1
|
Castañón-Cortés LG, Bravo-Vázquez LA, Santoyo-Valencia G, Medina-Feria S, Sahare P, Duttaroy AK, Paul S. Current advances in the development of microRNA-integrated tissue engineering strategies: a cornerstone of regenerative medicine. Front Bioeng Biotechnol 2024; 12:1484151. [PMID: 39479296 PMCID: PMC11521876 DOI: 10.3389/fbioe.2024.1484151] [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: 08/21/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Regenerative medicine is an innovative scientific field focused on repairing, replacing, or regenerating damaged tissues and organs to restore their normal functions. A central aspect of this research arena relies on the use of tissue-engineered scaffolds, which serve as structural supports that mimic the extracellular matrix, providing an environment that orchestrates cell growth and tissue formation. Remarkably, the therapeutic efficacy of these scaffolds can be improved by harnessing the properties of other molecules or compounds that have crucial roles in healing and regeneration pathways, such as phytochemicals, enzymes, transcription factors, and non-coding RNAs (ncRNAs). In particular, microRNAs (miRNAs) are a class of tiny (20-24 nt), highly conserved ncRNAs that play a critical role in the regulation of gene expression at the post-transcriptional level. Accordingly, miRNAs are involved in a myriad of biological processes, including cell differentiation, proliferation, and apoptosis, as well as tissue regeneration, angiogenesis, and osteogenesis. On this basis, over the past years, a number of research studies have demonstrated that miRNAs can be integrated into tissue-engineered scaffolds to create advanced therapeutic platforms that precisely modulate cellular behavior and offer a controlled and targeted release of miRNAs to optimize tissue repair and regeneration. Therefore, in this current review, we discuss the most recent advances in the development of miRNA-loaded tissue-engineered scaffolds and provide an overview of the future outlooks that should be aborded in this area of study in order to lay the groundwork for the clinical translation of these tissue engineering approaches.
Collapse
Affiliation(s)
| | | | | | - Sara Medina-Feria
- School of Engineering and Sciences, Tecnologico de Monterrey, Queretaro, Mexico
| | - Padmavati Sahare
- School of Engineering and Sciences, Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Queretaro, Mexico
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Queretaro, Mexico
| |
Collapse
|
2
|
Fratila DN, Virvescu DI, Luchian I, Hancianu M, Baciu ER, Butnaru O, Budala DG. Advances and Functional Integration of Hydrogel Composites as Drug Delivery Systems in Contemporary Dentistry. Gels 2024; 10:661. [PMID: 39451314 PMCID: PMC11507597 DOI: 10.3390/gels10100661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
This study explores the recent advances of and functional insights into hydrogel composites, materials that have gained significant attention for their versatile applications across various fields, including contemporary dentistry. Hydrogels, known for their high water content and biocompatibility, are inherently soft but often limited by mechanical fragility. Key areas of focus include the customization of hydrogel composites for biomedical applications, such as drug delivery systems, wound dressings, and tissue engineering scaffolds, where improved mechanical properties and bioactivity are critical. In dentistry, hydrogels are utilized for drug delivery systems targeting oral diseases, dental adhesives, and periodontal therapies due to their ability to adhere to the mucosa, provide localized treatment, and support tissue regeneration. Their unique properties, such as mucoadhesion, controlled drug release, and stimuli responsiveness, make them ideal candidates for treating oral conditions. This review highlights both experimental breakthroughs and theoretical insights into the structure-property relationships within hydrogel composites, aiming to guide future developments in the design and application of these multifunctional materials in dentistry. Ultimately, hydrogel composites represent a promising frontier for advancing materials science with far-reaching implications in healthcare, environmental technology, and beyond.
Collapse
Affiliation(s)
- Dragos Nicolae Fratila
- Department of Oral Diagnosis, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Dragos Ioan Virvescu
- Department of Dental Materials, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionut Luchian
- Department of Periodontology, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Monica Hancianu
- Department of Pharmacognosy, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Elena Raluca Baciu
- Department of Dental Materials, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Oana Butnaru
- Department of Biophysics, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Dana Gabriela Budala
- Department of Prosthodontics, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
3
|
Shiwaku Y, Okawa H, Suzuki I, Sakai S, Egusa H, Suzuki O. Induced pluripotent stem cell-derived neural stem cells promote bone formation in mice with calvarial defects. Acta Biomater 2024; 188:93-102. [PMID: 39241820 DOI: 10.1016/j.actbio.2024.08.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/27/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Nerve-derived factors have attracted attention in bone regeneration therapy due to their ability to promote bone regeneration and nerve innervation. Mesenchymal stem cells transported to target sites promote osteogenesis. However, there are few reports on the effects of neural stem cells on bone regeneration. Therefore, the aim of this study was to investigate the role of neural stem cells in osteogenesis. Here, embryoid bodies (EB) or primary neurospheres (1NS) were generated using mouse induced pluripotent stem cells (iPS cells), which were then seeded onto gelatin (Gel) sponges. The seeded Gel sponges were then transplanted into mouse calvarial bone defects. We noted that 1NS-seeded Gel promoted bone regeneration and the presence of tartrate-resistant acid phosphatase (TRAP)-positive cells, whereas the EB-seeded Gel did not. RNA-sequencing of the 1NS-seeded and EB seeded Gels showed an upregulation of the transforming growth factor (TGF)-β signaling pathway in the 1NS-seeded Gel group. Immunostaining confirmed the presence of Id3 positive cells in mice with bone defects treated with the 1NS-seeded Gel. These findings suggest that the transplantation of neural stem cells may contribute to the promotion of bone regeneration. STATEMENT OF SIGNIFICANCE: This study aimed to investigate whether neural stem cells, when seeded in Gel sponges, promoted bone regeneration. It has been well documented that bone is tightly linked with the nervous systems. Bioscaffolds comprising factors that promote innervation and bone regeneration have been investigated for use in bone therapy. However, there is limited research on the use of neural stem cells for promoting bone formation. To assess this relationship, we conducted both in vivo and in vitro assays to determine whether neural stem cells promoted bone formation. We noted that 1NS-seeded Gel sponges promoted bone formation significantly in mice with calvarial defects after 4 weeks. This study provides a novel approach of neural stem cells for bone therapy.
Collapse
Affiliation(s)
- Yukari Shiwaku
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Hiroko Okawa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Susumu Sakai
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|
4
|
Maphanao P, Phothikul Y, Choodet C, Puangmali T, Katewongsa K, Pinlaor S, Thanan R, Yordpratum U, Sakonsinsiri C. Development and in vitro evaluation of ursolic acid-loaded poly(lactic- co-glycolic acid) nanoparticles in cholangiocarcinoma. RSC Adv 2024; 14:24828-24837. [PMID: 39119279 PMCID: PMC11306966 DOI: 10.1039/d4ra03637a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Cholangiocarcinoma (CCA), an epithelial biliary tract malignancy, is a significant health concern in the Greater Mekong Subregion, particularly in northeastern Thailand. Prior to the development of advanced stages, CCA is typically asymptomatic, thereby limiting treatment options and chemotherapeutic effectiveness. Ursolic acid (UA), a triterpenoid derived from plants, was previously discovered to inhibit CCA cell growth through induction of apoptosis. Nevertheless, the therapeutic effectiveness of UA is limited by its poor solubility in water and low bioavailability; therefore, dimethyl sulfoxide (DMSO) is utilized as a solvent to treat UA with CCA cells. Enhancing cellular uptake and reducing toxicity, the utilization of polymeric nanoparticles (NPs) proves beneficial. In this study, UA-loaded PLGA nanoparticles (UA-PLGA NPs) were synthesized using nanoprecipitation and characterized through in silico formation analysis, average particle size, surface functional groups and ζ-potential measurements, electron microscopic imaging, drug loading efficiency and drug release studies, stability, hemo- and biocompatibility, cytotoxicity and cellular uptake assays. Molecular dynamics simulations validated the loading of UA into PLGA via hydrogen bonding. The synthesized UA-PLGA NPs had a spherical shape with an average size of 240 nm, a negative ζ-potential, good stability, great hemo- and bio-compatibility and an encapsulation efficiency of 98%. The NPs exhibited a characteristic of a simple diffusion-controlled Fickian process, as predicted by the Peppas-Sahlin drug release kinetic model. UA-PLGA NPs exhibited cytotoxic effects on KKU-213A and KKU-055 CCA cells even when dispersed in media without organic solvent, i.e., DMSO, highlighting the ability of PLGA NPs to overcome the poor water solubility of UA. Rhodamine 6G (R6G) was loaded into PLGA NPs using the same approach as UA-PLGA NPs, demonstrating effective delivery of the dye into CCA cells. These findings suggest that UA-PLGA NPs showed promise as a potential phytochemical delivery system for CCA treatment.
Collapse
Affiliation(s)
- Pornpattra Maphanao
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University Khon Kaen 40002 Thailand
| | - Yaowaret Phothikul
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
| | - Cherdpong Choodet
- Department of Physics, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Theerapong Puangmali
- Department of Physics, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Kanlaya Katewongsa
- Department of Biochemistry, Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Somchai Pinlaor
- Cholangiocarcinoma Research Institute, Khon Kaen University Khon Kaen 40002 Thailand
- Department of Parasitology, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
| | - Raynoo Thanan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University Khon Kaen 40002 Thailand
| | - Umaporn Yordpratum
- Department of Microbiology, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University Khon Kaen 40002 Thailand
| |
Collapse
|
5
|
Chen J, Luo J, Feng J, Wang Y, Lv H, Zhou Y. Spatiotemporal controlled released hydrogels for multi-system regulated bone regeneration. J Control Release 2024; 372:846-861. [PMID: 38955252 DOI: 10.1016/j.jconrel.2024.06.065] [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: 04/26/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Bone defect is one of the urgent problems to be solved in clinics, and it is very important to construct efficient scaffold materials to facilitate bone tissue regeneration. Hydrogels, characterized by their unique three-dimensional network structure, serve as excellent biological scaffold materials. Their internal pores are capable of loading osteogenic drugs to expedite bone formation. The rate and quality of new bone formation are intimately linked with immune regulation and vascular remodeling. The strategic sequential release of drugs to balance inflammation and regulate vascular remodeling is crucial for initiating the osteogenic process. Through the design of hydrogel microstructures, it is possible to achieve sequential drug release and the drug action time can be prolonged, thereby catering to the multi-systemic collaborative regulation needs of osteosynthesis. The drug release rate within the hydrogel is governed by swelling control systems, physical control systems, chemical control systems, and environmental control systems. Utilizing these control systems to design hydrogel materials capable of multi-drug delivery optimizes the construction of the bone microenvironment. Consequently, this facilitates the spatiotemporal controlled released of drugs, promoting bone tissue regeneration. This paper reviews the principles of the controlled release system of various sustained-release hydrogels and the advancements in research on hydrogel multi-drug delivery systems for bone tissue regeneration.
