials applied for periodontal tissue regeneration and to discuss future research directions in this field, especially focusing on research using nanomaterials to improve oral health. The biomimetic and physiochemical properties of nanomaterials such as metals and polymer composites are described in detail, including their effects on the regeneration of alveolar bone, periodontal ligament, cementum and gingiva. Finally, the biomedical safety issues of their application as regenerative materials are updated, with a discussion about their complications and future perspectives. Although the applications of bioactive nanomaterials in the oral cavity are still at an initial stage, and pose numerous challenges, recent research suggests that they are a promising alternative in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Chen Zong
- Department of Oral Health Sciences-Orthodontics, University of Leuven (KU Leuven) and Dentistry, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute, Faculty of Life Sciences, University of Hasselt, 3590 Diepenbeek, Belgium
| | - Guy Willems
- Department of Oral Health Sciences-Orthodontics, University of Leuven (KU Leuven) and Dentistry, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Hong He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Orthodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Center for Dentofacial Development and Sleep Medicine, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Maria Cadenas de Llano-Pérula
- Department of Oral Health Sciences-Orthodontics, University of Leuven (KU Leuven) and Dentistry, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
18
|
Amato M, Santonocito S, Polizzi A, Tartaglia GM, Ronsivalle V, Viglianisi G, Grippaudo C, Isola G. Local Delivery and Controlled Release Drugs Systems: A New Approach for the Clinical Treatment of Periodontitis Therapy. Pharmaceutics 2023; 15:pharmaceutics15041312. [PMID: 37111796 PMCID: PMC10143241 DOI: 10.3390/pharmaceutics15041312] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Periodontitis is an inflammatory disease of the gums characterized by the degeneration of periodontal ligaments, the formation of periodontal pockets, and the resorption of the alveolar bone, which results in the destruction of the teeth's supporting structure. Periodontitis is caused by the growth of diverse microflora (particularly anaerobes) in the pockets, releasing toxins and enzymes and stimulating the immune system. Various approaches, both local and systemic, have been used to treat periodontitis effectively. Successful treatment depends on reducing bacterial biofilm, bleeding on probing (BOP), and reducing or eliminating pockets. Currently, the use of local drug delivery systems (LDDSs) as an adjunctive therapy to scaling and root planing (SRP) in periodontitis is a promising strategy, resulting in greater efficacy and fewer adverse effects by controlling drug release. Selecting an appropriate bioactive agent and route of administration is the cornerstone of a successful periodontitis treatment plan. In this context, this review focuses on applications of LDDSs with varying properties in treating periodontitis with or without systemic diseases to identify current challenges and future research directions.
Collapse
Affiliation(s)
- Mariacristina Amato
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Gianluca Martino Tartaglia
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy
| | - Vincenzo Ronsivalle
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Gaia Viglianisi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Cristina Grippaudo
- Department of Head and Neck, Division of Oral Surgery and Implantology, Catholic University of the Sacred Heart, Fondazione Policlinico Gemelli IRCCS, 00168 Rome, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| |
Collapse
|
19
|
Zhao J, Wei Y, Xiong J, Liu H, Lv G, Zhao J, He H, Gou J, Yin T, Tang X, Zhang Y. Antibacterial-Anti-Inflammatory-Bone Restoration Procedure Achieved by MIN-Loaded PLGA Microsphere for Efficient Treatment of Periodontitis. AAPS PharmSciTech 2023; 24:74. [PMID: 36890400 DOI: 10.1208/s12249-023-02538-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/12/2023] [Indexed: 03/10/2023] Open
Abstract
The main development process of periodontitis involves periodontal pathogenic bacteria as the initiating factor causing the onset of destructive inflammation, which in turn stimulates the destruction of periodontal tissue. It is difficult to achieve the eradication of periodontitis due to the complex interaction among antibacterial, anti-inflammatory, and bone restoration. Herein, we propose an antibacterial-anti-inflammatory-bone restoration procedural treatment strategy with minocycline (MIN) for the efficient treatment of periodontitis. In brief, MIN was prepared into PLGA microspheres with tunable release behavior using different species of PLGA, respectively. The optimally selected PLGA microspheres (LA:GA with 50:50, 10 kDa, and carboxyl group) had a drug loading of 16.91%, an in vitro release of approximately 30 days, which also had a particle size of approximately 11.8 µm with a smooth appearance and a rounded morphology. The DSC and XRD results showed that the MIN was completely encapsulated in the microspheres as an amorphous state. Cytotoxicity tests demonstrated the safety and biocompatibility of the microspheres (cell viabilities at a concentration of 1-200 μg/mL were greater than 97%), and in vitro bacterial inhibition tests showed that the selected microspheres could achieve effective bacterial inhibition at the initial stage after administration. The favorable anti-inflammatory (low TNF-α and IL-10 levels) and bone restoration effects (BV/TV: 71.8869%; BMD: 0.9782 g/cm3; TB.Th: 0.1366 mm; Tb.N: 6.9318 mm-1; Tb.Sp: 0.0735 mm) were achieved in a SD rat periodontitis model after administering once a week for four weeks. The MIN-loaded PLGA microspheres were proved to be an efficient and safe treatment for periodontitis by procedural antibacterial, anti-inflammatory, and bone restoration.
