1
|
Tang H, Yu Y, Zhan X, Chai Y, Zheng Y, Liu Y, Xia D, Lin H. Zeolite imidazolate framework-8 in bone regeneration: A systematic review. J Control Release 2024; 365:558-582. [PMID: 38042375 DOI: 10.1016/j.jconrel.2023.11.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/19/2023] [Accepted: 11/26/2023] [Indexed: 12/04/2023]
Abstract
Zeolite imidazolate framework-8 (ZIF-8) is a biomaterial that has been increasingly studied in recent years. It has several applications such as bone regeneration, promotion of angiogenesis, drug loading, and antibacterial activity, and exerts multiple effects to deal with various problems in the process of bone regeneration. This systematic review aims to provide an overview of the applications and effectiveness of ZIF-8 in bone regeneration. A search of papers published in the PubMed, Web of Science, Embase, and Cochrane Library databases revealed 532 relevant studies. Title, abstract, and full-text screening resulted in 39 papers being included in the review, including 39 in vitro and 22 animal studies. Appropriate concentrations of nano ZIF-8 can promote cell proliferation and osteogenic differentiation by releasing Zn2+ and entering the cell, whereas high doses of ZIF-8 are cytotoxic and inhibit osteogenic differentiation. In addition, five studies confirmed that ZIF-8 exhibits good vasogenic activity. In all in vivo experiments, nano ZIF-8 promoted bone formation. These results indicate that, at appropriate concentrations, materials containing ZIF-8 promote bone regeneration more than materials without ZIF-8, and with characteristics such as promoting angiogenesis, drug loading, and antibacterial activity, it is expected to show promising applications in the field of bone regeneration. STATEMENT OF SIGNIFICANCE: This manuscript reviewed the use of ZIF-8 in bone regeneration, clarified the biocompatibility and effectiveness in promoting bone regeneration of ZIF-8 materials, and discussed the possible mechanisms and factors affecting its promotion of bone regeneration. Overall, this study provides a better understanding of the latest advances in the field of bone regeneration of ZIF-8, serves as a design guide, and contributes to the design of future experimental studies.
Collapse
Affiliation(s)
- Hao Tang
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yameng Yu
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Xinxin Zhan
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yuan Chai
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| | - Dandan Xia
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| | - Hong Lin
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China.
| |
Collapse
|
2
|
Zhu F, Nie G, Liu C. Engineered biomaterials in stem cell-based regenerative medicine. LIFE MEDICINE 2023; 2:lnad027. [PMID: 39872549 PMCID: PMC11749850 DOI: 10.1093/lifemedi/lnad027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/17/2023] [Indexed: 01/30/2025]
Abstract
Stem cell-based regenerative therapies, which harness the self-renewal and differentiation properties of stem cells, have been in the spotlight due to their widespread applications in treating degenerative, aging, and other, generally intractable diseases. Therapeutically effective hematopoietic stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells have been used in numerous basic and translational studies with exciting results. However, pre-/post-transplantation issues of poor cell survival and retention, uncontrolled differentiation, and insufficient numbers of cells engrafted into host tissues are the major challenges in stem cell-based regenerative therapies. Engineered biomaterials have adjustable biochemical and biophysical properties that significantly affect cell behaviors, such as cell engraftment, survival, migration, and differentiation outcomes, thereby enhancing the engraftment of implanted stem cells and guiding tissue regeneration. Therefore, the combination of stem cell biology with bioengineered materials is a promising strategy to improve the therapeutic outcomes of stem cell-based regenerative therapy. In this review, we summarize the advances in the modulation of behaviors of stem cells via engineered biomaterials. We then present different approaches to harnessing bioengineered materials to enhance the transplantation of stem cells. Finally, we will provide future directions in regenerative therapy using stem cells.
