1
|
Shi X, Hu X, Jiang N, Mao J. Regenerative endodontic therapy: From laboratory bench to clinical practice. J Adv Res 2025; 72:229-263. [PMID: 38969092 DOI: 10.1016/j.jare.2024.07.001] [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: 04/16/2024] [Revised: 06/16/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Maintaining the vitality and functionality of dental pulp is paramount for tooth integrity, longevity, and homeostasis. Aiming to treat irreversible pulpitis and necrosis, there has been a paradigm shift from conventional root canal treatment towards regenerative endodontic therapy. AIM OF REVIEW This extensive and multipart review presents crucial laboratory and practical issues related to pulp-dentin complex regeneration aimed towards advancing clinical translation of regenerative endodontic therapy and enhancing human life quality. KEY SCIENTIFIC CONCEPTS OF REVIEW In this multipart review paper, we first present a panorama of emerging potential tissue engineering strategies for pulp-dentin complex regeneration from cell transplantation and cell homing perspectives, emphasizing the critical regenerative components of stem cells, biomaterials, and conducive microenvironments. Then, this review provides details about current clinically practiced pulp regenerative/reparative approaches, including direct pulp capping and root revascularization, with a specific focus on the remaining hurdles and bright prospects in developing such therapies. Next, special attention was devoted to discussing the innovative biomimetic perspectives opened in establishing functional tissues by employing exosomes and cell aggregates, which will benefit the clinical translation of dental pulp engineering protocols. Finally, we summarize careful consideration that should be given to basic research and clinical applications of regenerative endodontics. In particular, this review article highlights significant challenges associated with residual infection and inflammation and identifies future insightful directions in creating antibacterial and immunomodulatory microenvironments so that clinicians and researchers can comprehensively understand crucial clinical aspects of regenerative endodontic procedures.
Collapse
Affiliation(s)
- Xin Shi
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiaohan Hu
- Outpatient Department Office, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Nan Jiang
- Central Laboratory, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Jing Mao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| |
Collapse
|
2
|
Zhuang Y, Wu D, Zhou L, Liu B, Zhao X, Yang J, Liu W, Wang Z, Zheng Y, Shi X. Electrospun Biomimetic Periosteum Promotes Diabetic Bone Defect Regeneration through Regulating Macrophage Polarization and Sequential Drug Release. ACS Biomater Sci Eng 2025; 11:1690-1704. [PMID: 39908041 DOI: 10.1021/acsbiomaterials.4c02095] [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: 02/06/2025]
Abstract
The inadequate vascularization and abnormal immune microenvironment in the diabetic bone defect region present a significant challenge to osteogenic regulation. Inspired by the distinctive characteristics of healing staged in diabetic bone defects and the structure-function relationship in the natural periosteum, we fabricated an electrospun bilayer biomimetic periosteum (Bilayer@E) to promote regeneration of diabetic bone defects. Here, the inner layer of biomimetic periosteum was fabricated using coaxial electrospinning fibers, with a shell incorporating zinc oxide nanoparticles (ZnO NPs) and a core containing silicon dioxide nanoparticles (SiO2 NPs) mimicking the cambium of periosteum; the outer layer consisted of randomly aligned electrospun fibers loaded with deferoxamine (DFO), simulating the fibrous layer of periosteum; finally, epigallocatechin-3-gallate (EGCG) was coated onto the bilayer membrane to obtain Bilayer@E. The presence of EGCG on the Bilayer@E surface efficiently triggers a phenotypic transition in macrophages, shifting them from an M1 proinflammatory state to an M2 anti-inflammatory state. Moreover, the sequential release of ZnO NPs, DFO, and SiO2 NPs exhibits antimicrobial characteristics while coordinating angiogenesis and promoting osteogenic mineralization in cells. Importantly, the biomimetic periosteum shows strong in vivo bone tissue and periosteal regeneration properties in diabetic rats. The integration of sequential drug release and immunomodulation, tailored to meet the specific healing requirements during bone regeneration, offers new insights for advancing the application of biomaterials in this field.
Collapse
Affiliation(s)
- Yu Zhuang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Dingwei Wu
- Department of Orthopedics, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, China
| | - Lvyang Zhou
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Boyuan Liu
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Xingkai Zhao
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Wenge Liu
- Department of Orthopedics, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, China
| | - Zhenyu Wang
- Department of Orthopedics, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou 350001, China
| | - Yunquan Zheng
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| |
Collapse
|
3
|
Tian Z, Chen H, Zhao P. Compliant immune response of silk-based biomaterials broadens application in wound treatment. Front Pharmacol 2025; 16:1548837. [PMID: 40012629 PMCID: PMC11861559 DOI: 10.3389/fphar.2025.1548837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025] Open
Abstract
The unique properties of sericin and silk fibroin (SF) favor their widespread application in biopharmaceuticals, particularly in wound treatment and bone repair. The immune response directly influences wound healing cycle, and the extensive immunomodulatory functions of silk-based nanoparticles and hydrogels have attracted wide attention. However, different silk-processing methods may trigger intense immune system resistance after implantation into the body. In this review, we elaborate on the inflammation and immune responses caused by the implantation of sericin and SF and also explore their anti-inflammatory properties and immune regulatory functions. More importantly, we describe the latest research progress in enhancing the immunotherapeutic and anti-inflammatory effects of composite materials prepared from silk from a mechanistic perspective. This review will provide a useful reference for using the correct processes to exploit silk-based biomaterials in different wound treatments.
Collapse
Affiliation(s)
- Zhiqiang Tian
- Biological Science Research Center, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Southwest University, Chongqing, China
| | - Hong Chen
- Department of Orthopedics, 903 Hospital of Joint Logistic Support Force of The People’s Liberation Army, Hangzhou, China
| | - Ping Zhao
- Biological Science Research Center, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Southwest University, Chongqing, China
| |
Collapse
|
4
|
Zhou X, Chen S, Pich A, He C. Advanced Bioresponsive Drug Delivery Systems for Promoting Diabetic Vascularized Bone Regeneration. ACS Biomater Sci Eng 2025; 11:182-207. [PMID: 39666445 DOI: 10.1021/acsbiomaterials.4c02037] [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: 12/14/2024]
Abstract
The treatment of bone defects in diabetes mellitus (DM) patients remains a major challenge since the diabetic microenvironments significantly impede bone regeneration. Many abnormal factors including hyperglycemia, elevated oxidative stress, increased inflammation, imbalanced osteoimmune, and impaired vascular system in the diabetic microenvironment will result in a high rate of impaired, delayed, or even nonhealing events of bone tissue. Stimuli-responsive biomaterials that can respond to endogenous biochemical signals have emerged as effective therapeutic systems to treat diabetic bone defects via the combination of microenvironmental regulation and enhanced osteogenic capacity. Following the natural bone healing processes, coupling of angiogenesis and osteogenesis by advanced bioresponsive drug delivery systems has proved to be of significant approach for promoting bone repair in DM. In this Review, we have systematically summarized the mechanisms and therapeutic strategies of DM-induced impaired bone healing, outlined the bioresponsive design for drug delivery systems, and highlighted the vascularization strategies for promoting bone regeneration. Accordingly, we then overview the recent advances in developing bioresponsive drug delivery systems to facilitate diabetic vascularized bone regeneration by remodeling the microenvironment and modulating multiple regenerative cues. Furthermore, we discuss the development of adaptable drug delivery systems with unique features for guiding DM-associated bone regeneration in the future.
Collapse
Affiliation(s)
- Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Shuo Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Andrij Pich
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
5
|
Sovar A, Patrick MD, Annamalai RT. Substrate curvature influences cytoskeletal rearrangement and modulates macrophage phenotype. Front Immunol 2025; 15:1478464. [PMID: 39835126 PMCID: PMC11743265 DOI: 10.3389/fimmu.2024.1478464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Inflammation is a vital immune response, tightly orchestrated through both biochemical and biophysical cues. Dysregulated inflammation contributes to chronic diseases, highlighting the need for novel therapies that modulate immune responses with minimal side effects. While several biochemical pathways of inflammation are well understood, the influence of physical properties such as substrate curvature on immune cell behavior remains underexplored. This study investigates how substrate curvature impacts macrophage cytoskeletal dynamics, gene expression, and immunophenotype through mechanosensitive pathways. Methods Gelatin-based microgels with tunable surface curvatures were fabricated via water-in-oil emulsification and crosslinked with genipin. Microgels were sorted into three size ranges, yielding high (40-50 µm), intermediate (150-250 µm), and low (350-400 µm) curvature profiles. Macrophages were seeded onto these microgels, and cytoskeletal dynamics were examined using confocal microscopy, SEM, and actin-specific staining. Gene expression of pro- and anti-inflammatory markers was quantified using qPCR. The role of actin polymerization was assessed using Latrunculin-A (Lat-A) treatment. Results Macrophages adhered effectively to both high- and low-curvature microgels, displaying curvature-dependent morphological changes. Confocal imaging revealed that macrophages on low-curvature microgels exhibited significantly higher F-actin density than those on high-curvature microgels. Correspondingly, qPCR analysis showed upregulation of pro-inflammatory markers (e.g., Tnf, Nos2) in high-curvature conditions, while anti-inflammatory markers (e.g., Arg1) were elevated in low-curvature conditions. Lat-A treatment reduced F-actin density and modulated gene expression patterns, confirming the cytoskeletal regulation of macrophage phenotype. Discussion These findings demonstrate that substrate curvature influences macrophage behavior by modulating cytoskeletal dynamics and associated immunophenotypic markers through actin-mediated transcriptional pathways. By controlling curvature, therapeutic biomaterials may direct immune responses, offering a new avenue for treating inflammatory diseases. This mechanobiological approach presents a promising strategy for precision immunomodulation in regenerative medicine.
Collapse
Affiliation(s)
- Austin Sovar
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, United States
| | - Matthew D. Patrick
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY, United States
| | - Ramkumar T. Annamalai
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
6
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
7
|
Hu Z, Lin H, Wang Z, Yi Y, Zou S, Liu H, Han X, Rong X. 3D Printing Hierarchical Porous Nanofibrous Scaffold for Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405406. [PMID: 39548932 DOI: 10.1002/smll.202405406] [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: 07/01/2024] [Revised: 09/16/2024] [Indexed: 11/18/2024]
Abstract
Current limitations in 3D printing pose significant challenges for the fabrication of hierarchical 3D scaffolds with nanofibrous structures that simulate the natural bone extracellular matrix (ECM) for enhanced bone regeneration. This study presents an innovative approach to 3D printing customized hierarchical porous scaffolds with nanofiber structures using biodegradable nanofibrous microspheres as the bio-ink. In vitro investigations demonstrate that the hierarchical porous architecture substantially enhances cell infiltration and proliferation rates, while the nanofiber topology provides physical cues to guide osteogenic differentiation and ECM deposition. When serving as a cell carrier, the 3D-printed nanofibrous scaffold promotes bone tissue regeneration and integration in vivo. Additionally, the facile and versatile chemical modification facilitates the precise tailoring of the scaffold's functionality. Using nanofibrous microspheres with highly biomimetic and versatile modification properties as the foundational constituent in this universal 3D printing methodology enables comprehensive manipulation of scaffolding biological properties, spanning from macroscopic external morphology to molecular-scale biochemical kinetics, thereby addressing a diverse spectrum of clinical requisites.