Collapse
Affiliation(s)
- Jiansong Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Ying Wei
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Jian Xiong
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Hongbing Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Gaoshuai Lv
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Jingyi Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
20
|
A multiple controlled-release hydrophilicity minocycline hydrochloride delivery system for the efficient treatment of periodontitis. Int J Pharm 2023; 636:122802. [PMID: 36894039 DOI: 10.1016/j.ijpharm.2023.122802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/04/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
The complexity of periodontitis, including the complex formation mechanisms and the complex periodontium physiological environment, as well as the complex association with multiple complications, often results in poor therapy effects. Herein, we aimed to design a nanosystem with a controlled release of minocycline hydrochloride (MH) and good retention to effectively treat periodontitis by inhibiting inflammation and repairing the alveolar bone. Firstly, insoluble ion-pairing (IIP) complexes were constructed to improve the encapsulation efficiency of hydrophilic MH in PLGA nanoparticles. Then, a nanogenerator was constructed and combined with a double emulsion method to encapsulate the complexes into PLGA nanoparticles (MH-NPs). The average particle size of MH-NPs was about 100 nm as observed by AFM and TEM, and the drug loading and encapsulation efficiency were 9.59% and 95.58%, respectively. Finally, a multifunctional system (MH-NPs-in-gels) was prepared by dispersing MH-NPs into thermosensitive gels, which could continue to release drug for 21 days in vitro. And the release mechanism showed that this controlled release behavior for MH was influenced by the insoluble ion-pairing complex, PLGA nanoparticles, and gels. In addition, the periodontitis rat model was established to investigate the pharmacodynamic effects. After 4 weeks of treatment, changes in the alveolar bone were assessed by Micro-CT (BV/TV: 70.88%; BMD: 0.97 g/cm3; TB.Th: 0.14 mm; Tb.N: 6.39 mm-1; Tb.Sp: 0.07 mm). The mechanism of MH-NPs-in-gels in vivo was clarified by the analysis of pharmacodynamic results, which showed that insoluble ion-pairing complexes with the aid of PLGA nanoparticles and gels achieved significant anti-inflammatory effects and bone repair capabilities. In conclusion, the multiple controlled-release hydrophilicity MH delivery system would have good prospects for the effective treatment of periodontitis.
Collapse
|
21
|
Wang M, Ge RL, Zhang F, Yu DG, Liu ZP, Li X, Shen H, Williams GR. Electrospun fibers with blank surface and inner drug gradient for improving sustained release. BIOMATERIALS ADVANCES 2023; 150:213404. [PMID: 37060792 DOI: 10.1016/j.bioadv.2023.213404] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/19/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
New engineering methods and advanced strategies are highly desired for creating novel drug sustained release nanomaterials. In this study, a trilayer concentric spinneret was explored to implement several multifluid electrospinning processes. A trilayer core-shell nanofiber was successfully fabricated, which comprise a drug-free polymeric coating and an inner drug gradient distribution, and then compared with bilayer core-shell and monolithic medicated nanofibers. All the electrospun nanofibers similarly consisted of two components (guest drug acetaminophen and host polymer cellulose acetate) and presented a linear morphology. Due to the secondary interactions within nanofibers, loaded drug with amorphous state was detected, as demonstrated by SEM, DSC, XRD, and FTIR determinations. In vitro and in vivo gavage treatments to rats tests were carried out, the trilayer nanofiber with an elaborate structure design were demonstrated to provide better drug sustained release profile than the bilayer core-shell nanofibers in term of initial burst release, later tail-off release and long sustained release time period. The synergistic mechanism for improving the drug sustained release behaviors is disclosed. By breaking the traditional concepts about the implementation of multifluid electrospinning and the strategy of combining surface properties and inner structural characteristics, the present protocols open a new way for developing material processing methods and generating novel functional nanomaterials.
Collapse
|
22
|
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: 9] [Impact Index Per Article: 4.5] [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
|
23
|
Functional biomaterials for comprehensive periodontitis therapy. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
24
|
Zhao P, Chen W, Feng Z, Liu Y, Liu P, Xie Y, Yu DG. Electrospun Nanofibers for Periodontal Treatment: A Recent Progress. Int J Nanomedicine 2022; 17:4137-4162. [PMID: 36118177 PMCID: PMC9480606 DOI: 10.2147/ijn.s370340] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/31/2022] [Indexed: 12/11/2022] Open
Abstract
Periodontitis is a major threat to oral health, prompting scientists to continuously study new treatment techniques. The nanofibrous membrane prepared via electrospinning has a large specific surface area and high porosity. On the one hand, electrospun nanofibers can improve the absorption capacity of proteins and promote the expression of specific genes. On the other hand, they can improve cell adhesion properties and prevent fibroblasts from passing through the barrier membrane. Therefore, electrospinning has unique advantages in periodontal treatment. At present, many oral nanofibrous membranes with antibacterial, anti-inflammatory, and tissue regeneration properties have been prepared for periodontal treatment. First, this paper introduces the electrospinning process. Then, the commonly used polymers of electrospun nanofibrous membranes for treating periodontitis are summarized. Finally, different types of nanofibrous membranes prepared via electrospinning for periodontal treatment are presented, and the future evolution of electrospinning to treat periodontitis is described.