Collapse
Affiliation(s)
- Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
3
|
Cell-Free Therapies: The Use of Cell Extracts to Mitigate Irradiation-Injured Salivary Glands. BIOLOGY 2023; 12:biology12020305. [PMID: 36829582 PMCID: PMC9953449 DOI: 10.3390/biology12020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023]
Abstract
Radiotherapy is a standard treatment for head and neck cancer patients worldwide. However, millions of patients who received radiotherapy consequently suffer from xerostomia because of irreversible damage to salivary glands (SGs) caused by irradiation (IR). Current treatments for IR-induced SG hypofunction only provide temporary symptom alleviation but do not repair the damaged SG, thus resulting in limited treatment efficacy. Therefore, there has recently been a growing interest in regenerative treatments, such as cell-free therapies. This review aims to summarize cell-free therapies for IR-induced SG, with a particular emphasis on utilizing diverse cell extract (CE) administrations. Cell extract is a group of heterogeneous mixtures containing multifunctional inter-cellular molecules. This review discusses the current knowledge of CE's components and efficacy. We propose optimal approaches to improve cell extract treatment from multiple perspectives (e.g., delivery routes, preparation methods, and other details regarding CE administration). In addition, the advantages and limitations of CE treatment are systematically discussed by comparing it to other cell-free (such as conditioned media and exosomes) and cell-based therapies. Although a comprehensive identification of the bioactive factors within CEs and their mechanisms of action have yet to be fully understood, we propose cell extract therapy as an effective, practical, user-friendly, and safe option to conventional therapies in IR-induced SG.
Collapse
|
4
|
Kuroda A, Mineo A, Shoji S, Inoue G, Saito W, Sekiguchi H, Takaso M, Uchida K. Effect of spheroid size on gene expression profiles of a mouse mesenchymal stem cell line in spheroid culture. Biomed Mater Eng 2023; 34:67-76. [PMID: 35694914 DOI: 10.3233/bme-221406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies offer potential for bone repair. MSC spheroid cultures may harbor enhanced therapeutic potential over MSC monolayers through increased secretion of trophic factors. However, the impact of spheroid size on trophic factor expression is unclear. OBJECTIVE We investigated the effect of spheroid size on trophic factor-related gene expression. METHODS KUM10, a murine MSC line was used. RNA-seq was used to screen the transcriptional profiles of MSC monolayer and spheroid cultures. Differentially expressed genes identified in RNA-seq were evaluated by q-PCR in cultures of 5 × 104 (S group), 5 × 105 (M group), 5 × 106 (L group) cells/well. RESULTS Comparison of expression levels between KUM10 monolayer and spheroid cultures identified 2140 differentially expressed genes, of which 1047 were upregulated and 1093 were downregulated in KUM10 spheroids. Among these, 12 upregulated genes (Bmp2, Fgf9, Fgf18, Ngf, Pdgfa, Pdgfb, Tgfb1, Vegfa, Vegfc, Wnt4, Wnt5a, Wnt10a) were associated with secretory growth factors. Of these, expression of Fgf9, Fgf18, Vegfa and Vegfc was elevated in the L group, and Pdgfb and Tgfb1 was elevated in the S group. CONCLUSIONS Spheroid size may impact trophic factor expression. Our results will be useful for future studies assessing the utility of MSC spheroids for treating bone injury.
Collapse
Affiliation(s)
- Akiyoshi Kuroda
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami ku, Kitasato, Sagamihara, Kanagawa, Japan
| | - Ayumi Mineo
- Department of Medical Engineering and Technology, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Shintaro Shoji
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami ku, Kitasato, Sagamihara, Kanagawa, Japan
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami ku, Kitasato, Sagamihara, Kanagawa, Japan
| | - Wataru Saito
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami ku, Kitasato, Sagamihara, Kanagawa, Japan
| | - Hiroyuki Sekiguchi
- Shonan University of Medical Sciences Research Institute, Chigasaki, Kanagawa, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami ku, Kitasato, Sagamihara, Kanagawa, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami ku, Kitasato, Sagamihara, Kanagawa, Japan.,Shonan University of Medical Sciences Research Institute, Chigasaki, Kanagawa, Japan
| |
Collapse
|
5
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
6
|
Zhao X, Li Q, Guo Z, Li Z. Constructing a cell microenvironment with biomaterial scaffolds for stem cell therapy. Stem Cell Res Ther 2021; 12:583. [PMID: 34809719 PMCID: PMC8607654 DOI: 10.1186/s13287-021-02650-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/03/2021] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapy is widely recognized as a promising strategy for exerting therapeutic effects after injury in degenerative diseases. However, limitations such as low cell retention and survival rates after transplantation exist in clinical applications. In recent years, emerging biomaterials that provide a supportable cellular microenvironment for transplanted cells have optimized the therapeutic efficacy of stem cells in injured tissues or organs. Advances in the engineered microenvironment are revolutionizing our understanding of stem cell-based therapies by co-transplanting with synthetic and tissue-derived biomaterials, which offer a scaffold for stem cells and propose an unprecedented opportunity to further employ significant influences in tissue repair and regeneration.