Collapse
Affiliation(s)
- Jingxia Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jiaxin Luo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jian Feng
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yihan Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Huixin Lv
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Li J, Zhang Z, Tang J, Hou Z, Li L, Li B. Emerging roles of nerve-bone axis in modulating skeletal system. Med Res Rev 2024; 44:1867-1903. [PMID: 38421080 DOI: 10.1002/med.22031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/25/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Over the past decades, emerging evidence in the literature has demonstrated that the innervation of bone is a crucial modulator for skeletal physiology and pathophysiology. The nerve-bone axis sparked extensive preclinical and clinical investigations aimed at elucidating the contribution of nerve-bone crosstalks to skeleton metabolism, homeostasis, and injury repair through the perspective of skeletal neurobiology. To date, peripheral nerves have been widely reported to mediate bone growth and development and fracture healing via the secretion of neurotransmitters, neuropeptides, axon guidance factors, and neurotrophins. Relevant studies have further identified several critical neural pathways that stimulate profound alterations in bone cell biology, revealing a complex interplay between the skeleton and nerve systems. In addition, inspired by nerve-bone crosstalk, novel drug delivery systems and bioactive materials have been developed to emulate and facilitate the process of natural bone repair through neuromodulation, eventually boosting osteogenesis for ideal skeletal tissue regeneration. Overall, this work aims to review the novel research findings that contribute to deepening the current understanding of the nerve-bone axis, bringing forth some schemas that can be translated into the clinical scenario to highlight the critical roles of neuromodulation in the skeletal system.
Collapse
Affiliation(s)
- Jingya Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhuoyuan Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinru Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zeyu Hou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Li X, Cui Y, He X, Mao L. Hydrogel-Based Systems in Neuro-Vascularized Bone Regeneration: A Promising Therapeutic Strategy. Macromol Biosci 2024; 24:e2300484. [PMID: 38241425 DOI: 10.1002/mabi.202300484] [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: 10/25/2023] [Revised: 12/16/2023] [Indexed: 01/21/2024]
Abstract
Blood vessels and nerve fibers are distributed throughout the skeletal tissue, which enhance the development and function of each other and have an irreplaceable role in bone formation and remodeling. Despite significant progress in bone tissue engineering, the inadequacy of nerve-vascular network reconstruction remains a major limitation. This is partly due to the difficulty of integrating and regulating multiple tissue types with artificial materials. Thus, understanding the anatomy and underlying coupling mechanisms of blood vessels and nerve fibers within bone to further develop neuro-vascularized bone implant biomaterials is an extremely critical aspect in the field of bone regeneration. Hydrogels have good biocompatibility, controllable mechanical characteristics, and osteoconductive and osteoinductive properties, making them important candidates for research related to neuro-vascularized bone regeneration. This review reports the classification and physicochemical properties of hydrogels, with a focus on the application advantages and status of hydrogels for bone regeneration. The authors also highlight the effect of neurovascular coupling on bone repair and regeneration and the necessity of achieving neuro-vascularized bone regeneration. Finally, the recent progress and design strategies of hydrogel-based biomaterials for neuro-vascularized bone regeneration are discussed.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| | - Ya Cui
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| | - Xiaoya He
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| | - Lixia Mao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200000, China
| |
Collapse
|
8
|
Damiati LA, El Soury M. Bone-nerve crosstalk: a new state for neuralizing bone tissue engineering-A mini review. Front Med (Lausanne) 2024; 11:1386683. [PMID: 38690172 PMCID: PMC11059066 DOI: 10.3389/fmed.2024.1386683] [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: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Neuro bone tissue engineering is a multidisciplinary field that combines both principles of neurobiology and bone tissue engineering to develop innovative strategies for repairing and regenerating injured bone tissues. Despite the fact that regeneration and development are considered two distinct biological processes, yet regeneration can be considered the reactivation of development in later life stages to restore missing tissues. It is noteworthy that the regeneration capabilities are distinct and vary from one organism to another (teleost fishes, hydra, humans), or even in the same organism can vary dependent on the injured tissue itself (Human central nervous system vs. peripheral nervous system). The skeletal tissue is highly innervated, peripheral nervous system plays a role in conveying the signals and connecting the central nervous system with the peripheral organs, moreover it has been shown that they play an important role in tissue regeneration. Their regeneration role is conveyed by the different cells' resident in it and in its endoneurium (fibroblasts, microphages, vasculature associated cells, and Schwann cells) these cells secrete various growth factors (NGF, BDNF, GDNF, NT-3, and bFGF) that contribute to the regenerative phenotype. The peripheral nervous system and central nervous system synchronize together in regulating bone homeostasis and regeneration through neurogenic factors and neural circuits. Receptors of important central nervous system peptides such as Serotonin, Leptin, Semaphorins, and BDNF are expressed in bone tissue playing a role in bone homeostasis, metabolism and regeneration. This review will highlight the crosstalk between peripheral nerves and bone in the developmental stages as well as in regeneration and different neuro-bone tissue engineering strategies for repairing severe bone injuries.
Collapse
Affiliation(s)
- Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Marwa El Soury
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| |
Collapse
|
9
|
Genedy HH, Humbert P, Laoulaou B, Le Moal B, Fusellier M, Passirani C, Le Visage C, Guicheux J, Lepeltier É, Clouet J. MicroRNA-targeting nanomedicines for the treatment of intervertebral disc degeneration. Adv Drug Deliv Rev 2024; 207:115214. [PMID: 38395361 DOI: 10.1016/j.addr.2024.115214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Low back pain stands as a pervasive global health concern, afflicting almost 80% of adults at some point in their lives with nearly 40% attributable to intervertebral disc degeneration (IVDD). As only symptomatic relief can be offered to patients there is a dire need for innovative treatments.Given the accumulating evidence that multiple microRNAs (miRs) are dysregulated during IVDD, they could have a huge potential against this debilitating condition. The way miRs can profoundly modulate signaling pathways and influence several cellular processes at once is particularly exciting to tackle this multifaceted disorder. However, miR delivery encounters extracellular and intracellular biological barriers. A promising technology to address this challenge is the vectorization of miRs within nanoparticles, providing both protection and enhancing their uptake within the scarce target cells of the degenerated IVD. This comprehensive review presents the diverse spectrum of miRs' connection with IVDD and demonstrates their therapeutic potential when vectorized in nanomedicines.
Collapse
Affiliation(s)
- Hussein H Genedy
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Paul Humbert
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Bilel Laoulaou
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Brian Le Moal
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Marion Fusellier
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Department of Diagnostic Imaging, CRIP, ONIRIS, College of Veterinary Medicine, Food Science and Engineering, Nantes F-44307, France
| | | | - Catherine Le Visage
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Élise Lepeltier
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France; Institut Universitaire de France (IUF), France.
| | - Johann Clouet
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| |
Collapse
|
10
|
Xu H, Cui Y, Tian Y, Dou M, Sun S, Wang J, Wu D. Nanoparticle-Based Drug Delivery Systems for Enhancing Bone Regeneration. ACS Biomater Sci Eng 2024; 10:1302-1322. [PMID: 38346448 DOI: 10.1021/acsbiomaterials.3c01643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The treatment of bone defects has been a long-standing challenge in clinical practice. Among the various bone tissue engineering approaches, there has been substantial progress in the development of drug delivery systems based on functional drugs and appropriate carrier materials owing to technological advances in recent years. A large number of materials based on functional nanocarriers have been developed and applied to improve the complex osteogenic microenvironment, including for promoting osteogenic activity, inhibiting osteoclast activity, and exerting certain antibacterial effects. This Review discusses the physicochemical properties, drug loading mechanisms, advantages and disadvantages of nanoparticles (NPs) used for constructing drug delivery systems. In addition, we provide an overview of the osteogenic microenvironment regulation mechanism of drug delivery systems based on nanoparticle (NP) carriers and the construction strategies of drug delivery systems. Finally, the advantages and disadvantages of NP carriers are summarized along with their prospects and future research trends in bone tissue engineering. This Review thus provides advanced strategies for the design and application of drug delivery systems based on NPs in the treatment of bone defects.
Collapse
Affiliation(s)
- Hang Xu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yutao Cui
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yuhang Tian
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Minghan Dou
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Shouye Sun
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Jingwei Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Dankai Wu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| |
Collapse
|
11
|
Huang X, Lou Y, Duan Y, Liu H, Tian J, Shen Y, Wei X. Biomaterial scaffolds in maxillofacial bone tissue engineering: A review of recent advances. Bioact Mater 2024; 33:129-156. [PMID: 38024227 PMCID: PMC10665588 DOI: 10.1016/j.bioactmat.2023.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Maxillofacial bone defects caused by congenital malformations, trauma, tumors, and inflammation can severely affect functions and aesthetics of maxillofacial region. Despite certain successful clinical applications of biomaterial scaffolds, ideal bone regeneration remains a challenge in maxillofacial region due to its irregular shape, complex structure, and unique biological functions. Scaffolds that address multiple needs of maxillofacial bone regeneration are under development to optimize bone regeneration capacity, costs, operational convenience. etc. In this review, we first highlight the special considerations of bone regeneration in maxillofacial region and provide an overview of the biomaterial scaffolds for maxillofacial bone regeneration under clinical examination and their efficacy, which provide basis and directions for future scaffold design. Latest advances of these scaffolds are then discussed, as well as future perspectives and challenges. Deepening our understanding of these scaffolds will help foster better innovations to improve the outcome of maxillofacial bone tissue engineering.