Collapse
Affiliation(s)
- Ping Zhao
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
| | - Wei Chen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
| | - Zhangbin Feng
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
| | - Yukang Liu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
| | - Ping Liu
- The Base of Achievement Transformation, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200433, People's Republic of China.,Institute of Orthopaedic Basic and Clinical Transformation, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Yufeng Xie
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, People's Republic of China.,Shanghai Engineering Technology Research Center for High-Performance Medical Device Materials, Shanghai, 200093, People's Republic of China
| |
Collapse
|
25
|
Petrescu N, Crisan B, Aghiorghiesei O, Sarosi C, Mirica IC, Lucaciu O, Iușan SAL, Dirzu N, Apostu D. Gradual Drug Release Membranes and Films Used for the Treatment of Periodontal Disease. MEMBRANES 2022; 12:895. [PMID: 36135916 PMCID: PMC9503414 DOI: 10.3390/membranes12090895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 06/16/2023]
Abstract
Periodontitis is an inflammatory disease that, if not treated, can cause a lot of harm to the oral cavity, to the patients' quality of life, and to the entire community. There is no predictable standardized treatment for periodontitis, but there have been many attempts, using antibiotics, tissue regeneration techniques, dental scaling, or root planning. Due to the limits of the above-mentioned treatment, the future seems to be local drug delivery systems, which could gradually release antibiotics and tissue regeneration inducers at the same time. Local gradual release of antibiotics proved to be more efficient than systemic administration. In this review, we have made a literature search to identify the articles related to this topic and to find out which carriers have been tested for drug release as an adjuvant in the treatment of periodontitis. Considering the inclusion and exclusion criteria, 12 articles were chosen to be part of this review. The selected articles indicated that the drug-releasing carriers in periodontitis treatment were membranes and films fabricated from different types of materials and through various methods. Some of the drugs released by the films and membranes in the selected articles include doxycycline, tetracycline, metronidazole, levofloxacin, and minocycline, all used with good outcome regarding their bactericide effect; BMP-2, Zinc-hydroxyapatite nanoparticles with regenerative effect. The conclusion derived from the selected studies was that gradual drug release in the periodontal pockets is a promising strategy as an adjuvant for the treatment of periodontal disease.
Collapse
Affiliation(s)
- Nausica Petrescu
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Bogdan Crisan
- Department of Maxillofacial Surgery and Oral Implantology, Iuliu Hatieganu University of Medicine and Pharmacy, 400029 Cluj-Napoca, Romania
| | - Ovidiu Aghiorghiesei
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Codruta Sarosi
- Institute of Chemistry Raluca Ripan, Department of Polymer Composites, Babes-Bolyai University, 400294 Cluj-Napoca, Romania
| | - Ioana Codruta Mirica
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ondine Lucaciu
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | | | - Noemi Dirzu
- Medfuture Research Center for Advanced Medicine, School of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Dragos Apostu
- Department of Orthopaedics and Traumatology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
26
|
Shetty K, Bhandari A, Yadav KS. Nanoparticles incorporated in nanofibers using electrospinning: A novel nano-in-nano delivery system. J Control Release 2022; 350:421-434. [PMID: 36002053 DOI: 10.1016/j.jconrel.2022.08.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/26/2022]
Abstract
Nanofibers are cutting-edge drug delivery systems that are being utilised to treat a variety of ailments. Nanofibers are mostly woven by electrospinning techniques that are majorly used in drug delivery, wound dressing, tissue engineering, sensors, etc. They have several limitations that can be addressed by developing nano-in-nano delivery techniques. Nanoparticles are incorporated into nanofibers in these nano-in-nano systems. They offer a lot of benefits over other nanosystems, including the ability to shield drugs from physical deterioration, the ability to provide prolonged drug release, high surface area to volume ratio, increased drug loading capacity and the potential to be employed in critical conditions such as cancer. These nanoparticles can be encapsulated, entrapped, or adsorbed onto nanofibers in a variety of ways. To include nanosystems into nanofibers, a variety of materials and different kinds of nanoparticles can be used. The present review gives an insight to the applications of nano - in - nano drug delivery system for different diseases/disorders. The review also brings forward the current state of these novel delivery systems along with future perspectives.
Collapse
Affiliation(s)
- Karishma Shetty
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai 400056, India
| | - Ayush Bhandari
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai 400056, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai 400056, India.
| |
Collapse
|
27
|
Progress in Polymeric Micelles for Drug Delivery Applications. Pharmaceutics 2022; 14:pharmaceutics14081636. [PMID: 36015262 PMCID: PMC9412594 DOI: 10.3390/pharmaceutics14081636] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 11/17/2022] Open
Abstract
Polymeric micelles (PMs) have made significant progress in drug delivery applications. A robust core-shell structure, kinetic stability and the inherent ability to solubilize hydrophobic drugs are the highlights of PMs. This review presents the recent advances and understandings of PMs with a focus on the latest drug delivery applications. The types, methods of preparation and characterization of PMs are described along with their applications in oral, parenteral, transdermal, intranasal and other drug delivery systems. The applications of PMs for tumor-targeted delivery have been provided special attention. The safety, quality and stability of PMs in relation to drug delivery are also provided. In addition, advanced polymeric systems and special PMs are also reviewed. The in vitro and in vivo stability assessment of PMs and recent understandings in this area are provided. The patented PMs and clinical trials on PMs for drug delivery applications are considered indicators of their tremendous future applications. Overall, PMs can help overcome many unresolved issues in drug delivery.