Collapse
Affiliation(s)
- Xiaotong Zhao
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China.,Department of Cardiology, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China. .,Department of Cardiology, Zhengzhou Seventh People's Hospital, Zhengzhou, China.
| | - Zongjin Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China. .,Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
7
|
Habibi H, Suzuki A, Hayashi K, Salimi H, Terai H, Hori Y, Tamai K, Orita K, Ohyama S, Yabu A, Maruf MH, Nakamura H. Expression and function of FGF9 in the hypertrophied ligamentum flavum of lumbar spinal stenosis patients. Spine J 2021; 21:1010-1020. [PMID: 33577925 DOI: 10.1016/j.spinee.2021.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/23/2020] [Accepted: 02/06/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Ligamentum flavum (LF) hypertrophy plays a dominant role in lumbar spinal stenosis (LSS). A previous study found that fibroblast growth factor 9 (FGF9) was upregulated with mechanical stress in rabbit LF. However, the expression and function of FGF9 are not well understood in human LF. PURPOSE To evaluate FGF9 expression and function in human LF with and without hypertrophy. STUDY DESIGN This study employed a basic research study design utilizing human LF tissue for histological analyses. PATIENT SAMPLES Hypertrophied LF tissue sample from patients with LSS, and nonhypertrophied (control) LFs from patients with lumbar disc herniation or other diseases were obtained during surgery. METHODS LF specimens were histologically analyzed for FGF9 and vascular endothelial growth factor A (VEGF-A) by immunohistochemistry. The number of total and FGF9 immuno-positive cells and blood vessels were counted and compared between LF with and without hypertrophy. For functional analysis, the effect of FGF9 on cell proliferation and migration was examined using a primary cell culture of human LF. RESULTS Histological studies revealed that the total cell number was significantly higher in the LF of patients with LSS than in the LF of control patients. Immunohistochemistry showed that the percentage of FGF9-positive cells was significantly higher in the LF of patients with LSS than in the controls, and it positively correlated with patients' age, regardless of disease. Double immune-positive cells for FGF9 and VEGF-A were often observed in vascular endothelial cells and fibroblasts in the fibrotic area of hypertrophied LF, and the number of double positive vessels was significantly higher in LF of LSS patients than in the LF of controls. Primary cell culture of human LF revealed that FGF9 promoted the proliferation and migration of LF cells. CONCLUSION The present study demonstrated that FGF9 expression is highly upregulated in hypertrophied human LF. FGF9 potentially plays a pivotal role in the process of hypertrophy of LF, which is associated with mechanical stress, through cell proliferation and migration. CLINICAL SIGNIFICANCE The results from this study partially reveal the molecular mechanisms of LF hypertrophy and suggest that FGF9 may be involved in the process of LF degeneration in elderly patients.
Collapse
Affiliation(s)
- Hasibullah Habibi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akinobu Suzuki
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - Kazunori Hayashi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hamidullah Salimi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hidetomi Terai
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Hori
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Tamai
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kumi Orita
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shoichiro Ohyama
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akito Yabu
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Mohammad Hasib Maruf
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
8
|
Tran HD, Park KD, Ching YC, Huynh C, Nguyen DH. A comprehensive review on polymeric hydrogel and its composite: Matrices of choice for bone and cartilage tissue engineering. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
9
|
Wan Y, Xu W, Ren X, Wang Y, Dong B, Wang L. Microporous Frameworks as Promising Platforms for Antibacterial Strategies Against Oral Diseases. Front Bioeng Biotechnol 2020; 8:628. [PMID: 32596233 PMCID: PMC7304413 DOI: 10.3389/fbioe.2020.00628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Abstract
Nowadays, the heavy burden of oral diseases such as dental caries, periodontitis, endodontic infections, etc., and their consequences on the patients' quality of life indicate a strong need for developing effective therapies. Bacterial infections played an important role in the field of oral diseases, in-depth insight of such oral diseases have given rise to the demand for antibacterial therapeutic strategies. Recently, microporous frameworks have attracted tremendous interest in antibacterial application due to their well-defined porous structures for drug delivery. In addition, intensive efforts have been made to enhance the antibacterial performance of microporous frameworks, such as ion doping, photosensitizer incorporation as building blocks, and surface modifications. This review article aims on the major recent developments of microporous frameworks for antibacterial applications against oral diseases. The first part of this paper puts concentration on the cutting-edge researches on the versatile antibacterial strategies of microporous materials via drug delivery, inherent activity, and structural modification. The second part discusses the antibacterial applications of microporous frameworks against oral diseases. The applications of microporous frameworks not only have promising therapeutic potential to inhibit bacterial plaque-initiated oral infectious diseases, but also have a wide applicability to other biomedical applications.