Collapse
Affiliation(s)
- Xiangya Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yaxin Lou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yihong Duan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - He Liu
- Division of Endodontics, Department of Oral Biological and Medical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jun Tian
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Ya Shen
- Division of Endodontics, Department of Oral Biological and Medical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
12
|
Yang J, Shuai J, Siow L, Lu J, Sun M, An W, Yu M, Wang B, Chen Q. MicroRNA-146a-loaded magnesium silicate nanospheres promote bone regeneration in an inflammatory microenvironment. Bone Res 2024; 12:2. [PMID: 38221522 PMCID: PMC10788347 DOI: 10.1038/s41413-023-00299-0] [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: 04/18/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 01/16/2024] Open
Abstract
Reconstruction of irregular oral-maxillofacial bone defects with an inflammatory microenvironment remains a challenge, as chronic local inflammation can largely impair bone healing. Here, we used magnesium silicate nanospheres (MSNs) to load microRNA-146a-5p (miR-146a) to fabricate a nanobiomaterial, MSN+miR-146a, which showed synergistic promoting effects on the osteogenic differentiation of human dental pulp stem cells (hDPSCs). In addition, miR-146a exhibited an anti-inflammatory effect on mouse bone marrow-derived macrophages (BMMs) under lipopolysaccharide (LPS) stimulation by inhibiting the NF-κB pathway via targeting tumor necrosis factor receptor-associated factor 6 (TRAF6), and MSNs could simultaneously promote M2 polarization of BMMs. MiR-146a was also found to inhibit osteoclast formation. Finally, the dual osteogenic-promoting and immunoregulatory effects of MSN+miR-146a were further validated in a stimulated infected mouse mandibular bone defect model via delivery by a photocuring hydrogel. Collectively, the MSN+miR-146a complex revealed good potential in treating inflammatory irregular oral-maxillofacial bone defects.
Collapse
Affiliation(s)
- Jiakang Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Jing Shuai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Lixuen Siow
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Jingyi Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Miao Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Wenyue An
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Baixiang Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China.
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
13
|
Zhang Y, Li C, Wei Q, Yuan Q, He W, Zhang N, Dong Y, Jing Z, Zhang L, Wang H, Cao X. MiRNA320a Inhibitor-Loaded PLGA-PLL-PEG Nanoparticles Contribute to Bone Regeneration in Trauma-Induced Osteonecrosis Model of the Femoral Head. Tissue Eng Regen Med 2024; 21:185-197. [PMID: 37828392 PMCID: PMC10764684 DOI: 10.1007/s13770-023-00580-7] [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: 05/11/2023] [Revised: 07/07/2023] [Accepted: 07/19/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND This study aimed to explore the effect of a nanomaterial-based miR-320a inhibitor sustained release system in trauma-induced osteonecrosis of the femoral head (TIONFH). METHODS The miR-320a inhibitor-loaded polyethylene glycol (PEG)- Poly(lactic-co-glycolic acid) (PLGA)- Poly-L-lysine (PLL) nanoparticles were constructed using the double emulsion method. The TIONFH rabbit model was established to observe the effects of miR-320a inhibitor nanoparticles in vivo. Hematoxylin-eosin staining and microcomputed tomography scanning were used for bone morphology analysis. Bone marrow mesenchymal stem cells (BMSCs), derived from TIONFH rabbits, were used for in vitro experiments. Cell viability was determined using the MTT assay. RESULTS High expression of miR-320a inhibited the osteogenic differentiation capacity of BMSCs in vitro by inhibiting the expression of the osteoblastic differentiation markers ALP and RUNX2. MiR-320a inhibitor-loaded PEG-PLGA-PLL nanoparticles were constructed with a mean loading efficiency of 1.414 ± 0.160%, and a mean encapsulation efficiency of 93.45 ± 1.24%, which released 50% of the loaded miR-320a inhibitor at day 12 and 80% on day 18. Then, inhibitor release entered the plateau. After treatment with the miR-320a inhibitor nanoparticle, the empty lacunae were decreased in the femoral head tissue of TIONFH rabbits, and the osteoblast surface/bone surface (Ob.S/BS), osteoblast number/bone perimeter (Ob.N/B.Pm), bone volume fraction, and bone mineral density increased. Additionally, the expression of osteogenic markers RUNX2 and ALP was significantly elevated in the TIONFH rabbit model. CONCLUSION The miR-320a inhibitor-loaded PEG-PLGA-PLL nanoparticle sustained drug release system significantly contributed to bone regeneration in the TIONFH rabbit model, which might be a promising strategy for the treatment of TIONFH.
Collapse
Affiliation(s)
- Ying Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), No. 82 Qiming South Road, Luoyang, 471002, Henan, China.
| | - Chuan Li
- Department of Orthopedics, 920Th Hospital of Joint Logistic Support Force, Kunming, 650032, Yunnan, China
| | - Qiushi Wei
- Institute of Orthopaedics of Guangzhou, University of Traditional Chinese Medicine, Guangzhou, 510240, China
- The Third Affiliated Hospital of Guangzhou, University of Traditional Chinese Medicine, Guangzhou, 510240, China
| | - Qiang Yuan
- Henan University of Traditional Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Wei He
- Institute of Orthopaedics of Guangzhou, University of Traditional Chinese Medicine, Guangzhou, 510240, China
- The Third Affiliated Hospital of Guangzhou, University of Traditional Chinese Medicine, Guangzhou, 510240, China
| | - Ning Zhang
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yiping Dong
- Henan University of Traditional Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Zhenhao Jing
- Henan University of Traditional Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Leilei Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), No. 82 Qiming South Road, Luoyang, 471002, Henan, China
| | - Haibin Wang
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xiangyang Cao
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), No. 82 Qiming South Road, Luoyang, 471002, Henan, China.
| |
Collapse
|
14
|
Hu S, Liang Y, Chen J, Gao X, Zheng Y, Wang L, Jiang J, Zeng M, Luo M. Mechanisms of hydrogel-based microRNA delivery systems and its application strategies in targeting inflammatory diseases. J Tissue Eng 2024; 15:20417314241265897. [PMID: 39092451 PMCID: PMC11292707 DOI: 10.1177/20417314241265897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
Hydrogels, composed of three-dimensional polymer networks, are excellent delivery carriers and have been extensively employed in the biomedical field. Inflammation acts as a protective mechanism to prevent harmful substances from entering living organisms, but chronic, long-lasting inflammation can cause oxidative stress, which damages tissue and organs and adversely affects patients' quality of life. The aberrant expression of microRNAs (miRNAs) has been found to play a significant part in the etiology and progression of inflammatory diseases, as suggested by growing evidence. Numerous hydrogels that can act as gene carriers for the intracellular delivery of miRNA have been described during ongoing research into innovative hydrogel materials. MiRNA hydrogel delivery systems, which are loaded with exogenous miRNA inhibitors or mimics, enable targeted miRNA intervention in inflammatory diseases and effectively prevent environmental stressors from degrading or inactivating miRNA. In this review, we summarize the classification of miRNA hydrogel delivery systems, the basic strategies and mechanisms for loading miRNAs into hydrogels, highlight the biomedical applications of miRNA hydrogel delivery systems in inflammatory diseases, and share our viewpoints on potential opportunities and challenges in the promising region of miRNA delivery systems. These findings may provide a new theoretical basis for the prevention and treatment of inflammation-related diseases and lay the foundation for clinical translation.
Collapse
Affiliation(s)
- Shaorun Hu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinxiang Chen
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Youkun Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Liqun Wang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Min Zeng
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
16
|
Liu J, Du C, Huang W, Lei Y. Injectable smart stimuli-responsive hydrogels: pioneering advancements in biomedical applications. Biomater Sci 2023; 12:8-56. [PMID: 37969066 DOI: 10.1039/d3bm01352a] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Hydrogels have established their significance as prominent biomaterials within the realm of biomedical research. However, injectable hydrogels have garnered greater attention compared with their conventional counterparts due to their excellent minimally invasive nature and adaptive behavior post-injection. With the rapid advancement of emerging chemistry and deepened understanding of biological processes, contemporary injectable hydrogels have been endowed with an "intelligent" capacity to respond to various endogenous/exogenous stimuli (such as temperature, pH, light and magnetic field). This innovation has spearheaded revolutionary transformations across fields such as tissue engineering repair, controlled drug delivery, disease-responsive therapies, and beyond. In this review, we comprehensively expound upon the raw materials (including natural and synthetic materials) and injectable principles of these advanced hydrogels, concurrently providing a detailed discussion of the prevalent strategies for conferring stimulus responsiveness. Finally, we elucidate the latest applications of these injectable "smart" stimuli-responsive hydrogels in the biomedical domain, offering insights into their prospects.