Collapse
|
28
|
Venugopal D, Vishwakarma S, Kaur I, Samavedi S. Electrospun fiber-based strategies for controlling early innate immune cell responses: Towards immunomodulatory mesh designs that facilitate robust tissue repair. Acta Biomater 2022; 163:228-247. [PMID: 35675893 DOI: 10.1016/j.actbio.2022.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 12/16/2022]
Abstract
Electrospun fibrous meshes are widely used for tissue repair due to their ability to guide a host of cell responses including phenotypic differentiation and tissue maturation. A critical factor determining the eventual biological outcomes of mesh-based regeneration strategies is the early innate immune response following implantation. The natural healing process involves a sequence of tightly regulated, temporally varying and delicately balanced pro-/anti-inflammatory events which together promote mesh integration with host tissue. Matrix designs that do not account for the immune milieu can result in dysregulation, chronic inflammation and fibrous capsule formation, thus obliterating potential therapeutic outcomes. In this review, we provide systematic insights into the effects of specific fiber/mesh properties and mechanical stimulation on the responses of early innate immune modulators viz., neutrophils, monocytes and macrophages. We identify matrix characteristics that promote anti-inflammatory immune phenotypes, and we correlate such responses with pro-regenerative in vivo outcomes. We also discuss recent advances in 3D fabrication technologies, bioactive functionalization approaches and biomimetic/bioinspired immunomodulatory mesh design strategies for tissue repair and wound healing. The mechanobiological insights and immunoregulatory strategies discussed herein can help improve the translational outcomes of fiber-based regeneration and may also be leveraged for intervention in degenerative diseases associated with dysfunctional immune responses. STATEMENT OF SIGNIFICANCE: The crucial role played by immune cells in promoting biomaterial-based tissue regeneration is being increasingly recognized. In this review focusing on the interactions of innate immune cells (primarily neutrophils, monocytes and macrophages) with electrospun fibrous meshes, we systematically elucidate the effects of the fiber microenvironment and mechanical stimulation on biological responses, and build upon these insights to inform the rational design of immunomodulatory meshes for effective tissue repair. We discuss state-of-the-art fabrication methods and mechanobiological advances that permit the orchestration of temporally controlled phenotypic switches in immune cells during different phases of healing. The design strategies discussed herein can also be leveraged to target several complex autoimmune and inflammatory diseases.
Collapse
|
29
|
Liu X, Wang C, Pang L, Pan L, Zhang Q. Combination of resolvin E1 and lipoxin A4 promotes the resolution of pulpitis by inhibiting NF-κB activation through upregulating sirtuin 7 in dental pulp fibroblasts. Cell Prolif 2022; 55:e13227. [PMID: 35411569 PMCID: PMC9136498 DOI: 10.1111/cpr.13227] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/24/2022] [Accepted: 03/15/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives To determine whether the combination of resolvin E1 (RvE1) and lipoxin A4 (LXA4) could promote resolution of pulpitis and to investigate the mechanism. Materials and Methods Preliminary screening was first conducted in four specialized pro‐resolving mediators (SPMs). Real‐time quantitative polymerase chain reaction, western blotting, enzyme‐linked immunosorbent assay and double‐immunofluorescence labelling were employed to assess the expression of RelA, SIRT1, SIRT6, SIRT7 and pro‐inflammatory factors. Dental pulp fibroblasts (DPFs) were transfected with siRNA to assess the biological role of SIRT7. A pulpitis model was utilized to evaluate the in vivo curative effect. Results Preliminary results showed that RvE1 and LXA4 reduced the expression of RelA more markedly than other two SPMs. Both RvE1 and LXA4 treatment downregulated nuclear factor kappa B (NF‐κB) activation and increased the expression of SIRT1, SIRT6 and SIRT7, more so in combination than alone. Double‐immunofluorescence labelling showed that SIRT7 co‐localized with p‐p65 and Ac‐p65 in the nucleus. Inhibiting ChemR23 and ALX reversed the expression of RelA mRNA, p‐p65 and Ac‐p65 proteins, pro‐inflammatory factors, SIRT1, SIRT6 and SIRT7. Silencing SIRT7 significantly increased p‐p65 and Ac‐p65 protein levels and decreased SIRT1 and SIRT6 expression. In vivo experiments showed that combined administration of RvE1 and LXA4 promoted pulpitis markedly to resolution. Conclusions Combination of RvE1 and LXA4 effectively inhibited NF‐κB activation by upregulating SIRT7 expression in DPFs, leading to reduced production of pro‐inflammatory factors and promotion of pulpitis resolution.