Collapse
Affiliation(s)
- Yao Wan
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Changchun, China
| | - Wenzhou Xu
- Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Changchun, China
- Department of Periodontology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Xuan Ren
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Changchun, China
| | - Yu Wang
- Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Changchun, China
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, China
| | - Lin Wang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, Changchun, China
| |
Collapse
|
10
|
Zhang Y, Yu T, Peng L, Sun Q, Wei Y, Han B. Advancements in Hydrogel-Based Drug Sustained Release Systems for Bone Tissue Engineering. Front Pharmacol 2020; 11:622. [PMID: 32435200 PMCID: PMC7218105 DOI: 10.3389/fphar.2020.00622] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Bone defects caused by injury, disease, or congenital deformity remain a major health concern, and efficiently regenerating bone is a prominent clinical demand worldwide. However, bone regeneration is an intricate process that requires concerted participation of both cells and bioactive factors. Mimicking physiological bone healing procedures, the sustained release of bioactive molecules plays a vital role in creating an optimal osteogenic microenvironment and achieving promising bone repair outcomes. The utilization of biomaterial scaffolds can positively affect the osteogenesis process by integrating cells with bioactive factors in a proper way. A high water content, tunable physio-mechanical properties, and diverse synthetic strategies make hydrogels ideal cell carriers and controlled drug release reservoirs. Herein, we reviewed the current advancements in hydrogel-based drug sustained release systems that have delivered osteogenesis-inducing peptides, nucleic acids, and other bioactive molecules in bone tissue engineering (BTE).
Collapse
Affiliation(s)
- Yunfan Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Liying Peng
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Qiannan Sun
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Bing Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
11
|
Samuel RZ, Lei P, Nam K, Baker OJ, Andreadis ST. Engineering the mode of morphogenetic signal presentation to promote branching from salivary gland spheroids in 3D hydrogels. Acta Biomater 2020; 105:121-130. [PMID: 31988042 DOI: 10.1016/j.actbio.2020.01.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/24/2022]
Abstract
Previously we developed a fibrin hydrogel (FH) decorated with laminin-111 peptides (L1p-FH) and supports three-dimensional (3D) gland microstructures containing polarized acinar cells. Here we expand on these results and show that co-culture of rat parotid Par-C10 cells with mesenchymal stem cells produces migrating branches of gland cells into the L1p-FH and we identify FGF-7 as the principal morphogenetic signal responsible for branching. On the other hand, another FGF family member and gland morphogen, FGF-10 increased proliferation but did not promote migration and therefore, limited the number and length of branched structures grown into the gel. By controlling the mode of growth factor presentation and delivery, we can control the length and cellularity of branches as well as formation of new nodes/clusters within the hydrogel. Such spatial delivery of two or more morphogens may facilitate engineering of anatomically complex tissues/mini organs such as salivary glands that can be used to address developmental questions or as platforms for drug discovery. STATEMENT OF SIGNIFICANCE: Hyposalivation leads to the development of a host of oral diseases. Current treatments only provide temporary relief. Tissue engineering may provide promising permanent solutions. Yet current models are limited to salivary spheroids with no branching networks. Branching structures are vital to an effective functioning gland as they increase the surface area/glandular volume ratio of the tissue, allowing a higher output from the small-sized gland. We describe a strategy that controls branch network formation in salivary glands that is a key in advancing the field of salivary gland tissue engineering.