Collapse
Affiliation(s)
- Zhiai Hu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hengyi Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yating Yi
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hao Liu
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xin Rong
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
8
|
Song P, Zhou D, Wang F, Li G, Bai L, Su J. Programmable biomaterials for bone regeneration. Mater Today Bio 2024; 29:101296. [PMID: 39469314 PMCID: PMC11513843 DOI: 10.1016/j.mtbio.2024.101296] [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: 07/12/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Programmable biomaterials are distinguished by their ability to adjust properties and functions on demand, in a periodic, reversible, or sequential manner. This contrasts with traditional biomaterials, which undergo irreversible, uncontrolled changes. This review synthesizes key advances in programmable biomaterials, examining their design principles, functionalities and applications in bone regeneration. It charts the transition from traditional to programmable biomaterials, emphasizing their enhanced precision, safety and control, which are critical from clinical and biosafety standpoints. We then classify programmable biomaterials into six types: dynamic nucleic acid-based biomaterials, electrically responsive biomaterials, bioactive scaffolds with programmable properties, nanomaterials for targeted bone regeneration, surface-engineered implants for sequential regeneration and stimuli-responsive release materials. Each category is analyzed for its structural properties and its impact on bone tissue engineering. Finally, the review further concludes by highlighting the challenges faced by programmable biomaterials and suggests integrating artificial intelligence and precision medicine to enhance their application in bone regeneration and other biomedical fields.
Collapse
Affiliation(s)
- Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Dongyang Zhou
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghaizhongye Hospital, Shanghai, 200941, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
9
|
Dobrzyńska‐Mizera M, Dodda JM, Liu X, Knitter M, Oosterbeek RN, Salinas P, Pozo E, Ferreira AM, Sadiku ER. Engineering of Bioresorbable Polymers for Tissue Engineering and Drug Delivery Applications. Adv Healthc Mater 2024; 13:e2401674. [PMID: 39233521 PMCID: PMC11616265 DOI: 10.1002/adhm.202401674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/15/2024] [Indexed: 09/06/2024]
Abstract
Herein, the recent advances in the development of resorbable polymeric-based biomaterials, their geometrical forms, resorption mechanisms, and their capabilities in various biomedical applications are critically reviewed. A comprehensive discussion of the engineering approaches for the fabrication of polymeric resorbable scaffolds for tissue engineering, drug delivery, surgical, cardiological, aesthetical, dental and cardiovascular applications, are also explained. Furthermore, to understand the internal structures of resorbable scaffolds, representative studies of their evaluation by medical imaging techniques, e.g., cardiac computer tomography, are succinctly highlighted. This approach provides crucial clinical insights which help to improve the materials' suitable and viable characteristics for them to meet the highly restrictive medical requirements. Finally, the aspects of the legal regulations and the associated challenges in translating research into desirable clinical and marketable materials of polymeric-based formulations, are presented.
Collapse
Affiliation(s)
| | - Jagan Mohan Dodda
- New Technologies – Research Centre (NTC)University of West BohemiaUniverzitní 8Pilsen30100Czech Republic
| | - Xiaohua Liu
- Chemical and Biomedical Engineering DepartmentUniversity of Missouri1030 Hill StreetColumbiaMissouri65211USA
| | - Monika Knitter
- Institute of Materials TechnologyPolymer DivisionPoznan University of TechnologyPoznanPoland
| | - Reece N. Oosterbeek
- Department of Engineering ScienceUniversity of OxfordParks RoadOxfordOX1 3PJUK
| | - Pablo Salinas
- Department of CardiologyHospital Clínico San CarlosMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Eduardo Pozo
- Department of CardiologyHospital Clínico San CarlosMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Ana Marina Ferreira
- School of EngineeringNewcastle UniversityNewcastle upon TyneNewcastleNE1 7RUUK
| | - Emmanuel Rotimi Sadiku
- Tshwane University of TechnologyDepartment of ChemicalMetallurgical and Materials EngineeringPolymer Division & Institute for Nano Engineering Research (INER)Pretoria West CampusPretoriaSouth Africa
| |
Collapse
|
10
|
Ming Y, He X, Zhao Z, Meng X, Zhu Y, Tan H, Yang G, Hu Y, Zheng L. Nanocarrier-Assisted Delivery of Berberine Promotes Diabetic Alveolar Bone Regeneration by Scavenging ROS and Improving Mitochondrial Dysfunction. Int J Nanomedicine 2024; 19:10263-10282. [PMID: 39399826 PMCID: PMC11471107 DOI: 10.2147/ijn.s475320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/15/2024] [Indexed: 10/15/2024] Open
Abstract
Purpose Oxidative stress and mitochondrial dysfunction are potential contributors to the compromised tissue regeneration capacity of alveolar bone in diabetic patients. Berberine, an active plant alkaloid, exhibits multiple pharmacological effects including antioxidation, blood glucose- and blood lipid-lowering properties. However, it remains uncertain whether berberine can improve impaired osteogenesis in type 2 diabetes mellitus (T2DM), and its poor solubility and oral bioavailability also constrain its applications in bone regeneration. Thus, our study aimed to probe the effects of berberine on bone marrow stem cells (BMSCs) in a diabetic microenvironment, with a greater emphasis on developing a suitable nano-delivery system for berberine and assessing its capability to repair diabetic alveolar bone defects. Methods Firstly, BMSCs were exposed to berberine within a high glucose and palmitate (HG+PA) environment. Reactive oxygen species levels, mitochondrial membrane potential, ATP generation, cell apoptosis, and osteogenic potential were subsequently assessed. Next, we explored the regulatory mechanism of autophagy flux in the positive effects of berberine. Furthermore, a nanocarrier based on emulsion electrospinning for sustained local delivery of berberine (Ber@SF/PCL) was established. We assessed its capacity to enhance bone healing in the alveolar bone defect of T2DM rats through micro-computed tomography and histology analysis. Results Berberine treatment could inhibit reactive oxygen species overproduction, mitochondrial dysfunction, apoptosis, and improve osteogenesis differentiation by restoring autophagy flux under HG+PA conditions. Notably, Ber@SF/PCL electrospun nanofibrous membrane with excellent physicochemical properties and good biological safety had the potential to promote alveolar bone remodeling in T2DM rats. Conclusion Our study shed new lights into the protective role of berberine on BMSCs under T2DM microenvironment. Furthermore, berberine-loaded composite electrospun membrane may serve as a promising approach for regenerating alveolar bone in diabetic patients.
Collapse
Affiliation(s)
- Ye Ming
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Xinyi He
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Zhenxing Zhao
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Xuehuan Meng
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Ye Zhu
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Hao Tan
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Guoyin Yang
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Yun Hu
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Leilei Zheng
- College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| |
Collapse
|
11
|
Han J, Sheng T, Zhang Y, Cheng H, Gao J, Yu J, Gu Z. Bioresponsive Immunotherapeutic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2209778. [PMID: 36639983 DOI: 10.1002/adma.202209778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The human immune system is an interaction network of biological processes, and its dysfunction is closely associated with a wide array of diseases, such as cancer, infectious diseases, tissue damage, and autoimmune diseases. Manipulation of the immune response network in a desired and controlled fashion has been regarded as a promising strategy for maximizing immunotherapeutic efficacy and minimizing side effects. Integration of "smart" bioresponsive materials with immunoactive agents including small molecules, biomacromolecules, and cells can achieve on-demand release of agents at targeted sites to reduce overdose-related toxicity and alleviate off-target effects. This review highlights the design principles of bioresponsive immunotherapeutic materials and discusses the critical roles of controlled release of immunoactive agents from bioresponsive materials in recruiting, housing, and manipulating immune cells for evoking desired immune responses. Challenges and future directions from the perspective of clinical translation are also discussed.
Collapse
Affiliation(s)
- Jinpeng Han
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tao Sheng
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Jianqing Gao
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
12
|
Desai N, Pande S, Vora L, Kommineni N. Correction to "Nanofibrous Microspheres: A Biomimetic Platform for Bone Tissue Regeneration". ACS APPLIED BIO MATERIALS 2024; 7:6325-6331. [PMID: 39162584 PMCID: PMC11409221 DOI: 10.1021/acsabm.4c01057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Indexed: 08/21/2024]
|
13
|
Zhang M, Yan S, Wang J, Zhong Y, Wang C, Zhang T, Xing D, Shao Y. Rational design of multifunctional hydrogels targeting the microenvironment of diabetic periodontitis. Int Immunopharmacol 2024; 138:112595. [PMID: 38950455 DOI: 10.1016/j.intimp.2024.112595] [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: 03/24/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
Periodontitis is a chronic inflammatory disease and is the primary contributor to adult tooth loss. Diabetes exacerbates periodontitis, accelerates periodontal bone resorption. Thus, effectively managing periodontitis in individuals with diabetes is a long-standing challenge. This review introduces the etiology and pathogenesis of periodontitis, and analyzes the bidirectional relationship between diabetes and periodontitis. In this review, we comprehensively summarize the four pathological microenvironments influenced by diabetic periodontitis: high glucose microenvironment, bacterial infection microenvironment, inflammatory microenvironment, and bone loss microenvironment. The hydrogel design strategies and latest research development tailored to the four microenvironments of diabetic periodontitis are mainly focused on. Finally, the challenges and potential solutions in the treatment of diabetic periodontitis are discussed. We believe this review will be helpful for researchers seeking novel avenues in the treatment of diabetic periodontitis.
Collapse
Affiliation(s)
- Miao Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Saisai Yan
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Yingjie Zhong
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Tingting Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yingchun Shao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
14
|
Siverino C, Metsemakers WJ, Sutter R, Della Bella E, Morgenstern M, Barcik J, Ernst M, D'Este M, Joeris A, Chittò M, Schwarzenberg P, Stoddart M, Vanvelk N, Richards G, Wehrle E, Weisemann F, Zeiter S, Zalavras C, Varga P, Moriarty TF. Clinical management and innovation in fracture non-union. Expert Opin Biol Ther 2024; 24:973-991. [PMID: 39126182 DOI: 10.1080/14712598.2024.2391491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION With the introduction and continuous improvement in operative fracture fixation, even the most severe bone fractures can be treated with a high rate of successful healing. However, healing complications can occur and when healing fails over prolonged time, the outcome is termed a fracture non-union. Non-union is generally believed to develop due to inadequate fixation, underlying host-related factors, or infection. Despite the advancements in fracture fixation and infection management, there is still a clear need for earlier diagnosis, improved prediction of healing outcomes and innovation in the treatment of non-union. AREAS COVERED This review provides a detailed description of non-union from a clinical perspective, including the state of the art in diagnosis, treatment, and currently available biomaterials and orthobiologics.Subsequently, recent translational development from the biological, mechanical, and infection research fields are presented, including the latest in smart implants, osteoinductive materials, and in silico modeling. EXPERT OPINION The first challenge for future innovations is to refine and to identify new clinical factors for the proper definition, diagnosis, and treatment of non-union. However, integration of in vitro, in vivo, and in silico research will enable a comprehensive understanding of non-union causes and correlations, leading to the development of more effective treatments.