Collapse
Affiliation(s)
- Xiaochen Liu
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Chunmeng Wang
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Liping Pang
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Liangliang Pan
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Qi Zhang
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
30
|
Costa SM, Fangueiro R, Ferreira DP. Drug Delivery Systems for Photodynamic Therapy: The Potentiality and Versatility of Electrospun Nanofibers. Macromol Biosci 2022; 22:e2100512. [PMID: 35247227 DOI: 10.1002/mabi.202100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Indexed: 11/07/2022]
Abstract
Recently, photodynamic therapy (PDT) has become a promising approach for the treatment of a broad range of diseases, including oncological and infectious diseases. This minimally invasive and localized therapy is based on the production of reactive oxygen species (ROS) able to destroy cancer cells and inactivate pathogens by combining the use of photosensitizers (PSs), light and molecular oxygen. To overcome the drawbacks of drug systemic administration, drug delivery systems (DDS) can be used to carrier the PSs, allowing higher therapeutic efficacy and minimal toxicological effects. Polymeric nanofibers produced by electrospinning emerged as powerful platforms for drug delivery applications. Electrospun nanofibers exhibit outstanding characteristics, such as large surface area to volume ratio associated with high drug loading, high porosity, flexibility, ability to incorporate and release a wide variety of therapeutic agents, biocompatibility and biodegradability. Due to the versatility of this technique, fibers with different morphologies and functionalities, including drug release profile can be produced. The possibility of scalability makes electrospinning even more attractive for the development of DDS. This review aims to explore and show an up to date of the huge potential of electrospun nanofibers as DDS for different PDT applications and discuss the opportunities and challenges in this field. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sofia M Costa
- Centre for Textile Science and Technology (2C2T), University of Minho, Guimarães, 4800-058, Portugal
| | - Raul Fangueiro
- Centre for Textile Science and Technology (2C2T), University of Minho, Guimarães, 4800-058, Portugal.,Department of Mechanical Engineering, University of Minho, Guimarães, 4800-058, Portugal
| | - Diana P Ferreira
- Centre for Textile Science and Technology (2C2T), University of Minho, Guimarães, 4800-058, Portugal
| |
Collapse
|
31
|
Dang X, Zhang H, Lin L, Li P, Ren L, Zhang W, Song R. The anti-inflammatory and osteogenic activity of chitosan/polyvinyl alcohol/graphene oxide/astaxanthin nanofibers membranes in vitro study. J Biomater Appl 2022; 36:1873-1881. [PMID: 35227102 DOI: 10.1177/08853282221076229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anti-inflammation and bone regeneration are the two major goals of periodontal therapy. We have demonstrated that chitosan (CS)/polyvinyl alcohol (PVA)/graphene oxide (GO)/astaxanthin (ASTA) nanofibers membranes prepared by electrospinning had favorable micro-morphology, good mechanical properties, and no cytotoxicity. In this study, CS/PVA/GO/ASTA nanofibers membranes were prepared to modulate both inflammatory response and osteogenic induction in vitro study. When the nanofibers membranes were co-cultured with RAW264.7 cells, glycoprotein nonmetastatic melanoma protein in the cells was highly expressed and RAW264.7 cells were polarized to M2 phenotype at the same time. In addition, following stimulation with nanofibers membranes, the messenger RNA (mRNA) and protein levels of Osteocalcin (OCN) and Runx2 in Bone marrow mesenchymal stem cells (BMSCs) were highly expressed. Taken together, these results suggested CS/PVA/GO/ASTA nanofibers membranes may promote the dissipation of inflammation and stimulate the differentiation of BMSCs into osteoblasts.
Collapse
Affiliation(s)
- Xuan Dang
- First Affiliated Hospital of Harbin Medical University, 74559College of Stomatology, Harbin Medical University, Harbin, China
| | - Hengfang Zhang
- First Affiliated Hospital of Harbin Medical University, 74559College of Stomatology, Harbin Medical University, Harbin, China
| | - Lexun Lin
- Department of pathogenic biology teaching center, Basic Medical College, 34707Harbin Medical University, Harbin, China
| | - Pie Li
- First Affiliated Hospital of Harbin Medical University, 74559College of Stomatology, Harbin Medical University, Harbin, China
| | - Liping Ren
- First Affiliated Hospital of Harbin Medical University, 74559College of Stomatology, Harbin Medical University, Harbin, China
| | - Wei Zhang
- nstitute of Materials Science and Engineering, 47820Northeast Forestry University, Harbin, China
| | - Rong Song
- First Affiliated Hospital of Harbin Medical University, 74559College of Stomatology, Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
Silk sericin/PLGA electrospun scaffolds with anti-inflammatory drug-eluting properties for periodontal tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112723. [DOI: 10.1016/j.msec.2022.112723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/22/2022] [Accepted: 02/15/2022] [Indexed: 12/13/2022]
|
33
|
Wang Y, Liu Y, Zhang X, Liu N, Yu X, Gao M, Wang W, Wu T. Engineering Electrospun Nanofibers for the Treatment of Oral Diseases. Front Chem 2022; 9:797523. [PMID: 34988063 PMCID: PMC8721107 DOI: 10.3389/fchem.2021.797523] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
With the increase of consumption of high-sugar foods, beverages, tobacco, and alcohol, the incidence rate of oral diseases has been increasing year by year. Statistics showed that the prevalence of oral diseases such as dental caries, dental pulpal disease, and periodontal disease has reached as high as 97% in 2015 in China. It is thus urgent to develop functional materials or products for the treatment of oral diseases. Electrospinning has been a widely used technology that is capable of utilizing polymer solution to generate micro/nano fibers under an appropriate high voltage condition. Owing to their excellent structures and biological performances, materials prepared by electrospinning technology have been used for a wide range of oral-related applications, such as tissue restoration, controlled drug release, anti-cancer, etc. In this regard, this article reviews the application and progress of electrospun nanofibers to various oral diseases in recent years. Firstly, engineering strategies of a variety of nanofiber structures together with their resultant functions will be introduced. Then, biological functions of electrospun nanofibers as well as their applications in the treatment of oral diseases are summarized and demonstrated. Finally, the development viewpoint of functional nanofibers is prospected, which is expected to lay the foundation and propose the direction for further clinical application.