Collapse
|
12
|
Nam K, Dean SM, Brown CT, Smith RJ, Lei P, Andreadis ST, Baker OJ. Synergistic effects of laminin-1 peptides, VEGF and FGF9 on salivary gland regeneration. Acta Biomater 2019; 91:186-194. [PMID: 31028910 DOI: 10.1016/j.actbio.2019.04.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/25/2019] [Accepted: 04/23/2019] [Indexed: 01/19/2023]
Abstract
Hyposalivation is associated with radiation therapy, Sjögren's syndrome and/or aging, and is a significant clinical problem that decreases oral health and overall health in many patients and currently lacks effective treatment. Hence, methods to regenerate salivary glands and restore saliva secretion are urgently needed. To this end, this study describes the modification of fibrin hydrogels with a combination of laminin-1 peptides (YIGSR and A99) and human growth factors (vascular endothelial growth factor and fibroblast growth factor 9) to enhance regeneration in a salivary gland injury mouse model. Our results indicate that these fortified hydrogels enhanced angiogenesis and neurogenesis while promoting formation of acinar structures, thereby leading to enhanced saliva secretion. Such functional recovery indicates salivary gland regeneration and suggests that our technology may be useful in promoting gland regeneration and reversing hyposalivation in a clinical setting. STATEMENT OF SIGNIFICANCE: We engineered Fibrin Hydrogels (FH) to contain multiple regenerative cues including laminin-1 peptides (L1p) and growth factors (GFs). L1p and GF modified FH were used to induce salivary gland regeneration in a wounded mouse model. Treatment with L1p and GF modified FH promoted salivary epithelial tissue regeneration, vascularization, neurogenesis and healing as compared to L1p-FH or FH alone. Results indicate that L1p and GF modified FH can be used for future therapeutic applications.
Collapse
Affiliation(s)
- Kihoon Nam
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States
| | - Spencer M Dean
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States
| | - Callie T Brown
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States
| | - Randall J Smith
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States
| | - Pedro Lei
- Department of Chemical and Biological Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States; Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States; Center of Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States.
| | - Olga J Baker
- School of Dentistry, The University of Utah, Salt Lake City, UT 84108, United States.
| |
Collapse
|
13
|
3D Bone Biomimetic Scaffolds for Basic and Translational Studies with Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19103150. [PMID: 30322134 PMCID: PMC6213614 DOI: 10.3390/ijms19103150] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are recognized as an attractive tool owing to their self-renewal and differentiation capacity, and their ability to secrete bioactive molecules and to regulate the behavior of neighboring cells within different tissues. Accumulating evidence demonstrates that cells prefer three-dimensional (3D) to 2D culture conditions, at least because the former are closer to their natural environment. Thus, for in vitro studies and in vivo utilization, great effort is being dedicated to the optimization of MSC 3D culture systems in view of achieving the intended performance. This implies understanding cell–biomaterial interactions and manipulating the physicochemical characteristics of biomimetic scaffolds to elicit a specific cell behavior. In the bone field, biomimetic scaffolds can be used as 3D structures, where MSCs can be seeded, expanded, and then implanted in vivo for bone repair or bioactive molecules release. Actually, the union of MSCs and biomaterial has been greatly improving the field of tissue regeneration. Here, we will provide some examples of recent advances in basic as well as translational research about MSC-seeded scaffold systems. Overall, the proliferation of tools for a range of applications witnesses a fruitful collaboration among different branches of the scientific community.
Collapse
|
14
|
Cengiz IF, Oliveira JM, Reis RL. Micro-CT - a digital 3D microstructural voyage into scaffolds: a systematic review of the reported methods and results. Biomater Res 2018; 22:26. [PMID: 30275969 PMCID: PMC6158835 DOI: 10.1186/s40824-018-0136-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Cell behavior is the key to tissue regeneration. Given the fact that most of the cells used in tissue engineering are anchorage-dependent, their behavior including adhesion, growth, migration, matrix synthesis, and differentiation is related to the design of the scaffolds. Thus, characterization of the scaffolds is highly required. Micro-computed tomography (micro-CT) provides a powerful platform to analyze, visualize, and explore any portion of interest in the scaffold in a 3D fashion without cutting or destroying it with the benefit of almost no sample preparation need. MAIN BODY This review highlights the relationship between the scaffold microstructure and cell behavior, and provides the basics of the micro-CT method. In this work, we also analyzed the original papers that were published in 2016 through a systematic search to address the need for specific improvements in the methods section of the papers including the amount of provided information from the obtained results. CONCLUSION Micro-CT offers a unique microstructural analysis of biomaterials, notwithstanding the associated challenges and limitations. Future studies that will include micro-CT characterization of scaffolds should report the important details of the method, and the derived quantitative and qualitative information can be maximized.