Collapse
Affiliation(s)
- Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
17
|
Karmakar R, Dey S, Alam A, Khandelwal M, Pati F, Rengan AK. Attributes of Nanomaterials and Nanotopographies for Improved Bone Tissue Engineering and Regeneration. ACS APPLIED BIO MATERIALS 2023; 6:4020-4041. [PMID: 37691480 DOI: 10.1021/acsabm.3c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Bone tissue engineering (BTE) is a multidisciplinary area that can solve the limitation of conventional grafting methods by developing viable and biocompatible bone replacements. The three essential components of BTE, i.e., Scaffold material and Cells and Growth factors altogether, facilitate support and guide for bone formation, differentiation of the bone tissues, and enhancement in the cellular activities and bone regeneration. However, there is a scarcity of the appropriate materials that can match the mechanical property as well as functional similarity to native tissue, considering the bone as hard tissue. In such scenarios, nanotechnology can be leveraged upon to achieve the desired aspects of BTE, and that is the key point of this review article. This review article examines the significant areas of nanotechnology research that have an impact on regeneration of bone: (a) scaffold with nanomaterials helps to enhance physicochemical interactions, biocompatibility, mechanical stability, and attachment; (b) nanoparticle-based approaches for delivering bioactive chemicals, growth factors, and genetic material. The article begins with the introduction of components and healing mechanisms of bone and the factors associated with them. The focus of this article is on the various nanotopographies that are now being used in scaffold formation, by describing how they are made, and how these nanotopographies affect the immune system and potential underlying mechanisms. The advantages of 4D bioprinting in BTE by using nanoink have also been mentioned. Additionally, we have investigated the importance of an in silico approach for finding the interaction between drugs and their related receptors, which can help to formulate suitable systems for delivery. This review emphasizes the role of nanoscale approach and how it helps to increase the efficacy of parameters of scaffold as well as drug delivery system for tissue engineering and bone regeneration.
Collapse
Affiliation(s)
- Rounik Karmakar
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Sreenath Dey
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Aszad Alam
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology, Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Mudrika Khandelwal
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology, Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| |
Collapse
|
18
|
Wang Y, Zhang Y, Yang Z, Zhang L, Chen X, Yang G, Zhan J, Li S, He F, Fan G. Mesoporous silica-based nanocarriers with dual response to pH and ROS for enhanced anti-inflammation therapy of 5-demethylnobiletin against psoriasis-like lesions. Int J Pharm 2023; 645:123373. [PMID: 37673281 DOI: 10.1016/j.ijpharm.2023.123373] [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: 04/28/2023] [Revised: 08/11/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Psoriasis is an inflammatory skin disease accompanied with chronic papulosquamous lesions and multiple comorbidities that considerably affect patients' quality of life. In order to develop an enhanced therapeutic strategy for psoriasis, 5-demethylnobiletin (5-DN), a kind of polymethoxyflavones (PMFs) with high anti-inflammatory activity, was delivered in vitro and in vivo by the nanocarrier of mesoporous silica nanoparticles (MSNs) both in the human keratinocytes HaCaT cell line and the mouse model with psoriasis-like lesions. The drug-loaded nanocarrier system (MSNs@5-DN) significantly improved the biocompatibility and bioavailability of 5-DN. Investigations at cell biological, histopathological, and molecular levels revealed the pharmacological mechanism of the drug delivery system, including the inhibition of inflammatory responses by downregulating the proinflammatory cytokine levels of tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6). The upregulation of anti‑inflammatory cytokine of transforming growth factor-β1 (TGF-β1) and microRNA-17-5p, a critical regulator of the PTEN/AKT pathway, was also observed. The psoriasis-like lesions were markedly ameliorated in the mouse models treated with MSNs@5-DN. The designed drug-loading system shows an enhanced therapeutic outcome for psoriasis-like lesion compared with free 5-DN. This study revealed the synergistic effect of functionalized MSNs loaded with PMFs on the clinical treatment of human psoriasis.
Collapse
Affiliation(s)
- Yimin Wang
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang 438000, PR China
| | - Yanan Zhang
- College of Chemistry and Chemical Engineering, Huanggang Normal University, Huanggang 438000, PR China
| | - Zhihui Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China
| | - Lei Zhang
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang 438000, PR China
| | - Xiangping Chen
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang 438000, PR China
| | - Guliang Yang
- National Research Center of Rice Deep Process and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Jianfeng Zhan
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang 438000, PR China
| | - Shiming Li
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang 438000, PR China; Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Feng He
- Li Shizhen College of Traditional Chinese Medicine, Huanggang Normal University, Huanggang 438000, PR China.
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China.
| |
Collapse
|
19
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [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: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
20
|
Ou Y, Zhu D. Enlarged pore of worm mesoporous silica nanoparticles improves anti-inflammatory drug absorption. Drug Deliv Transl Res 2023; 13:2475-2486. [PMID: 36940080 DOI: 10.1007/s13346-023-01326-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2023] [Indexed: 03/21/2023]
Abstract
Searching for an effective pore-enlarging agent to form mesoporous silica nanoparticles (MSN) with a creative surface frame is of great importance. Herein, several polymers were attempted to be pore-enlarging agents to form seven types of worm mesoporous silica nanoparticles (W-MSN) and analgesic indometacin that exerted functions on inflammatory diseases (breast disease, arthrophlogosis, etc.) was studied to enhance its delivery efficiency. The porous morphology differences between MSN and W-MSN were that MSN had independent mesopores while the enlarged mesopores of W-MSN were interrelated and shaped as a worm. Among all these W-MSN, WG-MSN templated by hydroxypropyl cellulose acetate succinate HG with the highest drug-loading capacity (24.78%), shortest loading time (10 h), drug dissolution improvement of almost 4 times compared to that of the raw drug, and highest bioavailability (5.48 times higher than that of raw drug and 1.52 times higher than that of MSN) was an outstanding drug carrier and can shoulder the mission to deliver drugs with high efficiency.
Collapse
Affiliation(s)
- Yuying Ou
- Department of Breast Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Demiao Zhu
- Department of Breast Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
21
|
Shams R, Behmanesh A, Mazhar FN, Vaghari AA, Hossein-Khannazer N, Agarwal T, Vosough M, Padrón JM. Developed Bone Biomaterials Incorporated with MicroRNAs to Promote Bone Regeneration: A Systematic Review, Bioinformatics, and Meta-analysis Study. ACS Biomater Sci Eng 2023; 9:5186-5204. [PMID: 37585807 DOI: 10.1021/acsbiomaterials.3c00178] [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: 08/18/2023]
Abstract
This systematic review and meta-analysis focused on the effectiveness of biomaterials integrated with specific microRNAs (miRNAs) for bone fracture repair treatment. We conducted a comprehensive search of the PubMed, Web of Science, and Scopus databases, identifying 42 relevant papers up to March 2022. Hydrogel-based scaffolds were the most commonly used, incorporating miRNAs like miR-26a, miR-21, and miR-222, with miR-26a being the most prevalent. The meta-analysis revealed significant benefits of incorporating miRNAs into scaffolds for bone repair, particularly in hydrogel scaffolds. However, some controversies were observed among studies, presenting challenges in selecting appropriate miRNAs for this purpose. The study concludes that incorporating specific miRNAs into bone biomaterials enhances bone regeneration, but further trials comparing different biomaterials and miRNAs are necessary to validate their potential applications for bone tissue regeneration.
Collapse
Affiliation(s)
- Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Ali Behmanesh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Farid Najd Mazhar
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Amir Ali Vaghari
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Tarun Agarwal
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh 522302, India
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 171 77 Stockholm, Sweden
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, P.O. Box 456, 38200 La Laguna, Spain
| |
Collapse
|
22
|
Gu L, Huang R, Ni N, Gu P, Fan X. Advances and Prospects in Materials for Craniofacial Bone Reconstruction. ACS Biomater Sci Eng 2023; 9:4462-4496. [PMID: 37470754 DOI: 10.1021/acsbiomaterials.3c00399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
The craniofacial region is composed of 23 bones, which provide crucial function in keeping the normal position of brain and eyeballs, aesthetics of the craniofacial complex, facial movements, and visual function. Given the complex geometry and architecture, craniofacial bone defects not only affect the normal craniofacial structure but also may result in severe craniofacial dysfunction. Therefore, the exploration of rapid, precise, and effective reconstruction of craniofacial bone defects is urgent. Recently, developments in advanced bone tissue engineering bring new hope for the ideal reconstruction of the craniofacial bone defects. This report, presenting a first-time comprehensive review of recent advances of biomaterials in craniofacial bone tissue engineering, overviews the modification of traditional biomaterials and development of advanced biomaterials applying to craniofacial reconstruction. Challenges and perspectives of biomaterial development in craniofacial fields are discussed in the end.
Collapse
Affiliation(s)
- Li Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Rui Huang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ni Ni
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
23
|
Zhao W, Tu H, Chen J, Wang J, Liu H, Zhang F, Li J. Functionalized hydrogels in neural injury repairing. Front Neurosci 2023; 17:1199299. [PMID: 37404462 PMCID: PMC10315583 DOI: 10.3389/fnins.2023.1199299] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/27/2023] [Indexed: 07/06/2023] Open
Abstract
Repairing injuries to the nervous system has always been a prominent topic in clinical research. Direct suturing and nerve displacement surgery are the primary treatment options, but they may not be suitable for long nerve injuries and may require sacrificing the functionality of other autologous nerves. With the emergence of tissue engineering, hydrogel materials have been identified as a promising technology with clinical translation potential for repairing nervous system injuries due to their excellent biocompatibility and ability to release or deliver functional ions. By controlling their composition and structure, hydrogels can be Functionalized and almost fully matched with nerve tissue and even simulate nerve conduction function and mechanical properties. Thus, they are suitable for repairing injuries to both the central and peripheral nervous systems. This article provides a review of recent research progress in functionalized hydrogels for nerve injury repair, highlighting the design differences among various materials and future research directions. We strongly believe that the development of functionalized hydrogels has great potential for improving the clinical treatment of nerve injuries.