Collapse
Affiliation(s)
- C Siverino
- AO Research Institute Davos, Davos Platz, Switzerland
| | - W-J Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven - University of Leuven, Leuven, Belgium
| | - R Sutter
- Radiology Department, Balgrist University Hospital, University of Zürich, Zürich, Switzerland
| | - E Della Bella
- AO Research Institute Davos, Davos Platz, Switzerland
| | - M Morgenstern
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | - J Barcik
- AO Research Institute Davos, Davos Platz, Switzerland
| | - M Ernst
- AO Research Institute Davos, Davos Platz, Switzerland
| | - M D'Este
- AO Research Institute Davos, Davos Platz, Switzerland
| | - A Joeris
- AO Innovation Translation Center, Davos Platz, Switzerland
| | - M Chittò
- AO Research Institute Davos, Davos Platz, Switzerland
| | | | - M Stoddart
- AO Research Institute Davos, Davos Platz, Switzerland
| | - N Vanvelk
- Trauma Research Unit, Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - G Richards
- AO Research Institute Davos, Davos Platz, Switzerland
| | - E Wehrle
- AO Research Institute Davos, Davos Platz, Switzerland
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - F Weisemann
- Department of Trauma Surgery, BG Unfallklinik Murnau, Murnau am Staffelsee, Germany
| | - S Zeiter
- AO Research Institute Davos, Davos Platz, Switzerland
| | - C Zalavras
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - P Varga
- AO Research Institute Davos, Davos Platz, Switzerland
| | - T F Moriarty
- AO Research Institute Davos, Davos Platz, Switzerland
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
15
|
Xia Y, Chen Z, Zheng Z, Chen H, Chen Y. Nanomaterial-integrated injectable hydrogels for craniofacial bone reconstruction. J Nanobiotechnology 2024; 22:525. [PMID: 39217329 PMCID: PMC11365286 DOI: 10.1186/s12951-024-02801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The complex anatomy and biology of craniofacial bones pose difficulties in their effective and precise reconstruction. Injectable hydrogels (IHs) with water-swollen networks are emerging as a shape-adaptive alternative for noninvasively rebuilding craniofacial bones. The advent of versatile nanomaterials (NMs) customizes IHs with strengthened mechanical properties and therapeutically favorable performance, presenting excellent contenders over traditional substitutes. Structurally, NM-reinforced IHs are energy dissipative and covalently crosslinked, providing the mechanics necessary to support craniofacial structures and physiological functions. Biofunctionally, incorporating unique NMs into IH expands a plethora of biological activities, including immunomodulatory, osteogenic, angiogenic, and antibacterial effects, further favoring controllable dynamic tissue regeneration. Mechanistically, NM-engineered IHs optimize the physical traits to direct cell responses, regulate intracellular signaling pathways, and control the release of biomolecules, collectively bestowing structure-induced features and multifunctionality. By encompassing state-of-the-art advances in NM-integrated IHs, this review offers a foundation for future clinical translation of craniofacial bone reconstruction.
Collapse
Affiliation(s)
- Yong Xia
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zihan Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zebin Zheng
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Huimin Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yuming Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
16
|
Shakya A, Li Y, Chang NW, Liu X. Supra-Alveolar Bone Regeneration: Progress, Challenges, and Future Perspectives. COMPOSITES. PART B, ENGINEERING 2024; 283:111673. [PMID: 39071449 PMCID: PMC11270636 DOI: 10.1016/j.compositesb.2024.111673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Periodontitis is a highly prevalent disease that damages the supporting tissues of a tooth, including the alveolar bone. Alveolar bone loss owing to periodontitis is broadly categorized as supra-alveolar and intra-alveolar bone loss. In intra-alveolar bone loss, the defect has an angular or oblique orientation to the long axis of the tooth in an apical direction. In contrast, the defect is perpendicular to the long axis of the tooth in supra-alveolar bone loss. Unlike intra-alveolar bone defects, supra-alveolar bone defects lack supporting adjacent space, which makes supra-alveolar bone regeneration more challenging. In addition, the limited availability of resources in terms of vascularity and underlying tissues is another obstacle to supra-alveolar bone regeneration. Currently, supra-alveolar bone loss is the least predictable periodontal defect type in regenerative periodontal therapy. In addition, supra-alveolar bone loss is much more common than other alveolar bone loss. Despite its prevalence, research on supra-alveolar bone regeneration remains sparse, indicating an unmet need for significant research efforts in this area. This review summarize recent advances, obstacles, and future directions in the field of supra-alveolar bone regeneration. We discuss the biomaterials, bioactive molecules, and cells that have been tested for supra-alveolar bone regeneration, followed by pre-clinical and clinical approaches employed in this field. Additionally, we highlight obstacles and present future directions that will propel supra-alveolar bone research forward.
Collapse
Affiliation(s)
- Ajay Shakya
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246
| | - Yingzi Li
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246
- Chemical and Biomedical Engineering Department, University of Missouri, Columbia, MO 65211
| | - Nai-wen Chang
- Department of Periodontology, Texas A&M University School of Dentistry, Dallas, TX 75246
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX 75246
- Chemical and Biomedical Engineering Department, University of Missouri, Columbia, MO 65211
| |
Collapse
|
17
|
Sovar A, Patrick M, Annamalai RT. Substrate Curvature Influences Cytoskeletal Rearrangement and Modulates Macrophage Phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607593. [PMID: 39211272 PMCID: PMC11361075 DOI: 10.1101/2024.08.12.607593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Inflammation serves as a critical defense mechanism against pathogens and tissue damage but can lead to chronic diseases, such as cardiovascular disease and diabetes, when dysregulated. Macrophages play a pivotal role in orchestrating inflammatory responses, transitioning from pro-inflammatory M1 to anti-inflammatory M2 phenotypes to resolve inflammation and promote tissue repair. Current approaches to modulate macrophage phenotype predominantly rely on biochemical cues, which may induce systemic side effects. Given the mechanosensitivity of macrophages, this study investigates biophysical cues, specifically substrate curvature, as a localized strategy to regulate macrophage phenotype and minimize systemic repercussions. We hypothesized that substrate curvature influences macrophage immunophenotype by modulating F-actin polymerization. To test this hypothesis, we fabricated spherical microgels with tunable curvatures and characterized their biophysical properties. Our findings indicate that macrophages adhere to microgel surfaces irrespective of curvature, but the curvature significantly alters F-actin dynamics. Furthermore, manipulating cytoskeletal dynamics via selective actin inhibition partially reversed curvature-induced changes in macrophage phenotype. These results underscore the pivotal role of substrate curvature in modulating macrophage behavior and immunophenotype. Overall, our study demonstrates that substrate curvature significantly influences macrophage cytoskeletal dynamics and resulting immunophenotype. This simple approach can be utilized as a localized immunomodulatory treatment for inflammatory diseases.
Collapse
|
18
|
Yang Y, He X, Zhao Z, Yi J. Macrophage-Centric Biomaterials for Bone Regeneration in Diabetes Mellitus: Contemporary Advancements, Challenges, and Future Trajectories. Cureus 2024; 16:e66621. [PMID: 39258053 PMCID: PMC11386247 DOI: 10.7759/cureus.66621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 09/12/2024] Open
Abstract
Increased susceptibility to bone fragility and the diminution of bone regenerative capacity are recognized as significant and frequent sequelae of diabetes mellitus. Research has elucidated the pivotal role of macrophages in the pathogenesis and repair of diabetic bone defects. Notwithstanding this, the therapeutic efficacy of traditional interventions remains predominantly inadequate. Concomitant with substantial advancements in tissue engineering in recent epochs, there has been an escalation in the development of biomaterials designed to modulate macrophage activity, thereby augmenting osseous tissue regeneration in the context of hyperglycemia. This review amalgamates insights from extant research and delineates recent progressions in the domain of biomaterials that target macrophages for the regeneration of diabetic bone, whilst also addressing the clinical challenges and envisaging future directions within this field.
Collapse
Affiliation(s)
- Yiyan Yang
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| | - Xiaoli He
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| | - Zhihe Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| | - Jianru Yi
- Department of Orthodontics, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, CHN
| |
Collapse
|
19
|
Lin H, Weng E, Rong X, Yu L, Chen Y, Jiang Y, Hu H, Wang Z, Zou S, Hu Z. ECM-Mimicking Strontium-Doped Nanofibrous Microspheres for Periodontal Tissue Regeneration in Osteoporosis. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39042857 DOI: 10.1021/acsami.4c06286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Regenerating periodontal defects in osteoporosis patients presents a significant clinical challenge. Unlike the relatively straightforward regeneration of homogeneous bone tissue, periodontal regeneration requires the intricate reconstruction of the cementum-periodontal ligament-alveolar bone interface. Strontium (Sr)-doped biomaterials have been extensively utilized in bone tissue engineering due to their remarkable pro-osteogenic attributes. However, their application in periodontal tissue regeneration has been scarcely explored. In this study, we synthesized an innovative injectable Sr-BGN/GNM scaffold by integrating Sr-doped bioactive glass nanospheres (Sr-BGNs) into the nanofiber architecture of gelatin nanofiber microspheres (GNMs). This design, mimicking the natural bone extracellular matrix (ECM), enhanced the scaffold's mechanical properties and effectively controlled the sustained release of Sr ions (Sr2+), thereby promoting the proliferation, osteogenic differentiation, and ECM secretion of PDLSCs and BMSCs, as well as enhancing vascularization in endothelial cells. In vivo experiments further indicated that the Sr-BGNs/GNMs significantly promoted osteogenesis and angiogenesis. Moreover, the scaffold's tunable degradation kinetics optimized the prolonged release and pro-regenerative effects of Sr2+ in vivo, matching the pace of periodontal regeneration and thereby facilitating the regeneration of functional periodontal tissues under osteoporotic conditions. Therefore, Sr-BGNs/GNMs emerge as a promising candidate for advancing periodontal regeneration strategies.
Collapse
Affiliation(s)
- Hengyi Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Enhuai Weng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xin Rong
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Yu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiling Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yukun Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haikun Hu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhiai Hu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
20
|
Cao X, Luo B, Mu Y, Wang C, Lu R, Yao Y, Chen S. The regulatory effect of TiO 2 nanotubes loaded with graphene oxide on macrophage polarization in an inflammatory environment. BMC Oral Health 2024; 24:824. [PMID: 39033148 PMCID: PMC11265100 DOI: 10.1186/s12903-024-04608-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Excessive inflammation is a major cause of implant failure. The surface morphology, hydrophilicity, and loading of biomaterials are major properties modulating anti-inflammatory macrophage activation. This paper investigates the regulatory effects of modifying the surface of Titanium dioxide nanotubes (TNTs) with graphene oxide (GO) on the polarization of mouse monocyte macrophages (RAW264.7). METHODS TNT was produced by the anodic oxidation of titanium. GO was subsequently electrodeposited on the TNT to obtain a TNT-GO composite. The samples were characterised through scanning electron microscopy (SEM), Raman spectroscopy, and X-ray diffraction. RAW264.7 cells were separately seeded onto the surface of three groups of samples: pure Ti, TNT, and TNT-GO. Under the condition of lipopolysaccharide stimulation, the influence of the sample surfaces on the gene expression profiles was investigated through RNA sequence analysis. In addition, cell spreading was observed through SEM, cell adhesion and proliferation were analysed using the CCK8 assay, and the expression of inflammation-related factors was investigated by ELISA and cellular immunofluorescence staining. The production of reactive oxygen species (ROS) in the RAW264.7 cells on the surface of the three groups was detected via immunofluorescence staining. RESULTS The CCK8 results indicated that the adhesion and proliferation of the RAW264.7 cells were reduced on the TNT and TNT-GO surfaces. ELISA results revealed significant differences in the pro-inflammatory factors tumour necrosis factor-α and interleukin-6 secretion among the three groups at 24 h (p < 0.05). The secretion of pro-inflammatory factors significantly reduced and the expression of anti-inflammatory factor IL-10 increased on the TNT and TNT-GO surfaces. The RNA sequencing, ELISA, and cell immunofluorescence staining test results suggested that the inflammatory response of M1 polarization was reduced and the M2 polarization of macrophages was induced on the TNT-GO surface, which may be attributed to the reduction in ROS production. CONCLUSIONS Under lipopolysaccharide stimulation, the inflammatory response of the RAW264.7 cells was reduced and the M2 polarization of macrophages was promoted on the TNT-GO surface, which may be caused by the reduced ROS production. Consequently, the designed TNT-GO material is promising for implants owing to its excellent inflammation regulation ability.