Collapse
Affiliation(s)
- Yuanfei Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yingnan Liu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Xiaopei Zhang
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China.,Qingdao Medical College, Qingdao University, Qingdao, China
| | - Na Liu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China.,Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xixi Yu
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Meihua Gao
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Wanchun Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Tong Wu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China.,Qingdao Medical College, Qingdao University, Qingdao, China.,Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
34
|
Sharma D, Mathur VP, Satapathy BK. Biodegradable and Biocompatible 3D Constructs for Dental Applications: Manufacturing Options and Perspectives. Ann Biomed Eng 2021; 49:2030-2056. [PMID: 34318403 DOI: 10.1007/s10439-021-02839-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Designing 3D constructs with appropriate materials and structural frameworks for complex dental restorative/regenerative procedures has always remained a multi-criteria optimization challenge. In this regard, 3D printing has long been known to be a potent tool for various tissue regenerative applications, however, the preparation of biocompatible, biodegradable, and stable inks is yet to be explored and revolutionized for overall performance improvisation. The review reports the currently employed manufacturing processes for the development of engineered self-supporting, easily processable, and cost-effective 3D constructs with target-specific tuneable mechanics, bioactivity, and degradability aspects in the oral cavity for their potential use in numerous dental applications ranging from soft pulp tissues to hard alveolar bone tissues. A hybrid synergistic approach, comprising of development of multi-layered, structurally stable, composite building blocks with desired physicomechanical performance and bioactivity presents an optimal solution to circumvent the major limitations and develop new-age advanced dental restorations and implants. Further, the review summarizes some manufacturing perspectives which may inspire the readers to design appropriate structures for clinical trials so as to pave the way for their routine applications in dentistry in the near future.
Collapse
Affiliation(s)
- Deepika Sharma
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Vijay Prakash Mathur
- Division of Pedodontics and Preventive Dentistry, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Bhabani K Satapathy
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
35
|
Bioinspired peptide adhesion on Ti implants alleviates wear particle-induced inflammation and improves interfacial osteogenesis. J Colloid Interface Sci 2021; 605:410-424. [PMID: 34332414 DOI: 10.1016/j.jcis.2021.07.079] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/02/2021] [Accepted: 07/14/2021] [Indexed: 01/18/2023]
Abstract
In the inflammatory peri-implant microenvironment, excessive polarization of macrophages to the proinflammatory M1 phenotype can trigger the secretion of inflammatory cytokines, which promote bone resorption and impede osteogenesis around implants. The direct consequence of this process is the failure of prosthetic implants due to aseptic loosening. To reverse the inflammatory microenvironment and prevent prosthesis loosening, a mussel adhesion-inspired surface strategy was used for bioengineering of titanium implants with integrin-binding ability. In our design, a mussel-inspired catecholic peptide with tetravalent 3,4-dihydroxy-l-phenylalanine (DOPA) and Arg-Gly-Asp (RGD) sequences was synthesized. The peptide can easily anchor to the surface of medical titanium materials through a mussel adhesive mechanism. We found that peptide-decorated titanium implants could effectively inhibit peri-implant inflammation in a wear particle model and could promote the polarization of macrophages to a pro-healing M2 phenotype by interfering with integrin-α2β1 and integrin-αvβ3. Moreover, the peptide coating increased the adherence of osteoblasts and promoted osteogenesis on titanium implants even under inflammatory conditions. This work suggested that this biomimetic catecholic integrin-binding peptide can provide facile tactics for surface bioengineering of medical prostheses with improved interfacial osteogenesis under inflammatory conditions, which might contribute greatly to the prevention of prosthesis loosening and the improvement of clinical outcomes.