Collapse
Affiliation(s)
- Wenqian Zhao
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Hui Tu
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jianxiao Chen
- Department of Nephrology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jing Wang
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Haoting Liu
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Fengshou Zhang
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jing Li
- Office of Science and Technology, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
24
|
Chen A, Deng S, Lai J, Li J, Chen W, Varma SN, Zhang J, Lei C, Liu C, Huang L. Hydrogels for Oral Tissue Engineering: Challenges and Opportunities. Molecules 2023; 28:3946. [PMID: 37175356 PMCID: PMC10179962 DOI: 10.3390/molecules28093946] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/20/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Oral health is crucial to daily life, yet many people worldwide suffer from oral diseases. With the development of oral tissue engineering, there is a growing demand for dental biomaterials. Addressing oral diseases often requires a two-fold approach: fighting bacterial infections and promoting tissue growth. Hydrogels are promising tissue engineering biomaterials that show great potential for oral tissue regeneration and drug delivery. In this review, we present a classification of hydrogels commonly used in dental research, including natural and synthetic hydrogels. Furthermore, recent applications of these hydrogels in endodontic restorations, periodontal tissues, mandibular and oral soft tissue restorations, and related clinical studies are also discussed, including various antimicrobial and tissue growth promotion strategies used in the dental applications of hydrogels. While hydrogels have been increasingly studied in oral tissue engineering, there are still some challenges that need to be addressed for satisfactory clinical outcomes. This paper summarizes the current issues in the abovementioned application areas and discusses possible future developments.
Collapse
Affiliation(s)
- Anfu Chen
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
- Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London, Royal National Orthopaedic Hospital, London HA4 4LP, UK
| | - Shuhua Deng
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
| | - Jindi Lai
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
| | - Jing Li
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
| | - Weijia Chen
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
| | - Swastina Nath Varma
- Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London, Royal National Orthopaedic Hospital, London HA4 4LP, UK
| | - Jingjing Zhang
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
| | - Caihong Lei
- Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China; (A.C.)
| | - Chaozong Liu
- Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London, Royal National Orthopaedic Hospital, London HA4 4LP, UK
| | - Lijia Huang
- Guangdong Provincial Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
25
|
Su D, Swearson S, Krongbaramee T, Sun H, Hong L, Amendt BA. Exploring microRNAs in craniofacial regenerative medicine. Biochem Soc Trans 2023; 51:841-854. [PMID: 37073783 PMCID: PMC11244734 DOI: 10.1042/bst20221448] [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: 11/14/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023]
Abstract
microRNAs (miRs) have been reported over the decades as important regulators in bone development and bone regeneration. They play important roles in maintaining the stem cell signature as well as regulating stem cell fate decisions. Thus, delivering miRs and miR inhibitors to the defect site is a potential treatment towards craniofacial bone defects. However, there are challenges in translation of basic research to clinics, including the efficiency, specificity, and efficacy of miR manipulation methods and the safety of miR delivery systems. In this review, we will compare miR oligonucleotides, mimics and antagomirs as therapeutic reagents to treat disease and regenerate tissues. Newer technology will be discussed as well as the efficiency and efficacy of using these technologies to express or inhibit miRs in treating and repairing oral tissues. Delivery of these molecules using extracellular vesicles and nanoparticles can achieve different results and depending on their composition will elicit specific effects. We will highlight the specificity, toxicity, stability, and effectiveness of several miR systems in regenerative medicine.
Collapse
Affiliation(s)
- Dan Su
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
| | - Samuel Swearson
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
| | - Tadkamol Krongbaramee
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
- Division of Endodontics, Department of Restorative Dentistry & Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Hongli Sun
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Liu Hong
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| |
Collapse
|
26
|
Chen H, Qiu X, Xia T, Li Q, Wen Z, Huang B, Li Y. Mesoporous Materials Make Hydrogels More Powerful in Biomedicine. Gels 2023; 9:gels9030207. [PMID: 36975656 PMCID: PMC10048667 DOI: 10.3390/gels9030207] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/12/2023] Open
Abstract
Scientists have been attempting to improve the properties of mesoporous materials and expand their application since the 1990s, and the combination with hydrogels, macromolecular biological materials, is one of the research focuses currently. Uniform mesoporous structure, high specific surface area, good biocompatibility, and biodegradability make the combined use of mesoporous materials more suitable for the sustained release of loaded drugs than single hydrogels. As a joint result, they can achieve tumor targeting, tumor environment stimulation responsiveness, and multiple therapeutic platforms such as photothermal therapy and photodynamic therapy. Due to the photothermal conversion ability, mesoporous materials can significantly improve the antibacterial ability of hydrogels and offer a novel photocatalytic antibacterial mode. In bone repair systems, mesoporous materials remarkably strengthen the mineralization and mechanical properties of hydrogels, aside from being used as drug carriers to load and release various bioactivators to promote osteogenesis. In hemostasis, mesoporous materials greatly elevate the water absorption rate of hydrogels, enhance the mechanical strength of the blood clot, and dramatically shorten the bleeding time. As for wound healing and tissue regeneration, incorporating mesoporous materials can be promising for enhancing vessel formation and cell proliferation of hydrogels. In this paper, we introduce the classification and preparation methods of mesoporous material-loaded composite hydrogels and highlight the applications of composite hydrogels in drug delivery, tumor therapy, antibacterial treatment, osteogenesis, hemostasis, and wound healing. We also summarize the latest research progress and point out future research directions. After searching, no research reporting these contents was found.
Collapse
Affiliation(s)
- Huangqin Chen
- Department of Stomatology, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xin Qiu
- Department of Stomatology, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Tian Xia
- Department of Stomatology, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Qing Li
- Department of Stomatology, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhehan Wen
- Department of Stomatology, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Bin Huang
- Department of Stomatology, School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
- Correspondence: (B.H.); (Y.L.)
| | - Yuesheng Li
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, Non-Power Nuclear Technology Collaborative Innovation Center, Hubei University of Science and Technology, Xianning 437100, China
- Correspondence: (B.H.); (Y.L.)
| |
Collapse
|
27
|
Guo J, Yao H, Li X, Chang L, Wang Z, Zhu W, Su Y, Qin L, Xu J. Advanced Hydrogel systems for mandibular reconstruction. Bioact Mater 2023; 21:175-193. [PMID: 36093328 PMCID: PMC9413641 DOI: 10.1016/j.bioactmat.2022.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/16/2022] [Accepted: 08/02/2022] [Indexed: 12/23/2022] Open
Abstract
Mandibular defect becomes a prevalent maxillofacial disease resulting in mandibular dysfunctions and huge psychological burdens to the patients. Considering the routine presence of oral contaminations and aesthetic restoration of facial structures, the current clinical treatments are however limited, incapable to reconstruct the structural integrity and regeneration, spurring the need for cost-effective mandibular tissue engineering. Hydrogel systems possess great merit for mandibular reconstruction with precise involvement of cells and bioactive factors. In this review, current clinical treatments and distinct mode(s) of mandible formation and pathological resorption are summarized, followed by a review of hydrogel-related mandibular tissue engineering, and an update on the advanced fabrication of hydrogels with improved mechanical property, antibacterial ability, injectable form, and 3D bioprinted hydrogel constructs. The exploration of advanced hydrogel systems will lay down a solid foundation for a bright future with more biocompatible, effective, and personalized treatment in mandibular reconstruction.
Collapse
Affiliation(s)
- Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zixuan Wang
- Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Wangyong Zhu
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yuxiong Su
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Corresponding author. Director of Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Corresponding author. Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
28
|
Wang X, Ma Y, Chen J, Liu Y, Liu G, Wang P, Wang B, Taketo MM, Bellido T, Tu X. A novel decellularized matrix of Wnt signaling-activated osteocytes accelerates the repair of critical-sized parietal bone defects with osteoclastogenesis, angiogenesis, and neurogenesis. Bioact Mater 2023; 21:110-128. [PMID: 36093329 PMCID: PMC9411072 DOI: 10.1016/j.bioactmat.2022.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/21/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cell source is the key to decellularized matrix (DM) strategy. This study compared 3 cell types, osteocytes with/without dominant active Wnt/β-catenin signaling (daCO and WTO) and bone marrow stromal cells (BMSCs) for their DMs in bone repair. Decellularization removes all organelles and >95% DNA, and retained >74% collagen and >71% GAG, maintains the integrity of cell basement membrane with dense boundaries showing oval and honeycomb structure in osteocytic DM and smooth but irregular shape in the BMSC-DM. DM produced higher cell survival rate (90%) and higher proliferative activity. In vitro, daCO-DM induces more and longer stress fibers in BMSCs, conducive to cell adhesion, spreading, and osteogenic differentiation. 8-wk after implantation of the critical-sized parietal bone defect model, daCO-DM formed tight structures, composed of a large number of densely-arranged type-I collagen under polarized light microscope, which is similar to and integrated with host bone. BV/TV (>54%) was 1.5, 2.9, and 3.5 times of WTO-DM, BMSC-DM, and none-DM groups, and N.Ob/T.Ar (3.2 × 102/mm2) was 1.7, 2.9, and 3.3 times. At 4-wk, daCO-DM induced osteoclastogenesis, 2.3 times higher than WTO-DM; but BMSC-DM or none-DM didn't. daCO-DM increased the expression of RANKL and MCSF, Vegfa and Angpt1, and Ngf in BMSCs, which contributes to osteoclastogenesis, angiogenesis, and neurogenesis, respectively. daCO-DM promoted H-type vessel formation and nerve markers β3-tubulin and NeuN expression. Conclusion: daCO-DM produces metabolic and neurovascularized organoid bone to accelerate the repair of bone defects. These features are expected to achieve the effect of autologous bone transplantation, suitable for transformation application. Decellularized matrix of osteocytes with dominant-active β-catenin (daCO-DM) promotes osteogenesis for regenerative repair. daCO-DM induces BMSCs to form stress fibers, conducive to cell adhesion, spreading, and differentiation towards osteoblasts. daCO-DM-induced osteoblasts have strong activity secreting dense and orderly-arranged type I collagen as host bone’s. daCO-DM induces BMSCs to express pre-osteoclastogenic cytokine RANKL and MCSF for osteoclastogenesis of marrow monocytes. daCO-DM enhances BMSCs to express angiogenic Vegfa and Angpt1, and neurogenic Ngf potentially for neurovascularization.