Collapse
Affiliation(s)
- Xu Cao
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Bin Luo
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yanting Mu
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Caiyun Wang
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Ran Lu
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Yao Yao
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Su Chen
- Laboratory of Biomaterials and Biomechanics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
21
|
Desai N, Pande S, Vora LK, Kommineni N. Nanofibrous Microspheres: A Biomimetic Platform for Bone Tissue Regeneration. ACS APPLIED BIO MATERIALS 2024; 7:4270-4292. [PMID: 38950103 PMCID: PMC11253102 DOI: 10.1021/acsabm.4c00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Bone, a fundamental constituent of the human body, is a vital scaffold for support, protection, and locomotion, underscoring its pivotal role in maintaining skeletal integrity and overall functionality. However, factors such as trauma, disease, or aging can compromise bone structure, necessitating effective strategies for regeneration. Traditional approaches often lack biomimetic environments conducive to efficient tissue repair. Nanofibrous microspheres (NFMS) present a promising biomimetic platform for bone regeneration by mimicking the native extracellular matrix architecture. Through optimized fabrication techniques and the incorporation of active biomolecular components, NFMS can precisely replicate the nanostructure and biochemical cues essential for osteogenesis promotion. Furthermore, NFMS exhibit versatile properties, including tunable morphology, mechanical strength, and controlled release kinetics, augmenting their suitability for tailored bone tissue engineering applications. NFMS enhance cell recruitment, attachment, and proliferation, while promoting osteogenic differentiation and mineralization, thereby accelerating bone healing. This review highlights the pivotal role of NFMS in bone tissue engineering, elucidating their design principles and key attributes. By examining recent preclinical applications, we assess their current clinical status and discuss critical considerations for potential clinical translation. This review offers crucial insights for researchers at the intersection of biomaterials and tissue engineering, highlighting developments in this expanding field.
Collapse
Affiliation(s)
- Nimeet Desai
- Department
of Biomedical Engineering, Indian Institute
of Technology Hyderabad, Kandi 502285, India
| | - Shreya Pande
- Department
of Biomedical Engineering, Indian Institute
of Technology Hyderabad, Kandi 502285, India
| | - Lalitkumar K. Vora
- School
of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Nagavendra Kommineni
- Center
for Biomedical Research, Population Council, New York, New York 10065, United States
| |
Collapse
|
22
|
Li Q, Ma C, Jing Y, Liu X. Multifunctional Nanofibrous Hollow Microspheres for Enhanced Periodontal Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402335. [PMID: 38757666 PMCID: PMC11267322 DOI: 10.1002/advs.202402335] [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: 03/04/2024] [Revised: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Destructive periodontitis destroys alveolar bone and eventually leads to tooth loss. While guided bone regeneration, which is based on creating a physical barrier to hinder the infiltration of epithelial and connective tissues into defect sites, has been widely used for alveolar bone regeneration, its outcomes remain variable. In this work, a multifunctional nanofibrous hollow microsphere (NFHMS) is developed for enhanced alveolar bone regeneration. The NFHMS is first prepared via combining a double emulsification and a thermally induced phase separation process. Next, E7, a short peptide with high specific affinity to bone marrow-derived stem cells (BMSCs), is conjugated onto the surface of NFHMS. After that, bone forming peptide (BFP), a short peptide derived from bone morphology protein 7 is loaded in calcium phosphate (CaP) nanoparticles, which are further encapsulated in the hollow space of the NFHMS-E7 to form NFHMS-E7-CaP/BFP. The NFHMS-E7-CaP/BFP selectively promoted the adhesion of BMSCs and expelled the adhesion of fibroblasts and epithelial cells. In addition, the BFP is sustainedly released from the NFHMS-E7-CaP/BFP to enhance the osteogenesis of BMSCs. A rat challenging fenestration defect model showed that the NFHMS-E7-CaP/BFP significantly enhanced alveolar bone tissue regeneration. This work provides a novel bioengineering approach for guided bone regeneration.
Collapse
Affiliation(s)
- Qian Li
- Department of Biomedical SciencesTexas A&M University School of DentistryDallasTX75246USA
- Chemical and Biomedical Engineering DepartmentUniversity of MissouriColumbiaMO65211USA
| | - Chi Ma
- Department of Biomedical SciencesTexas A&M University School of DentistryDallasTX75246USA
- Center of Excellence in HipScottish Rite for ChildrenDallasTX75219USA
- Department of Orthopedic SurgeryUniversity of Texas Southwestern Medical CenterDallasTX75390USA
| | - Yan Jing
- Department of OrthodonticsTexas A&M University School of DentistryDallasTX75246USA
| | - Xiaohua Liu
- Department of Biomedical SciencesTexas A&M University School of DentistryDallasTX75246USA
- Chemical and Biomedical Engineering DepartmentUniversity of MissouriColumbiaMO65211USA
| |
Collapse
|
23
|
Wang X, Xiang C, Huang C, Cheng H, Zhou Z, Zhang J, Xie H. The treatment efficacy of bone tissue engineering strategy for repairing segmental bone defects under diabetic condition. Front Bioeng Biotechnol 2024; 12:1379679. [PMID: 38737542 PMCID: PMC11082311 DOI: 10.3389/fbioe.2024.1379679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Background Diabetes mellitus is a systematic disease which exert detrimental effect on bone tissue. The repair and reconstruction of bone defects in diabetic patients still remain a major clinical challenge. This study aims to investigate the potential of bone tissue engineering approach to improve bone regeneration under diabetic condition. Methods In the present study, decalcified bone matrix (DBM) scaffolds were seeded with allogenic fetal bone marrow-derived mesenchymal stem cells (BMSCs) and cultured in osteogenic induction medium to fabricate BMSC/DBM constructs. Then the BMSC/DBM constructs were implanted in both subcutaneous pouches and large femoral bone defects in diabetic (BMSC/DBM in DM group) and non-diabetic rats (BMSC/DBM in non-DM group), cell-free DBM scaffolds were implanted in diabetic rats to serve as the control group (DBM in DM group). X-ray, micro-CT and histological analyses were carried out to evaluate the bone regenerative potential of BMSC/DBM constructs under diabetic condition. Results In the rat subcutaneous implantation model, quantitative micro-CT analysis demonstrated that BMSC/DBM in DM group showed impaired bone regeneration activity compared with the BMSC/DBM in non-DM group (bone volume: 46 ± 4.4 mm3 vs 58.9 ± 7.15 mm3, *p < 0.05). In the rat femoral defect model, X-ray examination demonstrated that bone union was delayed in BMSC/DBM in DM group compared with BMSC/DBM in non-DM group. However, quantitative micro-CT analysis showed that after 6 months of implantation, there was no significant difference in bone volume and bone density between the BMSC/DBM in DM group (199 ± 63 mm3 and 593 ± 65 mg HA/ccm) and the BMSC/DBM in non-DM group (211 ± 39 mm3 and 608 ± 53 mg HA/ccm). Our data suggested that BMSC/DBM constructs could repair large bone defects in diabetic rats, but with delayed healing process compared with non-diabetic rats. Conclusion Our study suggest that biomaterial sacffolds seeded with allogenic fetal BMSCs represent a promising strategy to induce and improve bone regeneration under diabetic condition.
Collapse
Affiliation(s)
- Xiangsheng Wang
- Department of Plastic Surgery, Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Hubei, China
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Can Xiang
- Department of Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunhua Huang
- Department of Plastic Surgery, Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Hubei, China
| | - Hanxiao Cheng
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhentao Zhou
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jufang Zhang
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Xie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
24
|
Wang B, Huang Y, Cai Q, Du Z, Li X. Biomaterials for diabetic bone repair: Influencing mechanisms, multi-aspect progress and future prospects. COMPOSITES PART B: ENGINEERING 2024; 274:111282. [DOI: 10.1016/j.compositesb.2024.111282] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
25
|
Song D, Wang C, Liang J, Jia T. Effect and mechanism of fluctuant glucose on restraining implant osseointegration in diabetes. Oral Dis 2024; 30:1583-1590. [PMID: 37338083 DOI: 10.1111/odi.14600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 06/21/2023]
Abstract
OBJECTIVES The objectives of the study was to discuss the effect and mechanism of fluctuant glucose (FG) on implant osseointegration in type 2 diabetic mellitus (T2DM). MATERIALS AND METHODS Rats were divided into control, T2DM and FG group, and the implants were inserted into their femurs. Micro-CT and histological analysis were used to evaluate the effect on osseointegration in vivo. And we investigated the effect of different conditions (normal, control, high glucose, and FG medium) on rat osteoblast in vitro. Then transmission electron microscope (TEM) and Western blot were used to evaluate the endoplasmic reticulum stress (ERS) response. Finally, 4-PBA, an inhibitor of ERS, was added into different conditions to observe the functions of osteoblast. RESULTS In vivo, Micro-CT and histological analysis showed that the percentage of osseointegration in FG rats were lower than other two group. In vitro, the results demonstrated that the adhesion of the cells becomes worst, and osteogenic ability was also severely impaired in FG group. In addition, FG could induce more serious ERS and 4-PBA could improve the dysfunction of osteoblasts induced by FG. CONCLUSION Fluctuant glucose could restrain the implant osseointegration in T2DM, and the effect was more obvious than consistent high glucose by a possible mechanism of activation ERS pathway.
Collapse
Affiliation(s)
- Dawei Song
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chenchen Wang
- Department of Oral Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jin Liang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tingting Jia
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
26
|
He Y, Jiang H, Dong S. Bioactives and Biomaterial Construction for Modulating Osteoclast Activities. Adv Healthc Mater 2024; 13:e2302807. [PMID: 38009952 DOI: 10.1002/adhm.202302807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/28/2023] [Indexed: 11/29/2023]
Abstract
Bone tissue constitutes 15-20% of human body weight and plays a crucial role in supporting the body, coordinating movement, regulating mineral homeostasis, and hematopoiesis. The maintenance of bone homeostasis relies on a delicate balance between osteoblasts and osteoclasts. Osteoclasts, as the exclusive "bone resorbers" in the human skeletal system, are of paramount significance yet often receive inadequate attention. When osteoclast activity becomes excessive, it frequently leads to various bone metabolic disorders, subsequently resulting in secondary bone injuries, such as fractures. This not only reduces life quality of patients, but also imposes a significant economic burden on society. In response to the pressing need for biomaterials in the treatment of osteoclast dysregulation, there is a surge of research and investigations aimed at osteoclast regulation. Promising progress is achieved in this domain. This review seeks to provide a comprehensive understanding of how to modulate osteoclast activities. It summarizes bioactive substances that influence osteoclasts and elucidates strategies for constructing related biomaterial systems. It offers practical insights and ideas for the development and application of biomaterials and tissue engineering, with the hope of guiding the clinical treatment of osteoclast-related bone diseases using biomaterials in the future.