Collapse
|
36
|
Wang XS, Yang JM, Ding RJ, Liu XZ, Jiang XB, Yang ZJ, Ling ZM, Hu TX, Wei FX. Fabrication of a Polylactide-Glycolide/Poly-ε-Caprolactone/Dextran/Plastrum Testudinis Extract Composite Anti-Inflammation Nanofiber Membrane via Electrospinning for Annulus Fibrosus Regeneration. J Biomed Nanotechnol 2021; 17:873-888. [PMID: 34082873 DOI: 10.1166/jbn.2021.3070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tissue engineering is a promising approach for the treatment of chronic lower back pain (LBP) caused by intervertebral disc degeneration (IDD) resulting from degeneration and inflammation of annulus fibrosus (AF) tissue. However, scaffold with an anti-inflammatory effect on AF cells has not been reported. In this study, we fabricated a polylactide-glycolide (PLGA)/poly-ε-caprolactone (PCL)Zdextran (DEX) composite membrane loaded with plastrum testudinis extract (PTE), a Traditional Chinese Medicine herbal extract, via electrospinning. The membranes were characterized by mechanical measurements and scanning electron microscopy (SEM). Using an in vitro inflammation model induced by interleukin (IL)-1β, the cytocompatibility and anti-inflammatory effects of the composites were investigated by CCK-8 assay and flow cytometry. Potential regulatory mechanisms were examined by RT-qPCR and Western blotting. The results showed that the P10P8D2 (PLGA 10 g, PCL 8 g, DEX 2 g) composite nanofiber membrane exhibited the most uniform diameter distribution, best mechanical properties, a moderate degradation rate, and the best cytocompatibility characteristics. The optimal concentration of PTE was 120 µg/mL. Importantly, P10P8D2 combined with PTE exhibited anti-inflammatory and cell proliferation promotion effects. Moreover, the NF-κBB/NLRP3/IL-β signaling pathway was inactivated. Our findings suggested that the nanofiber membrane composed of P10P8D2 and PTE has anti-inflammatory and pro-proliferation effects on AF cells. It may provide an effective strategy for AF tissue regeneration.
Collapse
Affiliation(s)
- Xiao-Shuai Wang
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Jia-Ming Yang
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Ren-Jie Ding
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Xi-Zhe Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology Orthopaedic Research Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Xiao-Bing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510080, P. R. China
| | - Zhi-Jian Yang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510030, P. R. China
| | - Ze-Min Ling
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology Orthopaedic Research Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Tian-Xue Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Fu-Xin Wei
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| |
Collapse
|
37
|
Wei Y, Deng Y, Ma S, Ran M, Jia Y, Meng J, Han F, Gou J, Yin T, He H, Wang Y, Zhang Y, Tang X. Local drug delivery systems as therapeutic strategies against periodontitis: A systematic review. J Control Release 2021; 333:269-282. [PMID: 33798664 DOI: 10.1016/j.jconrel.2021.03.041] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/27/2021] [Accepted: 03/27/2021] [Indexed: 12/14/2022]
Abstract
Periodontitis is a chronic inflammation of the soft tissue surrounding and supporting the teeth, which causes periodontal structural damage, alveolar bone resorption, and even tooth loss. Its prevalence is very high, with nearly 60% of the global population affected. Hence, periodontitis is an important public health concern, and the development of effective healing treatments for oral diseases is a major target of the health sciences. Currently, the application of local drug delivery systems (LDDS) as an adjunctive therapy to scaling and root planning (SRP) in periodontitis is a promising strategy, giving higher efficacy and fewer side effects by controlling drug release. The cornerstone of successful periodontitis therapy is to select an appropriate bioactive agent and route of administration. In this context, this review highlights applications of LDDS with different properties in the treatment of periodontitis with or without systemic diseases, in order to reveal existing challenges and future research directions.
Collapse
Affiliation(s)
- Ying Wei
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Yaxin Deng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Shuting Ma
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Meixin Ran
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Yannan Jia
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao 028000, Neimenggu, China
| | - Jia Meng
- Liaoning Institute of Basic Medicine, Shenyang 110016, Liaoning, China
| | - Fei Han
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Yanjiao Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| |
Collapse
|
38
|
Lan X, Wang H, Bai J, Miao X, Lin Q, Zheng J, Ding S, Li X, Tang Y. Multidrug-loaded electrospun micro/nanofibrous membranes: Fabrication strategies, release behaviors and applications in regenerative medicine. J Control Release 2021; 330:1264-1287. [PMID: 33232749 DOI: 10.1016/j.jconrel.2020.11.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 01/02/2023]
Abstract
Electrospun micro/nanofibrous membranes (EFMs) have been widely investigated as local drug delivery systems. Multiple drugs can be simultaneously incorporated into one EFM to create synergistic effects, reduce side effects, and play their respective roles in the complex physiological processes of tissue regeneration and postoperative adhesion prevention. Due to the versatile electrospinning techniques, sustained and programmed release behaviors of multiple drugs could be achieved by modulating the structure of the EFMs and the location of the drugs. In this review, various multidrug incorporation approaches based on electrospinning are overviewed. In particular, the advantages and limitations of each drug incorporation technique, the methods to control drug release and the effect of one drug release on another are discussed. Then the applications of multidrug-loaded EFMs in regenerative medicine, including wound healing, bone regeneration, vascular tissue engineering, nerve regeneration, periodontal regeneration and adhesion prevention are comprehensively reviewed. Finally, the future perspectives and challenges in the research of multidrug-loaded EFMs are discussed.