Collapse
Affiliation(s)
- Xiaofang Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Yufei Ma
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Chen
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Yujiao Liu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Guangliang Liu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Pengtao Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Bo Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Makoto M. Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Teresita Bellido
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, 72223, USA
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Corresponding author. Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
29
|
Chen Y, Zhang C. Role of noncoding RNAs in orthodontic tooth movement: new insights into periodontium remodeling. J Transl Med 2023; 21:101. [PMID: 36759852 PMCID: PMC9912641 DOI: 10.1186/s12967-023-03951-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Orthodontic tooth movement (OTM) is biologically based on the spatiotemporal remodeling process in periodontium, the mechanisms of which remain obscure. Noncoding RNAs (ncRNAs), especially microRNAs and long noncoding RNAs, play a pivotal role in maintaining periodontal homeostasis at the transcriptional, post-transcriptional, and epigenetic levels. Under force stimuli, mechanosensitive ncRNAs with altered expression levels transduce mechanical load to modulate intracellular genes. These ncRNAs regulate the biomechanical responses of periodontium in the catabolic, anabolic, and coupling phases throughout OTM. To achieve this, down or upregulated ncRNAs actively participate in cell proliferation, differentiation, autophagy, inflammatory, immune, and neurovascular responses. This review highlights the regulatory mechanism of fine-tuning ncRNAs in periodontium remodeling during OTM, laying the foundation for safe, precise, and personalized orthodontic treatment.
Collapse
Affiliation(s)
- Yuming Chen
- grid.284723.80000 0000 8877 7471Stomatological Hospital, Southern Medical University, Guangzhou, 510280 China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
30
|
Liu L, Wu D, Tu H, Cao M, Li M, Peng L, Yang J. Applications of Hydrogels in Drug Delivery for Oral and Maxillofacial Diseases. Gels 2023; 9:gels9020146. [PMID: 36826316 PMCID: PMC9956178 DOI: 10.3390/gels9020146] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023] Open
Abstract
Oral and maxillofacial diseases have an important impact on local function, facial appearance, and general health. As a multifunctional platform, hydrogels are widely used in the biomedical field due to their excellent physicochemical properties. In recent years, a large number of studies have been conducted to adapt hydrogels to the complex oral and maxillofacial environment by modulating their pore size, swelling, degradability, stimulus-response properties, etc. Meanwhile, many studies have attempted to use hydrogels as drug delivery carriers to load drugs, cytokines, and stem cells for antibacterial, anticancer, and tissue regeneration applications in oral and maxillofacial regions. This paper reviews the application and research progress of hydrogel-based drug delivery systems in the treatment of oral and maxillofacial diseases such as caries, endodontic diseases, periodontal diseases, maxillofacial bone diseases, mucosal diseases, oral cancer, etc. The characteristics and applications of hydrogels and drug-delivery systems employed for the treatment of different diseases are discussed in order to provide a reference for further research on hydrogel drug-delivery systems in the future.
Collapse
Affiliation(s)
- Lijia Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Heng Tu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mengjiao Cao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mengxin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Li Peng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence:
| |
Collapse
|
31
|
Sun S, Cui Y, Yuan B, Dou M, Wang G, Xu H, Wang J, Yin W, Wu D, Peng C. Drug delivery systems based on polyethylene glycol hydrogels for enhanced bone regeneration. Front Bioeng Biotechnol 2023; 11:1117647. [PMID: 36793443 PMCID: PMC9923112 DOI: 10.3389/fbioe.2023.1117647] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Drug delivery systems composed of osteogenic substances and biological materials are of great significance in enhancing bone regeneration, and appropriate biological carriers are the cornerstone for their construction. Polyethylene glycol (PEG) is favored in bone tissue engineering due to its good biocompatibility and hydrophilicity. When combined with other substances, the physicochemical properties of PEG-based hydrogels fully meet the requirements of drug delivery carriers. Therefore, this paper reviews the application of PEG-based hydrogels in the treatment of bone defects. The advantages and disadvantages of PEG as a carrier are analyzed, and various modification methods of PEG hydrogels are summarized. On this basis, the application of PEG-based hydrogel drug delivery systems in promoting bone regeneration in recent years is summarized. Finally, the shortcomings and future developments of PEG-based hydrogel drug delivery systems are discussed. This review provides a theoretical basis and fabrication strategy for the application of PEG-based composite drug delivery systems in local bone defects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dankai Wu
- Orthopaedic Medical Center, Second Hospital of Jilin University, Changchun, China
| | - Chuangang Peng
- Orthopaedic Medical Center, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
The recent advancement in the PLGA-based thermo-sensitive hydrogel for smart drug delivery. Int J Pharm 2023; 631:122484. [PMID: 36509221 DOI: 10.1016/j.ijpharm.2022.122484] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
To date, hydrogels have opened new prospects for potential applications for drug delivery. The thermo-sensitive hydrogels have the great potential to provide more effective and controllable release of therapeutic/bioactive agents in response to changes in temperature. PLGA is a safe FDA-approved copolymer with good biocompatibility and biodegradability. Recently, PLGA-based formulation have attracted a lot of interest for thermo-sensitive hydrogels. Thermo-sensitive PLGA-based hydrogels provide the delivery system with good spatial and temporal control, and have been widely applied in drug delivery. This review is focused on the recent progression of the thermo-sensitive and biodegradable PLGA-based hydrogels that have been reported for smart drug delivery to the different organs. Eventually, future perspectives and challenges of thermo-sensitive PLGA-based hydrogels are discussed briefly.
Collapse
|
33
|
Bashir MH, Korany NS, Farag DBE, Abbass MMS, Ezzat BA, Hegazy RH, Dörfer CE, Fawzy El-Sayed KM. Polymeric Nanocomposite Hydrogel Scaffolds in Craniofacial Bone Regeneration: A Comprehensive Review. Biomolecules 2023; 13:biom13020205. [PMID: 36830575 PMCID: PMC9953024 DOI: 10.3390/biom13020205] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
Nanocomposite biomaterials combine a biopolymeric matrix structure with nanoscale fillers. These bioactive and easily resorbable nanocomposites have been broadly divided into three groups, namely natural, synthetic or composite, based on the polymeric origin. Preparing such nanocomposite structures in the form of hydrogels can create a three-dimensional natural hydrophilic atmosphere pivotal for cell survival and new tissue formation. Thus, hydrogel-based cell distribution and drug administration have evolved as possible options for bone tissue engineering and regeneration. In this context, nanogels or nanohydrogels, created by cross-linking three-dimensional polymer networks, either physically or chemically, with high biocompatibility and mechanical properties were introduced as promising drug delivery systems. The present review highlights the potential of hydrogels and nanopolymers in the field of craniofacial tissue engineering and bone regeneration.
Collapse
Affiliation(s)
- Maha H. Bashir
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Nahed S. Korany
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Dina B. E. Farag
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Marwa M. S. Abbass
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Bassant A. Ezzat
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Radwa H. Hegazy
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 11553, Egypt
- Correspondence: ; Tel.: +49-431-500-26210
| |
Collapse
|
34
|
RANDHAWA AAYUSHI, DEB DUTTA SAYAN, GANGULY KEYA, V. PATIL TEJAL, LUTHFIKASARI RACHMI, LIM KITAEK. Understanding cell-extracellular matrix interactions for topology-guided tissue regeneration. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
35
|
Zhou Y, Yang Y, Liu R, Zhou Q, Lu H, Zhang W. Research Progress of Polydopamine Hydrogel in the Prevention and Treatment of Oral Diseases. Int J Nanomedicine 2023; 18:2623-2645. [PMID: 37213351 PMCID: PMC10199686 DOI: 10.2147/ijn.s407044] [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: 02/18/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
Oral diseases represent one of the most prevalent diseases globally and are associated with serious health and economic burdens, greatly altering the quality of life of affected individuals. Various biomaterials play important roles in the treatment of oral diseases. To some extent, the development of biomaterials has promoted progress in clinically available oral medicines. Hydrogels have unique tunable advantages that make them useful in the next generation of regenerative strategies and have been widely applied in both oral soft and hard tissues repair. However, most hydrogels lack self-adhesive properties, which may result in low repair efficacy. Polydopamine (PDA), the primary adhesive component, has attracted increasing attention in recent years. PDA-modified hydrogels exhibit reliable and suitable adherence to tissues and easily integrate into tissues to promote repair efficiency. This paper reviews the latest research progress on PDA hydrogels and elaborates on the mechanism of the reaction between PDA functional groups and hydrogels, and summarizes the biological properties and the applications of PDA hydrogels in the prevention and treatment of the field of oral diseases. It is also proposed that in future research we should simulate the complex microenvironment of the oral cavity as much as possible, coordinate and plan various biological events rationally, and realize the translation from scientific research to clinical practice.