Collapse
Affiliation(s)
- Yuwei He
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Hong Jiang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, 400038, P. R. China
| |
Collapse
|
27
|
Valamvanos TF, Dereka X, Katifelis H, Gazouli M, Lagopati N. Recent Advances in Scaffolds for Guided Bone Regeneration. Biomimetics (Basel) 2024; 9:153. [PMID: 38534838 PMCID: PMC10968314 DOI: 10.3390/biomimetics9030153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
The rehabilitation of alveolar bone defects of moderate to severe size is often challenging. Currently, the therapeutic approaches used include, among others, the guided bone regeneration technique combined with various bone grafts. Although these techniques are widely applied, several limitations and complications have been reported such as morbidity, suboptimal graft/membrane resorption rate, low structural integrity, and dimensional stability. Thus, the development of biomimetic scaffolds with tailor-made characteristics that can modulate cell and tissue interaction may be a promising tool. This article presents a critical consideration in scaffold's design and development while also providing information on various fabrication methods of these nanosystems. Their utilization as delivery systems will also be mentioned.
Collapse
Affiliation(s)
- Theodoros-Filippos Valamvanos
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Xanthippi Dereka
- Department of Periodontology, School of Dentistry, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Hector Katifelis
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- School of Science and Technology, Hellenic Open University, 26335 Patra, Greece
| | - Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Greece Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
28
|
Ozkendir O, Karaca I, Cullu S, Erdoğan OC, Yaşar HN, Dikici S, Owen R, Aldemir Dikici B. Engineering periodontal tissue interfaces using multiphasic scaffolds and membranes for guided bone and tissue regeneration. BIOMATERIALS ADVANCES 2024; 157:213732. [PMID: 38134730 DOI: 10.1016/j.bioadv.2023.213732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Periodontal diseases are one of the greatest healthcare burdens worldwide. The periodontal tissue compartment is an anatomical tissue interface formed from the periodontal ligament, gingiva, cementum, and bone. This multifaceted composition makes tissue engineering strategies challenging to develop due to the interface of hard and soft tissues requiring multiphase scaffolds to recreate the native tissue architecture. Multilayer constructs can better mimic tissue interfaces due to the individually tuneable layers. They have different characteristics in each layer, with modulation of mechanical properties, material type, porosity, pore size, morphology, degradation properties, and drug-releasing profile all possible. The greatest challenge of multilayer constructs is to mechanically integrate consecutive layers to avoid delamination, especially when using multiple manufacturing processes. Here, we review the development of multilayer scaffolds that aim to recapitulate native periodontal tissue interfaces in terms of physical, chemical, and biological characteristics. Important properties of multiphasic biodegradable scaffolds are highlighted and summarised, with design requirements, biomaterials, and fabrication methods, as well as post-treatment and drug/growth factor incorporation discussed.
Collapse
Affiliation(s)
- Ozgu Ozkendir
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Ilayda Karaca
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Selin Cullu
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Oğul Can Erdoğan
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Hüsniye Nur Yaşar
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Serkan Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Robert Owen
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey.
| |
Collapse
|
29
|
Baldeon-Gutierrez R, Ohkura N, Yoshiba K, Yoshiba N, Tohma A, Takeuchi R, Belal RSI, Edanami N, Takahara S, Gomez-Kasimoto S, Ida T, Noiri Y. Wound-healing Processes After Pulpotomy in the Pulp Tissue of Type 1 Diabetes Mellitus Model Rats. J Endod 2024; 50:196-204. [PMID: 37939821 DOI: 10.1016/j.joen.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION Patients with type 1 diabetes mellitus (DM1) tend to have delayed wound healing, even in the pulp tissue. We hypothesized that hyperglycemia affects odontoblast-like cell (OLC) differentiation and is involved in macrophage polarization. Accordingly, we evaluated dental pulp stem cell differentiation and macrophage phenotypes after pulpotomy. METHODS After modifying DM1 rat models by streptozotocin, 8-week-old rats' upper left first molars were pulpotomized with mineral trioxide aggregate. Meanwhile, the control group was administered saline. Immunohistochemical localization of nestin, osteopontin, α-smooth muscles (α-SMAs), and CD68 (pan-macrophage marker) was conducted 7 days after pulpotomy. The OLC differentiation stage was determined using double immunofluorescence of nestin and α-SMA. Double immunofluorescence of CD68 and iNOS was counted as M1 macrophages and CD68 and CD206 as M2 macrophages. Proliferating cell nuclear antigen and Thy-1 (CD90) were evaluated by immunofluorescence. RESULTS In DM1 rats, the reparative dentin bridge was not complete; however, the osteopontin-positive area did not differ significantly from that in controls. Proliferating cell nuclear antigen, indicative of cell proliferation, increased in positive cells in DM1 rats compared with controls. Double-positive cells for α-SMA and nestin indicated many immature OLCs in DM1. CD90 was positive only in controls. CD68-positive cells, especially M1 macrophages, were increased in DM1 rats, allowing the inflammatory stage to continue 7 days after pulpotomy. CONCLUSIONS The condition of DM1 model rats can interfere at various stages of the wound healing process, altering OLC differentiation and macrophage polarization. These findings highlight the importance of normal blood glucose concentrations during pulp wound healing.
Collapse
Affiliation(s)
- Rosa Baldeon-Gutierrez
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoto Ohkura
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Kunihiko Yoshiba
- Division of Oral Science for Health Promotion, Department of Oral Health and Welfare, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Nagako Yoshiba
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Aiko Tohma
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryosuke Takeuchi
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Razi Saifullah Ibn Belal
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Edanami
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shintaro Takahara
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Susan Gomez-Kasimoto
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takako Ida
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuichiro Noiri
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
30
|
Shi Y, Tao W, Yang W, Wang L, Qiu Z, Qu X, Dang J, He J, Fan H. Calcium phosphate coating enhances osteointegration of melt electrowritten scaffold by regulating macrophage polarization. J Nanobiotechnology 2024; 22:47. [PMID: 38297240 PMCID: PMC10829397 DOI: 10.1186/s12951-024-02310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Abstract
The osteoimmune microenvironment induced by implants plays a significant role in bone regeneration. It is essential to efficiently and timely switch the macrophage phenotype from M1 to M2 for optimal bone healing. This study examined the impact of a calcium phosphate (CaP) coating on the physiochemical properties of highly ordered polycaprolactone (PCL) scaffolds fabricated using melt electrowritten (MEW). Additionally, it investigated the influence of these scaffolds on macrophage polarization and their immunomodulation on osteogenesis. The results revealed that the CaP coated PCL scaffold exhibited a rougher surface topography and higher hydrophilicity in comparison to the PCL scaffold without coating. Besides, the surface morphology of the coating and the release of Ca2+ from the CaP coating were crucial in regulating the transition of macrophages from M1 to M2 phenotypes. They might activate the PI3K/AKT and cAMP-PKA pathways, respectively, to facilitate M2 polarization. In addition, the osteoimmune microenvironment induced by CaP coated PCL could not only enhance the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) in vitro but also promote the bone regeneration in vivo. Taken together, the CaP coating can be employed to control the phenotypic switching of macrophages, thereby creating a beneficial immunomodulatory microenvironment that promotes bone regeneration.
Collapse
Affiliation(s)
- Yubo Shi
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weidong Tao
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenjing Yang
- Xijing 986 Hospital Department, The Fourth Military Medical University, Xi'an, China
| | - Lei Wang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
31
|
Guo K, Wang Y, Feng ZX, Lin XY, Wu ZR, Zhong XC, Zhuang ZM, Zhang T, Chen J, Tan WQ. Recent Development and Applications of Polydopamine in Tissue Repair and Regeneration Biomaterials. Int J Nanomedicine 2024; 19:859-881. [PMID: 38293610 PMCID: PMC10824616 DOI: 10.2147/ijn.s437854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
The various tissue damages are a severe problem to human health. The limited human tissue regenerate ability requires suitable biomaterials to help damage tissue repair and regeneration. Therefore, many researchers devoted themselves to exploring biomaterials suitable for tissue repair and regeneration. Polydopamine (PDA) as a natural and multifunctional material which is inspired by mussel has been widely applied in different biomaterials. The excellent properties of PDA, such as strong adhesion, photothermal and high drug-loaded capacity, seem to be born for tissue repair and regeneration. Furthermore, PDA combined with different materials can exert unexpected effects. Thus, to inspire researchers, this review summarizes the recent and representative development of PDA biomaterials in tissue repair and regeneration. This article focuses on why apply PDA in these biomaterials and what PDA can do in different tissue injuries.
Collapse
Affiliation(s)
- Kai Guo
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Yong Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zi-Xuan Feng
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xiao-Ying Lin
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zhang-Rui Wu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xin-Cao Zhong
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Ze-Ming Zhuang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Tao Zhang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jian Chen
- Department of Ultrasonography, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang Province, People’s Republic of China
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
32
|
Wu L, Morrow B, Hong L, Rajasingh J. Preparation of Monodispersed Nanofibrous Gelatin Microspheres Using Homebuilt Microfluidics. Methods Mol Biol 2024; 2835:325-337. [PMID: 39105928 DOI: 10.1007/978-1-0716-3995-5_28] [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: 08/07/2024]
Abstract
Gelatin, a protein derivative from collagen, is a versatile material with promising applications in tissue engineering. Among the various forms of gelatin scaffolds, nanofibrous gelatin microspheres (NFGMs) are attracting research efforts due to their fibrous nature and injectability. However, current methods for synthesizing nanofibrous gelatin microspheres (NFGMs) have limitations, such as wide size distributions and the use of toxic solvents. To address these challenges, the article introduces a novel approach. First, it describes the creation of a microfluidic device using readily available supplies. Subsequently, it outlines a unique process for producing monodispersed NFGMs through a combination of the microfluidic device and thermally induced phase separation (TIPS). This innovative method eliminates the need for sieving and the use of toxic solvents, making it a more ecofriendly and efficient alternative.
Collapse
Affiliation(s)
- Linfeng Wu
- Department of Pediatric Dentistry & Community Oral Health, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Brian Morrow
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Liang Hong
- Department of Pediatric Dentistry & Community Oral Health, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
33
|
Hu Z, Rong X, Liu X. E7-Conjugated Bio-Inspired Microspheres as a Biological Barrier for Guided Tissue Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58136-58150. [PMID: 38063848 PMCID: PMC10862379 DOI: 10.1021/acsami.3c12213] [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: 08/16/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Guided tissue regeneration (GTR), which is based on creating a physical barrier to prevent the downgrowth of epithelial and connective tissues into the defect site, has been widely used in clinical practice for periodontal regeneration for many years. However, its outcomes remain variable due to highly specific indications, the demand for proficient surgical skills, and frequent occurrence of complications. In this study, we developed a new GTR biomaterial that acts as a biological barrier for epithelial cells and fibroblasts while also serving as a scaffold for bone marrow-derived mesenchymal stem cells (BMSCs) and periodontal ligament stem cells (PDLSCs). This innovative GTR biomaterial is bioinspired injectable microspheres that are self-assembled from nanofibers, and their surfaces are conjugated with E7, a short peptide that selectively promotes BMSC and PDLSC adhesion but inhibits the attachment and spreading of epithelial cells and gingival fibroblasts. The selective affinity afforded by E7 on the surfaces of the nanofibrous microspheres facilitated the colonization of BMSCs in the periodontal defect, thereby substantially improving functional periodontal regeneration, as evidenced by enhanced new bone formation, reduced root exposure, and diminished attachment loss. The remarkable superiority of the bioinspired microspheres over conventional GTR materials in promoting periodontal regeneration underscores the potential of this innovative approach to enhance the efficacy of functional periodontal tissue regeneration.