Collapse
Affiliation(s)
- Xingzi Lan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Han Wang
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianfu Bai
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaomin Miao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Quan Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianpei Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shukai Ding
- Materials Institute of Atomic and Molecular Science, ShaanXi University of Science and Technology, Xi'an 710021, China
| | - Xiaoran Li
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, China
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China.
| |
Collapse
|
39
|
Chen J, Xu H, Xia K, Cheng S, Zhang Q. Resolvin E1 accelerates pulp repair by regulating inflammation and stimulating dentin regeneration in dental pulp stem cells. Stem Cell Res Ther 2021; 12:75. [PMID: 33482900 PMCID: PMC7821538 DOI: 10.1186/s13287-021-02141-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
Background Unresolved inflammation and tissue destruction are considered to underlie the failure of dental pulp repair. As key mediators of the injury response, dental pulp stem cells (DPSCs) play a critical role in pulp tissue repair and regeneration. Resolvin E1 (RvE1), a major dietary omega-3 polyunsaturated fatty-acid metabolite, is effective in resolving inflammation and activating wound healing. However, whether RvE1 facilitates injured pulp-tissue repair and regeneration through timely resolution of inflammation and rapid mobilization of DPSCs is unknown. Therefore, we established a pulp injury model and investigated the effects of RvE1 on DPSC-mediated inflammation resolution and injured pulp repair. Methods A pulp injury model was established using 8-week-old Sprague-Dawley rats. Animals were sacrificed on days 1, 3, 7, 14, 21, and 28 after pulp capping with a collagen sponge immersed in PBS with RvE1 or PBS. Hematoxylin-eosin and Masson’s trichrome staining, immunohistochemistry, and immunohistofluorescence were used to evaluate the prohealing properties of RvE1. hDPSCs were incubated with lipopolysaccharide (LPS) to induce an inflammatory response, and the expression of inflammatory factors after RvE1 application was measured. Effects of RvE1 on hDPSC proliferation, chemotaxis, and odontogenic differentiation were evaluated by CCK-8 assay, transwell assay, alkaline phosphatase (ALP) staining, alizarin red staining, and quantitative PCR, and possible signaling pathways were explored using western blotting. Results In vivo, RvE1 reduced the necrosis rate of damaged pulp and preserved more vital pulps, and promoted injured pulp repair and reparative dentin formation. Further, it enhanced dentin matrix protein 1 and dentin sialoprotein expression and accelerated pulp inflammation resolution by suppressing TNF-α and IL-1β expression. RvE1 enhanced the recruitment of CD146+ and CD105+ DPSCs to the damaged molar pulp mesenchyme. Isolated primary cells exhibited the mesenchymal stem cell immunophenotype and differentiation. RvE1 promoted hDPSC proliferation and chemotaxis. RvE1 significantly attenuated pro-inflammatory cytokine (TNF-α, IL-1β, and IL-6) release and enhanced ALP activity, nodule mineralization, and especially, expression of the odontogenesis-related genes DMP1, DSPP, and BSP in LPS-stimulated DPSCs. RvE1 regulated AKT, ERK, and rS6 phosphorylation in LPS-stimulated DPSCs. Conclusions RvE1 promotes pulp inflammation resolution and dentin regeneration and positively influences the proliferation, chemotaxis, and differentiation of LPS-stimulated hDPSCs. This response is, at least partially, dependent on AKT, ERK, and rS6-associated signaling in the inflammatory microenvironment. RvE1 has promising application potential in regenerative endodontics. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02141-y.
Collapse
Affiliation(s)
- Jie Chen
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Huaxing Xu
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Kun Xia
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Shuhua Cheng
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yan Chang Road, Shanghai, 200072, China
| | - Qi Zhang
- Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yan Chang Road, Shanghai, 200072, China.
| |
Collapse
|
40
|
Xu X, Ren S, Li L, Zhou Y, Peng W, Xu Y. Biodegradable engineered fiber scaffolds fabricated by electrospinning for periodontal tissue regeneration. J Biomater Appl 2020; 36:55-75. [PMID: 32842852 DOI: 10.1177/0885328220952250] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Considering the specificity of periodontium and the unique advantages of electrospinning, this technology has been used to fabricate biodegradable tissue engineering materials for functional periodontal regeneration. For better biomedical quality, a continuous technological progress of electrospinning has been performed. Based on property of materials (natural, synthetic or composites) and additive novel methods (drug loading, surface modification, structure adjustment or 3 D technique), various novel membranes and scaffolds that could not only relief inflammation but also influence the biological behaviors of cells have been fabricated to achieve more effective periodontal regeneration. This review provides an overview of the usage of electrospinning materials in treatments of periodontitis, in order to get to know the existing research situation and find treatment breakthroughs of the periodontal diseases.
Collapse
Affiliation(s)
- Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China *These authors contributed equally to this article
| | - Shuangshuang Ren
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China *These authors contributed equally to this article
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China *These authors contributed equally to this article
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China *These authors contributed equally to this article
| | - Wenzao Peng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China *These authors contributed equally to this article
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China *These authors contributed equally to this article
| |
Collapse
|