Collapse
Affiliation(s)
- Yuqi Zhou
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuanmeng Yang
- Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Rongpu Liu
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Qin Zhou
- Department of Oral Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Haixia Lu
- Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Haixia Lu, Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China, Email
| | - Wenjie Zhang
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Wenjie Zhang, Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China, Email
| |
Collapse
|
36
|
Wang J, Cui Y, Liu H, Li S, Sun S, Xu H, Peng C, Wang Y, Wu D. MicroRNA-loaded biomaterials for osteogenesis. Front Bioeng Biotechnol 2022; 10:952670. [PMID: 36199361 PMCID: PMC9527286 DOI: 10.3389/fbioe.2022.952670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
The large incidence of bone defects in clinical practice increases not only the demand for advanced bone transplantation techniques but also the development of bone substitute materials. A variety of emerging bone tissue engineering materials with osteogenic induction ability are promising strategies for the design of bone substitutes. MicroRNAs (miRNAs) are a class of non-coding RNAs that regulate intracellular protein expression by targeting the non-coding region of mRNA3′-UTR to play an important role in osteogenic differentiation. Several miRNA preparations have been used to promote the osteogenic differentiation of stem cells. Therefore, multiple functional bone tissue engineering materials using miRNA as an osteogenic factor have been developed and confirmed to have critical efficacy in promoting bone repair. In this review, osteogenic intracellular signaling pathways mediated by miRNAs are introduced in detail to provide a clear understanding for future clinical treatment. We summarized the biomaterials loaded with exogenous cells engineered by miRNAs and biomaterials directly carrying miRNAs acting on endogenous stem cells and discussed their advantages and disadvantages, providing a feasible method for promoting bone regeneration. Finally, we summarized the current research deficiencies and future research directions of the miRNA-functionalized scaffold. This review provides a summary of a variety of advanced miRNA delivery system design strategies that enhance bone regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dankai Wu
- *Correspondence: Yanbing Wang, ; Dankai Wu,
| |
Collapse
|
37
|
Zhang Z, Hao Z, Xian C, Fang Y, Cheng B, Wu J, Xia J. Neuro-bone tissue engineering: Multiple potential translational strategies between nerve and bone. Acta Biomater 2022; 153:1-12. [PMID: 36116724 DOI: 10.1016/j.actbio.2022.09.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/01/2022]
Abstract
Numerous tissue regeneration paradigms show evident neurological dependence, including mammalian fingertip, skin, and bone regeneration. The mature skeleton is innervated by an abundant nervous system that infiltrates the developing axial and appendicular bones and maintains the stability of the systemic skeletal system by controlling blood flow, regulating bone metabolism, secreting neurotransmitters, and regulating stem cell behavior. In recent years, neurotization in tissue-engineered bone has been considered as a promising strategy to effectively overcome the challenge of vascularization and innervation regeneration in the central zone of "critical-sized bone defects" that conventional tissue-engineered scaffolds are unable to handle, however, further validation is needed in relevant clinical applications. Therefore, this study reviews the mechanisms by which the nervous system regulates bone metabolism and regeneration through a variety of neurogenic or non-neurogenic factors, as well as the recent progress and design strategies of neuralized tissue-engineered bone, to provide new ideas for further studies on subsequent neural bone tissue engineering. STATEMENT OF SIGNIFICANCE: The interaction of nerve and bone tissue during skeletal development and repair has attracted widespread attention, with emerging evidences highlighting the regulation of bone metabolism and regeneration by the nervous system, but the underlying mechanisms have not been elucidated. Thus, further applications of neuro-bone tissue engineering still needs careful consideration. In this review, we summarize the numerous neurogenic and non-neurogenic factors which are involved in bone repair and regeneration, and further explore the current status of their application and biomaterial design in neuro-bone tissue engineering, and finally discuss the challenge and prospective for neuro-bone tissue engineering to facilitate its further development.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhichao Hao
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, China
| | - Caihong Xian
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Yifen Fang
- Department of Cardiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, China.
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, China.
| |
Collapse
|
38
|
Ansari MJ, Rajendran RR, Mohanto S, Agarwal U, Panda K, Dhotre K, Manne R, Deepak A, Zafar A, Yasir M, Pramanik S. Poly( N-isopropylacrylamide)-Based Hydrogels for Biomedical Applications: A Review of the State-of-the-Art. Gels 2022; 8:454. [PMID: 35877539 PMCID: PMC9323937 DOI: 10.3390/gels8070454] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/21/2022] Open
Abstract
A prominent research topic in contemporary advanced functional materials science is the production of smart materials based on polymers that may independently adjust their physical and/or chemical characteristics when subjected to external stimuli. Smart hydrogels based on poly(N-isopropylacrylamide) (PNIPAM) demonstrate distinct thermoresponsive features close to a lower critical solution temperature (LCST) that enhance their capability in various biomedical applications such as drug delivery, tissue engineering, and wound dressings. Nevertheless, they have intrinsic shortcomings such as poor mechanical properties, limited loading capacity of actives, and poor biodegradability. Formulation of PNIPAM with diverse functional constituents to develop hydrogel composites is an efficient scheme to overcome these defects, which can significantly help for practicable application. This review reports on the latest developments in functional PNIPAM-based smart hydrogels for various biomedical applications. The first section describes the properties of PNIPAM-based hydrogels, followed by potential applications in diverse fields. Ultimately, this review summarizes the challenges and opportunities in this emerging area of research and development concerning this fascinating polymer-based system deep-rooted in chemistry and material science.
Collapse
Affiliation(s)
- Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Rahul R. Rajendran
- Department of Mechanical Engineering and Mechanics, Lehigh University, 19 Memorial Drive West, Bethlehem, PA 18015, USA;
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College and Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - Unnati Agarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar-Delhi, Grand Trunk Road, Phagwara 144001, Punjab, India;
| | - Kingshuk Panda
- Department of Applied Microbiology, Vellore Institute of Technology, School of Bioscience and Technology, Vellore 632014, Tamilnadu, India;
| | - Kishore Dhotre
- I.C.M.R.—National Institute of Virology, Pune 411021, Maharashtra, India;
| | - Ravi Manne
- Chemtex Environmental Lab, Quality Control and Assurance Department, 3082 25th Street, Port Arthur, TX 77642, USA;
| | - A. Deepak
- Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 600124, Tamil Nadu, India;
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia; or
| | - Mohd Yasir
- Department of Pharmacy, College of Health Science, Arsi University, Asella 396, Ethiopia;
| | - Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| |
Collapse
|
39
|
Vallet-Regí M, Schüth F, Lozano D, Colilla M, Manzano M. Engineering mesoporous silica nanoparticles for drug delivery: where are we after two decades? Chem Soc Rev 2022; 51:5365-5451. [PMID: 35642539 PMCID: PMC9252171 DOI: 10.1039/d1cs00659b] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Collapse
Affiliation(s)
- María Vallet-Regí
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ferdi Schüth
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Daniel Lozano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Montserrat Colilla
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
40
|
Guan S, Zhang Z, Wu J. Non-coding RNA delivery for bone tissue engineering: progress, challenges and potential solutions. iScience 2022; 25:104807. [PMID: 35992068 PMCID: PMC9385673 DOI: 10.1016/j.isci.2022.104807] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
More than 20 million individuals worldwide suffer from congenital or acquired bone defects annually. The development of bone scaffold materials that simulate natural bone for bone defect repair remains challenging. Recently, ncRNA-based therapies for bone defects have attracted increasing interest because of the great potential of ncRNAs in disease treatment. Various types of ncRNAs regulate gene expression in osteogenesis-related cells via multiple mechanisms. The delivery of ncRNAs to the site of bone loss through gene vectors or scaffolds is a potential therapeutic option for bone defect repair. Therefore, this study discusses and summarizes the regulatory mechanisms of miRNAs, siRNAs, and piRNAs in osteogenic signaling and reviews the widely used current RNA delivery vectors and scaffolds for bone defect repair. Additionally, current challenges and potential solutions of delivery scaffolds for bone defect repair are proposed, with the aim of providing a theoretical basis for their future clinical applications.
Collapse
|
41
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
42
|
Chemically engineered mesoporous silica nanoparticles-based intelligent delivery systems for theranostic applications in multiple cancerous/non-cancerous diseases. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214309] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Huang M, Huang Y, LIU H, Tang Z, Chen Y, Huang Z, Xu S, Du J, Jia B. Hydrogels for Treatment of Oral and Maxillofacial Diseases: Current Research, Challenge, and Future Directions. Biomater Sci 2022; 10:6413-6446. [DOI: 10.1039/d2bm01036d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Oral and maxillofacial diseases such as infection and trauma often involve various organs and tissues, resulting in structural defects, dysfunctions and/or adverse effects on facial appearance. Hydrogels have been applied...
Collapse
|
44
|
Mellati A, Hasanzadeh E, Gholipourmalekabadi M, Enderami SE. Injectable nanocomposite hydrogels as an emerging platform for biomedical applications: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112489. [PMID: 34857275 DOI: 10.1016/j.msec.2021.112489] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022]
Abstract
Hydrogels have attracted much attention for biomedical and pharmaceutical applications due to the similarity of their biomimetic structure to the extracellular matrix of natural living tissues, tunable soft porous microarchitecture, superb biomechanical properties, proper biocompatibility, etc. Injectable hydrogels are an exciting type of hydrogels that can be easily injected into the target sites using needles or catheters in a minimally invasive manner. The more comfortable use, less pain, faster recovery period, lower costs, and fewer side effects make injectable hydrogels more attractive to both patients and clinicians in comparison to non-injectable hydrogels. However, it is difficult to achieve an ideal injectable hydrogel using just a single material (i.e., polymer). This challenge can be overcome by incorporating nanofillers into the polymeric matrix to engineer injectable nanocomposite hydrogels with combined or synergistic properties gained from the constituents. This work aims to critically review injectable nanocomposite hydrogels, their preparation methods, properties, functionalities, and versatile biomedical and pharmaceutical applications such as tissue engineering, drug delivery, and cancer labeling and therapy. The most common natural and synthetic polymers as matrices together with the most popular nanomaterials as reinforcements, including nanoceramics, carbon-based nanostructures, metallic nanomaterials, and various nanosized polymeric materials, are highlighted in this review.
Collapse
Affiliation(s)
- Amir Mellati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Elham Hasanzadeh
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Ehsan Enderami
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
45
|
Wang L, Jin Y, Wu L, Zhong T. Hybrid colloidal gels assembled from inorganic and polymeric nanoparticles as a drug-delivery platform. Chem Phys Lett 2021. [DOI: 10.1016/j.cplett.2021.139122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
46
|
Recent advancements and future submissions of silica core-shell nanoparticles. Int J Pharm 2021; 609:121173. [PMID: 34627997 DOI: 10.1016/j.ijpharm.2021.121173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022]
Abstract
The core-shell silica-based nanoparticles (CSNPs) possess outstanding properties for developing next-generation therapeutics. CSNPs provide greater surface area owing to their mesoporous structure, which offers a high opportunity for surface modification. This review highlights the potential of core-shell silica-based nanoparticle (CSNP) based injectable nanotherapeutics (INT); its role in drug delivery, biomedical imaging, light-triggered phototherapy, Plasmonic enhancers, gene delivery, magnetic hyperthermia, immunotherapy, and potential as next-generation theragnostic. Specifically, the conceptual crosstalk on modern synthetic strategies, biodistribution profiles with a mechanistic view on the therapeutics loading and release modeling are dealt in detail. The manuscript also converses the challenges associated with CSNPs, regulatory hurdles, and their current market position.