Collapse
Affiliation(s)
- Zhiai Hu
- Department
of Biomedical Sciences, Texas A&M University
School of Dentistry, Dallas, Texas 75246, United States
| | - Xin Rong
- Department
of Biomedical Sciences, Texas A&M University
School of Dentistry, Dallas, Texas 75246, United States
| | - Xiaohua Liu
- Department
of Biomedical Sciences, Texas A&M University
School of Dentistry, Dallas, Texas 75246, United States
- Chemical
and Biomedical Engineering Department, University
of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
34
|
Chen C, Gao X, Li H, Pan X, Wang S. Intravertebral insertion of interbody fusion cage via transpedicular approach for the treatment of stage III Kümmell disease: a technical note and case presentation. Br J Neurosurg 2023; 37:1909-1914. [PMID: 33843392 DOI: 10.1080/02688697.2021.1892590] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/16/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Kümmell disease usually occurs in the elderly osteoporosis population and develops gradually into symptomatic, progressive kyphosis of the spine. However, current surgical methods to deal with stage III Kümmell disease are less satisfying. The objective of this study was to describe a less invasive technique for treating stage III Kümmell disease. TECHNIQUE A less invasive technique of intravertebral insertion of interbody fusion cage via transpedicular approach with posterior spine stabilization was applied to treat stage III Kümmell disease. RESULTS This study details a modified technique applied in a patient with stage III Kümmell disease, showing significant improvement in pain relief, anterior column height recovery, and kyphotic angle correction. And no complications were reported during our follow-up. CONCLUSIONS Intravertebral insertion of interbody fusion cage via transpedicular approach provides advantages of acceptable correction of kyphosis, bony fusion, minimal invasion. Thus, our method was a good alternative choice for stage III Kümmell disease.
Collapse
Affiliation(s)
- Changjun Chen
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan, People's Republic of China
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xianlei Gao
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan, People's Republic of China
| | - Hao Li
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan, People's Republic of China
| | - Xin Pan
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan, People's Republic of China
| | - Songgang Wang
- Department of Orthopaedics, Shandong University Qilu Hospital, Jinan, People's Republic of China
| |
Collapse
|
35
|
Sheng N, Xing F, Wang J, Zhang QY, Nie R, Li-Ling J, Duan X, Xie HQ. Recent progress in bone-repair strategies in diabetic conditions. Mater Today Bio 2023; 23:100835. [PMID: 37928253 PMCID: PMC10623372 DOI: 10.1016/j.mtbio.2023.100835] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/02/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023] Open
Abstract
Bone regeneration following trauma, tumor resection, infection, or congenital disease is challenging. Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia. It can result in complications affecting multiple systems including the musculoskeletal system. The increased number of diabetes-related fractures poses a great challenge to clinical specialties, particularly orthopedics and dentistry. Various pathological factors underlying DM may directly impair the process of bone regeneration, leading to delayed or even non-union of fractures. This review summarizes the mechanisms by which DM hampers bone regeneration, including immune abnormalities, inflammation, reactive oxygen species (ROS) accumulation, vascular system damage, insulin/insulin-like growth factor (IGF) deficiency, hyperglycemia, and the production of advanced glycation end products (AGEs). Based on published data, it also summarizes bone repair strategies in diabetic conditions, which include immune regulation, inhibition of inflammation, reduction of oxidative stress, promotion of angiogenesis, restoration of stem cell mobilization, and promotion of osteogenic differentiation, in addition to the challenges and future prospects of such approaches.
Collapse
Affiliation(s)
- Ning Sheng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jie Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Duan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| |
Collapse
|
36
|
Liu Y, Suarez-Arnedo A, Caston EL, Riley L, Schneider M, Segura T. Exploring the Role of Spatial Confinement in Immune Cell Recruitment and Regeneration of Skin Wounds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304049. [PMID: 37721722 PMCID: PMC10874253 DOI: 10.1002/adma.202304049] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/09/2023] [Indexed: 09/19/2023]
Abstract
Microporous annealed particle (MAP) scaffolds are injectable granular materials comprised of micron sized hydrogel particles (microgels). The diameter of these microgels directly determines the size of the interconnected void space between particles where infiltrating or encapsulated cells reside. This tunable porosity allows the authors to use MAP scaffolds to study the impact of spatial confinement (SC) on both cellular behaviors and the host response to biomaterials. Despite previous studies showing that pore size and SC influence cellular phenotypes, including mitigating macrophage inflammatory response, there is still a gap in knowledge regarding how SC within a biomaterial modulates immune cell recruitment in vivo in wounds and implants. Thus, the immune cell profile within confined and unconfined biomaterials is studied using small (40 µm), medium (70 µm), and large (130 µm) diameter spherical microgels, respectively. This work uncovered that MAP scaffolds impart regenerative wound healing with an IgG1-biased Th2 response. MAP scaffolds made with large microgels promote a balanced pro-regenerative macrophage response, resulting in enhanced wound healing with mature collagen regeneration and reduced inflammation levels.
Collapse
Affiliation(s)
- Yining Liu
- Department of Biomedical Engineering, Duke University, 101 Science Drive Campus Box 90281, Durham, NC 27708, USA
| | - Alejandra Suarez-Arnedo
- Department of Biomedical Engineering, Duke University, 101 Science Drive Campus Box 90281, Durham, NC 27708, USA
| | - Eleanor L.P. Caston
- Department of Biomedical Engineering, Duke University, 101 Science Drive Campus Box 90281, Durham, NC 27708, USA
| | - Lindsay Riley
- Department of Biomedical Engineering, Duke University, 101 Science Drive Campus Box 90281, Durham, NC 27708, USA
| | - Michelle Schneider
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, 101 Science Drive Campus Box 90281, Durham, NC 27708, USA
- Clinical Science Departments of Neurology and Dermatology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
37
|
Yin H, Mao K, Huang Y, Guo A, Shi L. Tendon stem/progenitor cells are promising reparative cell sources for multiple musculoskeletal injuries of concomitant articular cartilage lesions associated with ligament injuries. J Orthop Surg Res 2023; 18:869. [PMID: 37968672 PMCID: PMC10647040 DOI: 10.1186/s13018-023-04313-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/23/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Trauma-related articular cartilage lesions usually occur in conjunction with ligament injuries. Torn ligaments are frequently reconstructed with tendon autograft and has been proven to achieve satisfactory clinical outcomes. However, treatments for the concomitant articular cartilage lesions are still very insufficient. The current study was aimed to evaluate whether stem cells derived from tendon tissue can be considered as an alternative reparative cell source for cartilage repair. METHODS Primary human tendon stem/progenitor cells (hTSPCs) were isolated from 4 male patients (32 ± 8 years) who underwent ACL reconstruction surgery with autologous semitendinosus and gracilis tendons. The excessive tendon tissue after graft preparation was processed for primary cell isolation with an enzyme digestion protocol. Decellularization cartilage matrix (DCM) was used to provide a chondrogenic microenvironment for hTSPCs. Cell viability, cell morphology on the DCM, as well as their chondrogenic differentiation were evaluated. RESULTS DAPI staining and DNA quantitative analysis (61.47 μg per mg dry weight before and 2.64 μg/mg after decellularization) showed that most of the cells in the cartilage lacuna were removed after decellularization process. Whilst, the basic structure of the cartilage tissue was preserved and the main ECM components, collagen type II and sGAG were retained after decellularization, which were revealed by DMMB assay and histology. Live/dead staining and proliferative assay demonstrated that DCM supported attachment, survival and proliferation of hTSPCs with an excellent biocompatibility. Furthermore, gene expression analysis indicated that chondrogenic differentiation of hTSPC was induced by the DCM microenvironment, with upregulation of chondrogenesis-related marker genes, COL 2 and SOX9, without the use of exogenous growth factors. CONCLUSION DCM supported hTSPCs attachment and proliferation with high biocompatibility. Moreover, TSPCs underwent a distinct chondrogenesis after the induction of a chondrogenic microenvironment provided by DCM. These results indicated that TSPCs are promising reparative cell sources for promoting cartilage repair. Particularly, in the cohort that articular cartilage lesions occur in conjunction with ligament injuries, autologous TSPCs can be isolated from a portion of the tendon autograph harvested for ligaments reconstruction. In future clinical practice, combined ligament reconstruction with TSPCs- based therapy for articular cartilage repair can to be considered to achieve superior repair of these associated injuries, in which autologous TSPCs can be isolated from a portion of the tendon autograph harvested for ligaments reconstruction.
Collapse
Affiliation(s)
- Heyong Yin
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Kelei Mao
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Yufu Huang
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Ai Guo
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China.
| | - Lin Shi
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
38
|
Bai X, Cao R, Wu D, Zhang H, Yang F, Wang L. Dental Pulp Stem Cells for Bone Tissue Engineering: A Literature Review. Stem Cells Int 2023; 2023:7357179. [PMID: 37868704 PMCID: PMC10586346 DOI: 10.1155/2023/7357179] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/03/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Bone tissue engineering (BTE) is a promising approach for repairing and regenerating damaged bone tissue, using stem cells and scaffold structures. Among various stem cell sources, dental pulp stem cells (DPSCs) have emerged as a potential candidate due to their multipotential capabilities, ability to undergo osteogenic differentiation, low immunogenicity, and ease of isolation. This article reviews the biological characteristics of DPSCs, their potential for BTE, and the underlying transcription factors and signaling pathways involved in osteogenic differentiation; it also highlights the application of DPSCs in inducing scaffold tissues for bone regeneration and summarizes animal and clinical studies conducted in this field. This review demonstrates the potential of DPSC-based BTE for effective bone repair and regeneration, with implications for clinical translation.
Collapse
Affiliation(s)
- Xiaolei Bai
- Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Ruijue Cao
- Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Danni Wu
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Huicong Zhang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Fan Yang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Linhong Wang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| |
Collapse
|
39
|
Moradi L, Witek L, Vivekanand Nayak V, Cabrera Pereira A, Kim E, Good J, Liu CJ. Injectable hydrogel for sustained delivery of progranulin derivative Atsttrin in treating diabetic fracture healing. Biomaterials 2023; 301:122289. [PMID: 37639975 PMCID: PMC11232488 DOI: 10.1016/j.biomaterials.2023.122289] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/22/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Hydrogels with long-term storage stability, controllable sustained-release properties, and biocompatibility have been garnering attention as carriers for drug/growth factor delivery in tissue engineering applications. Chitosan (CS)/Graphene Oxide (GO)/Hydroxyethyl cellulose (HEC)/β-glycerol phosphate (β-GP) hydrogel is capable of forming a 3D gel network at physiological temperature (37 °C), rendering it an excellent candidate for use as an injectable biomaterial. This work focused on an injectable thermo-responsive CS/GO/HEC/β-GP hydrogel, which was designed to deliver Atsttrin, an engineered derivative of a known chondrogenic and anti-inflammatory growth factor-like molecule progranulin. The combination of the CS/GO/HEC/β-GP hydrogel and Atsttrin provides a unique biochemical and biomechanical environment to enhance fracture healing. CS/GO/HEC/β-GP hydrogels with increased amounts of GO exhibited rapid sol-gel transition, higher viscosity, and sustained release of Atsttrin. In addition, these hydrogels exhibited a porous interconnected structure. The combination of Atsttrin and hydrogel successfully promoted chondrogenesis and osteogenesis of bone marrow mesenchymal stem cells (bmMSCs) in vitro. Furthermore, the work also presented in vivo evidence that injection of Atsttrin-loaded CS/GO/HEC/β-GP hydrogel stimulated diabetic fracture healing by simultaneously inhibiting inflammatory and stimulating cartilage regeneration and endochondral bone formation signaling pathways. Collectively, the developed injectable thermo-responsive CS/GO/HEC/βG-P hydrogel yielded to be minimally invasive, as well as capable of prolonged and sustained delivery of Atsttrin, for therapeutic application in impaired fracture healing, particularly diabetic fracture healing.