Collapse
Affiliation(s)
- Ibrahim Fatih Cengiz
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim Miguel Oliveira
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| |
Collapse
|
15
|
Huang J, Lin C, Fang J, Li X, Wang J, Deng S, Zhang S, Su W, Feng X, Chen B, Cheng D, Shuai X. pH-Sensitive Nanocarrier-Mediated Codelivery of Simvastatin and Noggin siRNA for Synergistic Enhancement of Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2018; 10:28471-28482. [PMID: 30067011 DOI: 10.1021/acsami.8b10521] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The inexpensive hypolipidemic drug simvastatin (SIM), which promotes bone regeneration by enhancing bone morphogenetic protein 2 (BMP-2) expression, has been regarded as an ideal alternative to BMP-2 therapy. However, SIM has low bioavailability and may induce the upregulation of the BMP-2-antagonistic noggin protein, which greatly limits the osteogenic effect. Here, a pH-sensitive copolymer, monomethoxy-poly(ethylene glycol)- b-branched polyethyleneimine- b-poly( N-( N', N'-diisopropylaminoethyl)- co-benzylamino)aspartamide (mPEG-bPEI-PAsp(DIP-BzA)) (PBP), was synthesized and self-assembled into a cationic micelle. SIM and siRNA targeting the noggin gene (N-siRNA) were loaded into the PAsp(DIP-BzA) core and the cationic bPEI interlayer of the micelle via hydrophobic and electrostatic interactions, respectively. The SIM-loaded micelle effectively delivered SIM into preosteoblast MC3T3-E1 cells and rapidly released it inside the acidic lysosome, resulting in the elevated expression of BMP-2. Meanwhile, the codelivered N-siRNA effectively suppressed the expression of noggin. Consequently, SIM and N-siRNA synergistically increased the BMP-2/noggin ratio and resulted in an obviously higher osteogenetic effect than did simvastatin or N-siRNA alone, both in vitro and in vivo.
Collapse
Affiliation(s)
- Jinsheng Huang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
| | - Chaowen Lin
- Department of Orthopaedics and Traumatology, Nanfang Hospital , Southern Medical University , Guangzhou 510515 , China
| | - Jintao Fang
- Department of Microsurgery & Orthopedic Trauma , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou 510080 , China
| | - Xiaoxia Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
| | - Jin Wang
- The Third Affiliated Hospital , Sun Yat-sen University , Guangzhou 510630 , China
| | - Shaohui Deng
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
| | - Sheng Zhang
- Department of Orthopaedics and Traumatology, Nanfang Hospital , Southern Medical University , Guangzhou 510515 , China
| | - Wanhan Su
- Department of Spinal Surgery, Longyan First Hospital , Fujian Medical University , Longyan 364000 , Fujian , China
| | - Xiaoreng Feng
- Department of Orthopaedics and Traumatology, Nanfang Hospital , Southern Medical University , Guangzhou 510515 , China
| | - Bin Chen
- Department of Orthopaedics and Traumatology, Nanfang Hospital , Southern Medical University , Guangzhou 510515 , China
| | - Du Cheng
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
- The Third Affiliated Hospital , Sun Yat-sen University , Guangzhou 510630 , China
| |
Collapse
|
16
|
Chaves Filho GP, de Sousa AFG, Câmara RBG, Rocha HAO, de Medeiros SRB, Moreira SMG. Genotoxicity and osteogenic potential of sulfated polysaccharides from Caulerpa prolifera seaweed. Int J Biol Macromol 2018; 114:565-571. [PMID: 29578018 DOI: 10.1016/j.ijbiomac.2018.03.132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/06/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022]
Abstract
Marine algae are sources of novel bioactive molecules and present a great potential for biotechnological and biomedical applications. Although green algae are the least studied type of seaweed, several of their biological activities have already been described. Here, we investigated the osteogenic potential of Sulfated Polysaccharide (SP)-enriched samples extracted from the green seaweed Caulerpa prolifera on human mesenchymal stem cells isolated from Wharton jelly (hMSC-WJ). In addition, the potential genotoxicity of these SPs was determined by cytokinesis-block micronucleus (CBMN) assay. SP-enriched samples did not show significant cytotoxicity towards hMSCs-WJ at a concentration of up to 10μg/mL, and after 72h of exposure. SP enrichment also significantly increased alkaline phosphatase (ALP) activity, promoting calcium accumulation in the extracellular matrix. Among the SP-enriched samples, the CP0.5 subfraction (at 5μg/mL) presented the most promising results. In this sample, ALP activity was increased approximately by 60%, and calcium accumulation was approximately 6-fold above the negative control, indicating high osteogenic potential. This subfraction also proved to be non-genotoxic, according to the CBMN assay, as it did not induce micronuclei. The results of this study highlight, for the first time, the potential of these SPs for the development of new therapies for bone regeneration.