Collapse
|
47
|
Activated Carbon Fiber Cloth/Biomimetic Apatite: A Dual Drug Delivery System. Int J Mol Sci 2021; 22:ijms222212247. [PMID: 34830128 PMCID: PMC8624510 DOI: 10.3390/ijms222212247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 02/07/2023] Open
Abstract
A biomaterial that is both bioactive and capable of controlled drug release is highly attractive for bone regeneration. In previous works, we demonstrated the possibility of combining activated carbon fiber cloth (ACC) and biomimetic apatite (such as calcium-deficient hydroxyapatite (CDA)) to develop an efficient material for bone regeneration. The aim to use the adsorption properties of an activated carbon/biomimetic apatite composite to synthetize a biomaterial to be used as a controlled drug release system after implantation. The adsorption and desorption of tetracycline and aspirin were first investigated in the ACC and CDA components and then on ACC/CDA composite. The results showed that drug adsorption and release are dependent on the adsorbent material and the drug polarity/hydrophilicity, leading to two distinct modes of drug adsorption and release. Consequently, a double adsorption approach was successfully performed, leading to a multifunctional and innovative ACC-aspirin/CDA-tetracycline implantable biomaterial. In a second step, in vitro tests emphasized a better affinity of the drug (tetracycline or aspirin)-loaded ACC/CDA materials towards human primary osteoblast viability and proliferation. Then, in vivo experiments on a large cortical bone defect in rats was carried out to test biocompatibility and bone regeneration ability. Data clearly highlighted a significant acceleration of bone reconstruction in the presence of the ACC/CDA patch. The ability of the aspirin-loaded ACC/CDA material to release the drug in situ for improving bone healing was also underlined, as a proof of concept. This work highlights the possibility of bone patches with controlled (multi)drug release features being used for bone tissue repair.
Collapse
|
48
|
Wang H, Zhang Y, Xu X, Wang A. An injectable mesoporous silica-based analgesic delivery system prolongs the duration of sciatic nerve block in mice with minimal toxicity. Acta Biomater 2021; 135:638-649. [PMID: 34520884 DOI: 10.1016/j.actbio.2021.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 12/16/2022]
Abstract
The major limitation of traditional local anesthetics is the finite duration of a single injection. The present study developed two kinds of novel injectable anesthetic nanocomposites based on mesoporous silica, and evaluated their long-lasting analgesic effect and biosafety. The nanoparticulate carriers, mesoporous silica nanoparticles (MSNs) and mesoporous silica-coated gold nanorods (GNR@MSN), were firstly constructed using the oil-water biphase reaction approach and then ropivacaine (RPC), a local anesthetic, was loaded into the mesoporous carriers by vacuum suction. Transmission electron microscopic images showed the well-ordered mesoporous structure for drug loading. RPC-loaded MSNs and RPC-loaded GNR@MSN exhibited a sustained-release pattern in vitro, and the latter also showed a controlled-release manner triggered by near-infrared (NIR) irradiation. RPC-loaded MSNs and RPC-loaded GNR@MSN caused an initial sensory blockade in mice that lasted for 6 h, almost 2.5 folds of that from free RPC solution. Furthermore, upon NIR irradiation, the latter induced three additional periods of the blockade. Neither of them showed motor nerve block, which may be due to the sustained release manner. The low myotoxicity and low neurotoxicity of the two nanocomposites were presented both in vitro and in vivo. These results demonstrate the potential of the mesoporous silica-based analgesic nanocomposites in effectively controlling postoperative pain, maybe RPC-loaded MSNs for moderate pain and RPC-loaded GNR@MSN for severe pain. STATEMENT OF SIGNIFICANCE: Adequate postoperative analgesia helps early functional exercise after surgery and accelerates rapid recovery, while uncontrolled postoperative pain probably develops chronic post-surgical pain that impacts the life quality of patients for a long time. However, postoperative pain management is still a challenge. The current treatment drugs are always accompanied by some side effects due to their systemic effect. Opioids have risks of addiction and respiratory depression, and nonsteroidal anti-inflammatory drugs can lead to gastrointestinal reaction. Therefore, the long-lasting local anesthetic formulation with good biocompatibility is the most promising solution to manage post-surgical pain. The present study developed novel injectable anesthetic nanocomposites based on mesoporous silica, providing long-lasting pain relief in mice with minimal toxicity.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China
| | - Yu Zhang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China
| | - Xiaotao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China.
| |
Collapse
|
49
|
Zhou XP, Li QW, Shu ZZ, Liu Y. TP53-mediated miR-2861 promotes osteogenic differentiation of BMSCs by targeting Smad7. Mol Cell Biochem 2021; 477:283-293. [PMID: 34709507 DOI: 10.1007/s11010-021-04276-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/11/2021] [Indexed: 12/20/2022]
Abstract
Bone defect seriously affects the quality of life. Meanwhile, osteogenic differentiation in BMSCs could regulate the progression of bone defect. Transcription factors are known to regulate the osteogenic differentiation in BMSCs. The study aimed to investigate the detailed mechanism by which TP53 regulates the osteogenic differentiation. To study bone defect in vitro, BMSCs were isolated from spinal cord injury rats. CCK-8 assay was applied to test the cell viability. The mineralized nodules in BMSCs was tested by alizarin red staining. Meanwhile, TUNEL staining and flow cytometry were performed to test the cell apoptosis. mRNA expression was tested by qRT-PCR. Starbase and dual-luciferase reporter assay were used to predict the downstream mRNA of miR-2861. Moreover, western blot was applied to detect the protein expressions (TP53 and Smad7). BMSCs were successfully isolated from rats. The expressions of miR-2861 were significantly upregulated in osteogenic medium, compared with growth medium. MiR-2861 inhibitor significantly decreased the levels of OCN, ALP, BSP, and Runx2 in BMSCs. In addition, miR-2861 inhibitor notably inhibited the mineralized nodules, viability, and induced the apoptosis of BMSCs. Smad7 was identified to be the downstream target of miR-2861, and knockdown of Smad7 notably reversed miR-2861 inhibitor-induced inhibition of osteogenic differentiation and promotion of apoptosis in BMSCs. Moreover, miR-2861 was transcriptionally regulated by TP53 in BMSCs. TP53-meidiated miR-2861 promotes osteogenic differentiation of BMSCs by targeting Smad7. Thereby, our research might provide new methods for bone defect treatment.
Collapse
Affiliation(s)
- Xian-Pei Zhou
- Department of Hand and Foot Surgery, Brain Hospital of Hunan Province, No. 427, Section 3 of Furong Middle Road, Changsha, 410007, Hunan Province, China.
| | - Qi-Wei Li
- Department of Hand and Foot Surgery, Brain Hospital of Hunan Province, No. 427, Section 3 of Furong Middle Road, Changsha, 410007, Hunan Province, China
| | - Zi-Zhen Shu
- Department of Hand and Foot Surgery, Brain Hospital of Hunan Province, No. 427, Section 3 of Furong Middle Road, Changsha, 410007, Hunan Province, China
| | - Yang Liu
- Department of Gastrointestinal Surgery, Second Xiangaya Hospital, Central South University, No. 139 Renmin Road, Furong District, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
50
|
Ren Y, Wang X, Liang H, He W, Zhao X. Mechanism of miR-30b-5p-Loaded PEG-PLGA Nanoparticles for Targeted Treatment of Heart Failure. Front Pharmacol 2021; 12:745429. [PMID: 34658880 PMCID: PMC8514665 DOI: 10.3389/fphar.2021.745429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/10/2021] [Indexed: 12/02/2022] Open
Abstract
Objective: Exploring the effectiveness of miR-30b-5p-loaded PEG-PLGA nanoparticles (NPs) for the treatment of heart failure and the underlying mechanism. Methods: PEG-PLGA characteristics with different loading amounts were first examined to determine the loading, encapsulation, and release of miR-30b-5p from NPs. The effects of miR-30b-5p NPs on cardiac function and structure were assessed by immunofluorescence, echocardiography, HE/Masson staining, and TUNEL staining. The effects of NPs on the expression of factors related to cardiac hypertrophy and inflammation were examined by RT-PCR and western blotting, and the mechanism of miR-30b-5p treatment on heart failure was explored by dual luciferase reporter assay and RT-PCR. Results: The size of PEG-PLGA NPs with different loading amounts ranged from 200 to 300 nm, and the zeta potential of PEG-PLGA NPs was negative. The mean entrapment efficiency of the NPs for miR-30b-5p was high (81.8 ± 2.1%), and the release rate reached 5 days with more than 90% release. Distribution experiments showed that NPs were mainly distributed in the heart and had a protective effect on myocardial injury and cardiac function. Compared with a rat model of cardiac failure and miR-30b-5p-non-loaede NP groups, the expression of cardiac hypertrophy markers (ANP, BNPβ-MHC) and inflammatory factors (IL-1β, IL-6) were significantly decreased. Dual luciferase reporter assay assays indicated that miR-30b-5p exerted its effects mainly by targeting TGFBR2. Conclusion: PEG-PLGA NPs loaded with miR-30b-5p improved cardiac function, attenuated myocardial injury, and regulated the expression of factors associated with cardiac hypertrophy and inflammation by targeting TGFBR2.
Collapse
Affiliation(s)
- Yu Ren
- Scientific Research Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Xiao Wang
- Scientific Research Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Hongyu Liang
- Scientific Research Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Wenshuai He
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Xingsheng Zhao
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, China
| |
Collapse
|