Collapse
Affiliation(s)
- Lida Moradi
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA; Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Lukasz Witek
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA; Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, 11201, USA
| | - Vasudev Vivekanand Nayak
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Angel Cabrera Pereira
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Ellen Kim
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA
| | - Julia Good
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA; Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
40
|
Shen Y, Tang Q, Wang J, Zhou Z, Yin Y, Zhang Y, Zheng W, Wang X, Chen G, Sun J, Chen L. Targeting RORα in macrophages to boost diabetic bone regeneration. Cell Prolif 2023; 56:e13474. [PMID: 37051760 PMCID: PMC10542986 DOI: 10.1111/cpr.13474] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Diabetes mellitus (DM) has become a serious threat to human health. Bone regeneration deficiency and nonunion caused by DM is perceived as a worldwide epidemic, with a very high socioeconomic impact on public health. Here, we find that targeted activation of retinoic acid-related orphan receptor α (RORα) by SR1078 in the early stage of bone defect repair can significantly promote in situ bone regeneration of DM rats. Bone regeneration relies on the activation of macrophage RORα in the early bone repair, but RORα of DM rats fails to upregulation as hyperglycemic inflammatory microenvironment induced IGF1-AMPK signalling deficiency. Mechanistic investigations suggest that RORα is vital for macrophage-induced migration and proliferation of bone mesenchymal stem cells (BMSCs) via a CCL3/IL-6 depending manner. In summary, our study identifies RORα expressed in macrophages during the early stage of bone defect repair is crucial for in situ bone regeneration, and offers a novel strategy for bone regeneration therapy and fracture repair in DM patients.
Collapse
Affiliation(s)
- Yufeng Shen
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
- Department of Stomatology, The First Affiliated Hospital, School of MedicineShihezi UniversityShihezi 832000China
| | - Qingming Tang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Jiajia Wang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Zheng Zhou
- Department of Stomatology, The First Affiliated Hospital, School of MedicineShihezi UniversityShihezi 832000China
| | - Ying Yin
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Yifan Zhang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Wenhao Zheng
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Xinyuan Wang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Guangjin Chen
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Jiwei Sun
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Lili Chen
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| |
Collapse
|
41
|
Liu Y, Suarez-Arnedo A, Caston E, Riley L, Schneider M, Segura T. Exploring the Role of Spatial Confinement in Immune Cell Recruitment and Regeneration of Skin Wounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.30.538879. [PMID: 37162980 PMCID: PMC10168413 DOI: 10.1101/2023.04.30.538879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Microporous annealed particle (MAP) scaffolds are injectable granular materials comprised of micron sized hydrogel particles (microgels). The diameter of these microgels directly determines the size of the interconnected void space between particles where infiltrating or encapsulated cells reside. This tunable porosity allows us to use MAP scaffolds to study the impact of spatial confinement (SC) on both cellular behaviors and the host response to biomaterials. Despite previous studies showing that pore size and SC influence cellular phenotypes, including mitigating the macrophage inflammatory response, there is still a gap in knowledge regarding how SC within a biomaterial modulates immune cell recruitment in vivo in wounds and implants. Thus, we studied the immune cell profile within confined and unconfined biomaterials using small (40 μm), medium (70 μm), and large (130 μm) diameter spherical microgels, respectively. We discovered that MAP scaffolds imparted regenerative wound healing with an IgG1-biased Th2 response. MAP scaffolds generated from 130 μm diameter microgels have a median pore size that can accommodate ∼40 µm diameter spheres induced a more balanced pro-regenerative macrophage response and better wound healing outcomes with more mature collagen regeneration and reduced levels of inflammation.
Collapse
|
42
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
43
|
Xu T, Xie K, Wang C, Ivanovski S, Zhou Y. Immunomodulatory nanotherapeutic approaches for periodontal tissue regeneration. NANOSCALE 2023; 15:5992-6008. [PMID: 36896757 DOI: 10.1039/d2nr06149j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Periodontitis is an infection-induced inflammatory disease characterized by progressive destruction of tooth supporting tissues, which, if left untreated, can result in tooth loss. The destruction of periodontal tissues is primarily caused by an imbalance between the host immune protection and immune destruction mechanisms. The ultimate goal of periodontal therapy is to eliminate inflammation and promote the repair and regeneration of both hard and soft tissues, so as to restore the physiological structure and function of periodontium. Advancement in nanotechnologies has enabled the development of nanomaterials with immunomodulatory properties for regenerative dentistry. This review discusses the immune mechanisms of the major effector cells in the innate and adaptive immune systems, the physicochemical and biological properties of nanomaterials, and the research advancements in immunomodulatory nanotherapeutic approaches for the management of periodontitis and the regeneration of periodontal tissues. The current challenges, and prospects for future applications of nanomaterials are then discussed so that researchers at the intersections of osteoimmunology, regenerative dentistry and materiobiology will continue to advance the development of nanomaterials for improved periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tian Xu
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Kunke Xie
- Clinical Laboratory, Bo'Ai Hospital of Zhongshan, 6 Chenggui Road, East District, Zhongshan 528403, Guangdong, China
| | - Cong Wang
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Yinghong Zhou
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| |
Collapse
|
44
|
Cai F, Liu Y, Liu K, Zhao R, Chen W, Yusufu A, Liu Y. Diabetes mellitus impairs bone regeneration and biomechanics. J Orthop Surg Res 2023; 18:169. [PMID: 36872328 PMCID: PMC9987049 DOI: 10.1186/s13018-023-03644-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 02/24/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND With the rise of high-calorie diets and the aging of populations, the incidence of diabetes was increased dramatically in the world and the number of people with diabetes was predicted to rise to 600 million by 2045. Numerous studies have confirmed that several organ systems, including the skeletal system, are seriously affected by diabetes. In that study, the bone regeneration and the biomechanics of the newly regenerated bone were investigated in diabetic rats, which may provide a supplement for previous studies. METHODS A total of 40 SD rats were randomly divided into the type 2 diabetes mellitus (T2DM) group (n = 20) and the control group (n = 20). Beyond that high fat diet and streptozotocin (STZ) were jointly used in the T2DM group, there were no differences between the two groups in terms of treatment conditions. Distraction osteogenesis was used in all animals for the next experimental observation. The evaluation criterion of the regenerated bone was based on radioscopy (once a week), micro-computed tomography (CT), general morphology, biomechanics (including ultimate load, modulus of elasticity, energy to failure, and stiffness), histomorphometry (including von Kossa, Masson trichrome, Goldner trichrome, and safranin O staining), and immunohistochemistry. RESULTS All rats in the T2DM group with fasting glucose levels (FGL, > 16.7 mmol/L) were allowed to complete the following experiments. The results showed that rats with T2DM have a higher body weight (549.01 g ± 31.34 g) than rats in the control group (488.60 g ± 33.60 g) at the end of observation. Additionally, compared to the control group, slower bone regeneration in the distracted segments was observed in the T2DM group according to radiography, micro-CT, general morphology, and histomorphometry. Furthermore, a biomechanical test showed that there was a worse ultimate load (31.01 ± 3.39%), modulus of elasticity (34.44 ± 5.06%), energy to failure (27.42 ± 5.87%), and stiffness (34.55 ± 7.66%) than the control group (45.85 ± 7.61%, 54.38 ± 9.33%, 59.41 ± 10.96%, and 54.07 ± 9.30%, respectively). Furthermore, the decreased expressions of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) were presented in T2DM group by immunohistochemistry. CONCLUSION The present study demonstrated that diabetes mellitus impairs bone regeneration and biomechanics in newly regenerated bone, a phenomenon that might be related to oxidative stress and poor angiogenesis brought on by the disease.
Collapse
Affiliation(s)
- Feiyu Cai
- Department of Burns and Plastic Surgery and Wound Repair Surgery, The Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yanshi Liu
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Kai Liu
- Department of Trauma and Micro Reconstructive Surgery, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Ruomei Zhao
- Department of Burns and Plastic Surgery and Wound Repair Surgery, The Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Wenjiao Chen
- Department of Burns and Plastic Surgery and Wound Repair Surgery, The Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Aihemaitijiang Yusufu
- Department of Trauma and Micro Reconstructive Surgery, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China.
| | - Yi Liu
- Department of Burns and Plastic Surgery and Wound Repair Surgery, The Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
45
|
Wang H, Chang X, Ma Q, Sun B, Li H, Zhou J, Hu Y, Yang X, Li J, Chen X, Song J. Bioinspired drug-delivery system emulating the natural bone healing cascade for diabetic periodontal bone regeneration. Bioact Mater 2023; 21:324-339. [PMID: 36185747 PMCID: PMC9483739 DOI: 10.1016/j.bioactmat.2022.08.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 12/01/2022] Open
Abstract
Diabetes mellitus (DM) aggravates periodontitis, resulting in accelerated periodontal bone resorption. Disordered glucose metabolism in DM causes reactive oxygen species (ROS) overproduction resulting in compromised bone healing, which makes diabetic periodontal bone regeneration a major challenge. Inspired by the natural bone healing cascade, a mesoporous silica nanoparticle (MSN)-incorporated PDLLA (poly(dl-lactide))-PEG-PDLLA (PPP) thermosensitive hydrogel with stepwise cargo release is designed to emulate the mesenchymal stem cell "recruitment-osteogenesis" cascade for diabetic periodontal bone regeneration. During therapy, SDF-1 quickly escapes from the hydrogel due to diffusion for early rat bone marrow stem cell (rBMSC) recruitment. Simultaneously, slow degradation of the hydrogel starts to gradually expose the MSNs for sustained release of metformin, which can scavenge the overproduced ROS under high glucose conditions to reverse the inhibited osteogenesis of rBMSCs by reactivating the AMPK/β-catenin pathway, resulting in regulation of the diabetic microenvironment and facilitation of osteogenesis. In vitro experiments indicate that the hydrogel markedly restores the inhibited migration and osteogenic capacities of rBMSCs under high glucose conditions. In vivo results suggest that it can effectively recruit rBMSCs to the periodontal defect and significantly promote periodontal bone regeneration under type 2 DM. In conclusion, our work provides a novel therapeutic strategy of a bioinspired drug-delivery system emulating the natural bone healing cascade for diabetic periodontal bone regeneration.