Collapse
|
17
|
Ndong JDLC, Stephenson Y, Davis ME, García AJ, Goudy S. Controlled JAGGED1 delivery induces human embryonic palate mesenchymal cells to form osteoblasts. J Biomed Mater Res A 2017; 106:552-560. [PMID: 28913955 DOI: 10.1002/jbm.a.36236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/09/2017] [Accepted: 08/25/2017] [Indexed: 12/12/2022]
Abstract
Osteoblast commitment and differentiation are controlled by multiple growth factors including members of the Notch signaling pathway. JAGGED1 is a cell surface ligand of the Notch pathway that is necessary for murine bone formation. The delivery of JAGGED1 to induce bone formation is complicated by its need to be presented in a bound form to allow for proper Notch receptor signaling. In this study, we investigate whether the sustained release of JAGGED1 stimulates human mesenchymal cells to commit to osteoblast cell fate using polyethylene glycol malemeide (PEG-MAL) hydrogel delivery system. Our data demonstrated that PEG-MAL hydrogel constructs are stable in culture for at least three weeks and maintain human mesenchymal cell viability with little cytotoxicity in vitro. JAGGED1 loaded on PEG-MAL hydrogel (JAGGED1-PEG-MAL) showed continuous release from the gel for up to three weeks, with induction of Notch signaling using a CHO cell line with a Notch1 reporter construct, and qPCR gene expression analysis in vitro. Importantly, JAGGED1-PEG-MAL hydrogel induced mesenchymal cells towards osteogenic differentiation based on increased Alkaline phosphatase activity and osteoblast genes expression including RUNX2, ALP, COL1, and BSP. These results thus indicated that JAGGED1 delivery in vitro using PEG-MAL hydrogel induced osteoblast commitment, suggesting that this may be a viable in vivo approach to bone regeneration. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 552-560, 2018.
Collapse
Affiliation(s)
| | | | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, Georgia
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Steven Goudy
- Emory University, Department of Otolaryngology, Atlanta, Georgia.,Emory University, Department of Pediatrics, Atlanta, Georgia
| |
Collapse
|
18
|
Nam K, Maruyama CL, Wang CS, Trump BG, Lei P, Andreadis ST, Baker OJ. Laminin-111-derived peptide conjugated fibrin hydrogel restores salivary gland function. PLoS One 2017; 12:e0187069. [PMID: 29095857 PMCID: PMC5667805 DOI: 10.1371/journal.pone.0187069] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/12/2017] [Indexed: 12/28/2022] Open
Abstract
Hyposalivation reduces the patient quality of life, as saliva is important for maintaining oral health. Current treatments for hyposalivation are limited to medications such as the muscarinic receptor agonists, pilocarpine and cevimeline. However, these therapies only provide temporary relief. Therefore, alternative therapies are essential to restore salivary gland function. An option is to use bioengineered scaffolds to promote functional salivary gland regeneration. Previous studies demonstrated that the laminin-111 protein is critical for intact salivary gland cell cluster formation and organization. However, laminin-111 protein as a whole is not suitable for clinical applications as some protein domains may contribute to unwanted side effects such as degradation, tumorigenesis and immune responses. Conversely, the use of synthetic laminin-111 peptides makes it possible to minimize the immune reactivity or pathogen transfer. In addition, it is relatively simple and inexpensive as compared to animal-derived proteins. Therefore, the goal of this study was to demonstrate whether a 20 day treatment with laminin-111-derived peptide conjugated fibrin hydrogel promotes tissue regeneration in submandibular glands of a wound healing mouse model. In this study, laminin-111-derived peptide conjugated fibrin hydrogel significantly accelerated formation of salivary gland tissue. The regenerated gland tissues displayed not only structural but also functional restoration.
Collapse
Affiliation(s)
- Kihoon Nam
- School of Dentistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Christina L. Maruyama
- School of Dentistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Ching-Shuen Wang
- School of Dentistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Bryan G. Trump
- School of Dentistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
- Center of Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Olga J. Baker
- School of Dentistry, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
19
|
M. Živković J, Đ. Vukelić - Nikolić M, G. Najdanović J, Stojanović S, S. Vitorović J, B. Radenković M, J. Najman S. BON E TISSUE ENGINEER ING BA SED ON BONE MARROW I N BLOOD CLOT LOADED ON MINER AL MATRIX CARRIER: E XPERIMENTAL STUDY IN SUBCUTANEOUS MICE MODEL. ACTA MEDICA MEDIANAE 2017. [DOI: 10.5633/amm.2017.0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
20
|
Noori A, Ashrafi SJ, Vaez-Ghaemi R, Hatamian-Zaremi A, Webster TJ. A review of fibrin and fibrin composites for bone tissue engineering. Int J Nanomedicine 2017; 12:4937-4961. [PMID: 28761338 PMCID: PMC5516781 DOI: 10.2147/ijn.s124671] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Tissue engineering has emerged as a new treatment approach for bone repair and regeneration seeking to address limitations associated with current therapies, such as autologous bone grafting. While many bone tissue engineering approaches have traditionally focused on synthetic materials (such as polymers or hydrogels), there has been a lot of excitement surrounding the use of natural materials due to their biologically inspired properties. Fibrin is a natural scaffold formed following tissue injury that initiates hemostasis and provides the initial matrix useful for cell adhesion, migration, proliferation, and differentiation. Fibrin has captured the interest of bone tissue engineers due to its excellent biocompatibility, controllable biodegradability, and ability to deliver cells and biomolecules. Fibrin is particularly appealing because its precursors, fibrinogen, and thrombin, which can be derived from the patient's own blood, enable the fabrication of completely autologous scaffolds. In this article, we highlight the unique properties of fibrin as a scaffolding material to treat bone defects. Moreover, we emphasize its role in bone tissue engineering nanocomposites where approaches further emulate the natural nanostructured features of bone when using fibrin and other nanomaterials. We also review the preparation methods of fibrin glue and then discuss a wide range of fibrin applications in bone tissue engineering. These include the delivery of cells and/or biomolecules to a defect site, distributing cells, and/or growth factors throughout other pre-formed scaffolds and enhancing the physical as well as biological properties of other biomaterials. Thoughts on the future direction of fibrin research for bone tissue engineering are also presented. In the future, the development of fibrin precursors as recombinant proteins will solve problems associated with using multiple or single-donor fibrin glue, and the combination of nanomaterials that allow for the incorporation of biomolecules with fibrin will significantly improve the efficacy of fibrin for numerous bone tissue engineering applications.
Collapse
Affiliation(s)
- Alireza Noori
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran
| | | | - Roza Vaez-Ghaemi
- Department of Chemical and Biological Engineering, Faculty of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | | | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
21
|
Chen J, Zhang X, Huang C, Cai H, Hu S, Wan Q, Pei X, Wang J. Osteogenic activity and antibacterial effect of porous titanium modified with metal-organic framework films. J Biomed Mater Res A 2016; 105:834-846. [PMID: 27885785 DOI: 10.1002/jbm.a.35960] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/30/2016] [Accepted: 11/04/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Junyu Chen
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- College of Chemistry; Sichuan University; Chengdu, Sichuan 610041 China
| | - Chao Huang
- College of Chemistry; Sichuan University; Chengdu, Sichuan 610041 China
| | - He Cai
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
| | - Shanshan Hu
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
| | - Jian Wang
- State Key Laboratory of Oral Diseases; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
- Department of Prosthodontics; West China Hospital of Stomatology, Sichuan University; Chengdu Sichuan 610041 China
| |
Collapse
|
22
|
Chen J, Zhang X, Cai H, Chen Z, Wang T, Jia L, Wang J, Wan Q, Pei X. Osteogenic activity and antibacterial effect of zinc oxide/carboxylated graphene oxide nanocomposites: Preparation and in vitro evaluation. Colloids Surf B Biointerfaces 2016; 147:397-407. [DOI: 10.1016/j.colsurfb.2016.08.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/11/2016] [Accepted: 08/17/2016] [Indexed: 11/28/2022]
|