Collapse
Affiliation(s)
- He Wang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Xiaowei Chang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi′an Jiaotong University, Xi′an, 710049, China
| | - Qian Ma
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Boyang Sun
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Han Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Jinmin Zhou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Yiyao Hu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Xiaoyu Yang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Jie Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi′an Jiaotong University, Xi′an, 710049, China
| | - Jinlin Song
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| |
Collapse
|
46
|
Kwan JC, Dondani J, Iyer J, Muaddi HA, Nguyen TT, Tran SD. Biomimicry and 3D-Printing of Mussel Adhesive Proteins for Regeneration of the Periodontium-A Review. Biomimetics (Basel) 2023; 8:biomimetics8010078. [PMID: 36810409 PMCID: PMC9944831 DOI: 10.3390/biomimetics8010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Innovation in the healthcare profession to solve complex human problems has always been emulated and based on solutions proven by nature. The conception of different biomimetic materials has allowed for extensive research that spans several fields, including biomechanics, material sciences, and microbiology. Due to the atypical characteristics of these biomaterials, dentistry can benefit from these applications in tissue engineering, regeneration, and replacement. This review highlights an overview of the application of different biomimetic biomaterials in dentistry and discusses the key biomaterials (hydroxyapatite, collagen, polymers) and biomimetic approaches (3D scaffolds, guided bone and tissue regeneration, bioadhesive gels) that have been researched to treat periodontal and peri-implant diseases in both natural dentition and dental implants. Following this, we focus on the recent novel application of mussel adhesive proteins (MAPs) and their appealing adhesive properties, in addition to their key chemical and structural properties that relate to the engineering, regeneration, and replacement of important anatomical structures in the periodontium, such as the periodontal ligament (PDL). We also outline the potential challenges in employing MAPs as a biomimetic biomaterial in dentistry based on the current evidence in the literature. This provides insight into the possible increased functional longevity of natural dentition that can be translated to implant dentistry in the near future. These strategies, paired with 3D printing and its clinical application in natural dentition and implant dentistry, develop the potential of a biomimetic approach to overcoming clinical problems in dentistry.
Collapse
Affiliation(s)
- Jan C. Kwan
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Jay Dondani
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Janaki Iyer
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Hasan A. Muaddi
- Department of Oral and Maxillofacial Surgery, King Khalid University, Abha 62529, Saudi Arabia
| | - Thomas T. Nguyen
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Division of Periodontics, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Correspondence:
| |
Collapse
|
47
|
Li D, Li X, Zhang J, Tang Z, Tian A. The immunomodulatory effect of IL-4 accelerates bone substitute material-mediated osteogenesis in aged rats via NLRP3 inflammasome inhibition. Front Immunol 2023; 14:1121549. [PMID: 37153554 PMCID: PMC10157059 DOI: 10.3389/fimmu.2023.1121549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Background Bone defect repair by implanting bone substitute materials has been a common clinical treatment. With the understanding of substance-immune system interactions and increasing evidence indicating that the post-implantation immune response determines the fate of bone substitute materials, active modulation of host macrophage polarization is considered a promising strategy. However, whether the same regulatory effects exist when an individual immune system is altered with aging is unclear. Methods In this study, we mechanistically investigated the effect of immunosenescence on the active regulation of macrophage polarization by establishing a cranial bone defect model in young and aged rats implanted with Bio-Oss®. Forty-eight young and 48 aged specific pathogen-free (SPF) male SD rats were randomly divided into two groups. In the experimental group, 20 μL of IL-4 (0.5 μg/mL) was injected locally on the third to seventh postoperative days, while an equal volume of PBS was injected in the control group. Specimens were collected at 1, 2, 6, and 12 weeks postoperatively, and bone regeneration at the defect site was evaluated by micro-CT, histomorphometry, immunohistochemistry, double-labeling immunofluorescence, and RT-qPCR. Results The application of exogenous IL-4 reduced activation of NLRP3 inflammasomes by promoting the polarization of M1 macrophages to M2 macrophages, thus promoting bone regeneration at the site of bone defects in aged rats. However, this effect was gradually weakened after the IL-4 intervention was discontinued. Conclusion Our data confirmed that a strategy to regulate macrophage polarization is also feasible under conditions of immunosenescence, i.e., the local inflammatory microenvironment can be regulated by reducing M1-type macrophages. However, further experiments are needed to determine an exogenous IL-4 intervention that can maintain a more sustained effect.
Collapse
Affiliation(s)
- Duchenhui Li
- Department of Prosthodontics and Implantology, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, China
- Department of Physiology and Pathology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiao Li
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, Guizhou, China
| | - Jie Zhang
- Department of Prosthodontics and Implantology, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhenglong Tang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, China
- Department of Physiology and Pathology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- *Correspondence: Ai Tian, ; Zhenglong Tang,
| | - Ai Tian
- Department of Prosthodontics and Implantology, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, Guizhou, China
- *Correspondence: Ai Tian, ; Zhenglong Tang,
| |
Collapse
|
48
|
Daghrery A, Ferreira JA, Xu J, Golafshan N, Kaigler D, Bhaduri SB, Malda J, Castilho M, Bottino MC. Tissue-specific melt electrowritten polymeric scaffolds for coordinated regeneration of soft and hard periodontal tissues. Bioact Mater 2023; 19:268-281. [PMID: 35574052 PMCID: PMC9058963 DOI: 10.1016/j.bioactmat.2022.04.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/14/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Periodontitis is a chronic inflammatory condition that often causes serious damage to tooth-supporting tissues. The limited successful outcomes of clinically available approaches underscore the need for therapeutics that cannot only provide structural guidance to cells but can also modulate the local immune response. Here, three-dimensional melt electrowritten (i.e., poly(ε-caprolactone)) scaffolds with tissue-specific attributes were engineered to guide differentiation of human-derived periodontal ligament stem cells (hPDLSCs) and mediate macrophage polarization. The investigated tissue-specific scaffold attributes comprised fiber morphology (aligned vs. random) and highly-ordered architectures with distinct strand spacings (small 250 μm and large 500 μm). Macrophages exhibited an elongated morphology in aligned and highly-ordered scaffolds, while maintaining their round-shape on randomly-oriented fibrous scaffolds. Expressions of periostin and IL-10 were more pronounced on the aligned and highly-ordered scaffolds. While hPDLSCs on the scaffolds with 500 μm strand spacing show higher expression of osteogenic marker (Runx2) over 21 days, cells on randomly-oriented fibrous scaffolds showed upregulation of M1 markers. In an orthotopic mandibular fenestration defect model, findings revealed that the tissue-specific scaffolds (i.e., aligned fibers for periodontal ligament and highly-ordered 500 μm strand spacing fluorinated calcium phosphate [F/CaP]-coated fibers for bone) could enhance the mimicking of regeneration of natural periodontal tissues.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Jessica A. Ferreira
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Jinping Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Nasim Golafshan
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Utrecht, the Netherlands
| | - Darnell Kaigler
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Sarit B. Bhaduri
- Department of Mechanical, Industrial and Manufacturing Engineering, University of Toledo, Toledo, OH, United States
- EEC Division, Directorate of Engineering, The National Science Foundation, Alexandria, VA, United States
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Utrecht, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Marco C. Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
49
|
Suturin AC, Krüger AJD, Neidig K, Klos N, Dolfen N, Bund M, Gronemann T, Sebers R, Manukanc A, Yazdani G, Kittel Y, Rommel D, Haraszti T, Köhler J, De Laporte L. Annealing High Aspect Ratio Microgels into Macroporous 3D Scaffolds Allows for Higher Porosities and Effective Cell Migration. Adv Healthc Mater 2022; 11:e2200989. [PMID: 36100464 PMCID: PMC11469137 DOI: 10.1002/adhm.202200989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/25/2022] [Indexed: 01/28/2023]
Abstract
Growing millimeter-scaled functional tissue remains a major challenge in the field of tissue engineering. Therefore, microporous annealed particles (MAPs) are emerging as promising porous biomaterials that are formed by assembly of microgel building blocks. To further vary the pore size and increase overall MAP porosity of mechanically stable scaffolds, rod-shaped microgels with high aspect ratios up to 20 are chemically interlinked into highly porous scaffolds. Polyethylene glycol based microgels (width 10 µm, lengths up to 200 µm) are produced via in-mold polymerization and covalently interlinked into stable 3D scaffolds via epoxy-amine chemistry. For the first time, MAP porosities can be enhanced by increasing the microgel aspect ratio (mean pore sizes ranging from 39 to 82 µm, porosities from 65 to 90%). These porosities are significantly higher compared to constructs made from spherical or lower aspect ratio rod-shaped microgels. Rapid filling of the pores by either murine or primary human fibroblasts is ensured as cells migrate and grow extensively into these scaffolds. Overall, this study demonstrates that highly porous, stable macroporous hydrogels can be achieved with a very low partial volume of synthetic, high aspect ratio microgels, leading to large empty volumes available for cell ingrowth and cell-cell interactions.
Collapse
Affiliation(s)
- Alisa C. Suturin
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
| | - Andreas J. D. Krüger
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
| | - Kathrin Neidig
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Nina Klos
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Nina Dolfen
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Michelle Bund
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Till Gronemann
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Rebecca Sebers
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Anna Manukanc
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Ghazaleh Yazdani
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
| | - Yonca Kittel
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
| | - Dirk Rommel
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
| | - Tamás Haraszti
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
| | - Jens Köhler
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
| | - Laura De Laporte
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 252074AachenGermany
- Advanced Materials for Biomedicine (AMB)Institute of Applied Medical Engineering (AME)University Hospital RWTH AachenCenter for Biohybrid Medical Systems (CMBS)Forckenbeckstraße 5552074AachenGermany
| |
Collapse
|
50
|
Li Y, Yang L, Hou Y, Zhang Z, Chen M, Wang M, Liu J, Wang J, Zhao Z, Xie C, Lu X. Polydopamine-mediated graphene oxide and nanohydroxyapatite-incorporated conductive scaffold with an immunomodulatory ability accelerates periodontal bone regeneration in diabetes. Bioact Mater 2022; 18:213-227. [PMID: 35387166 PMCID: PMC8961429 DOI: 10.1016/j.bioactmat.2022.03.021] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022] Open
Abstract
Regenerating periodontal bone tissues in the aggravated inflammatory periodontal microenvironment under diabetic conditions is a great challenge. Here, a polydopamine-mediated graphene oxide (PGO) and hydroxyapatite nanoparticle (PHA)-incorporated conductive alginate/gelatin (AG) scaffold is developed to accelerate periodontal bone regeneration by modulating the diabetic inflammatory microenvironment. PHA confers the scaffold with osteoinductivity and PGO provides a conductive pathway for the scaffold. The conductive scaffold promotes bone regeneration by transferring endogenous electrical signals to cells and activating Ca2+ channels. Moreover, the scaffold with polydopamine-mediated nanomaterials has a reactive oxygen species (ROS)-scavenging ability and anti-inflammatory activity. It also exhibits an immunomodulatory ability that suppresses M1 macrophage polarization and activates M2 macrophages to secrete osteogenesis-related cytokines by mediating glycolytic and RhoA/ROCK pathways in macrophages. The scaffold induces excellent bone regeneration in periodontal bone defects of diabetic rats because of the synergistic effects of good conductive, ROS-scavenging, anti-inflammatory, and immunomodulatory abilities. This study provides fundamental insights into the synergistical effects of conductivity, osteoinductivity, and immunomodulatory abilities on bone regeneration and offers a novel strategy to design immunomodulatory biomaterials for treatment of immune-related diseases and tissue regeneration. The conductive PGO-PHA-AG scaffold can activate Ca2+ channels. •The PGO-PHA-AG scaffold had ROS-scavenging and anti-inflammatory activities. •The scaffold exhibited an immunomodulatory ability. •The scaffold induced excellent periodontal bone regeneration in diabetes.
Collapse
Affiliation(s)
- Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lu Yang
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yue Hou
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Maoxia Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jin Liu
- Lab for Aging Research and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Corresponding author.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Corresponding author.
| | - Chaoming Xie
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Corresponding author.
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Corresponding author.
| |
Collapse
|