1
|
Zhao F, Zhu J, Dong X, Guo X, Lai C, Zhao J, Zong X, Song G, Jin X. The Influence of Extracellular Vesicles Secreted by Dural Cells on Osteoblasts. Mol Biotechnol 2024; 66:3674-3687. [PMID: 38040933 DOI: 10.1007/s12033-023-00974-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 12/03/2023]
Abstract
To explore the influence of extracellular vesicles secreted by dural cells (Dura-EVs) on osteoblasts. Our methodology involves assessing the effects of these EVs at concentrations of 50ug/ml, 100ug/ml, and 200ug/ml on osteoblasts proliferation, differentiation, migration, osteogenesis, and inhibition of apoptosis. We also treated a cranial defect model with injections of these Dura-EVs and monitored the healing rate of cranial defects. Tissue sections were analyzed using Hematoxylin and Eosin (H & E), Masson's trichrome, and immunofluorescence (IF) staining. Our results suggest that Dura-EVs can enhance osteoblasts proliferation, migration, differentiation, and osteogenesis in a dose-dependent manner in vitro. In vivo, Dura-EVs may promote the repair of skull defects. Dura-EVs have an important influence on osteoblasts, our findings shed light on a novel aspect of the dura mater's contribution to cranial osteogenesis.
Collapse
Affiliation(s)
- Fangning Zhao
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Jinglin Zhu
- The Adipose Remodeling Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Xinhang Dong
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Xiaoshuang Guo
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Chenzhi Lai
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Jingyi Zhao
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Xianlei Zong
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Guodong Song
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Xiaolei Jin
- The 4th Craniomaxillofacial Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
2
|
Albougha MS, Sugii H, Adachi O, Mardini B, Soeno S, Hamano S, Hasegawa D, Yoshida S, Itoyama T, Obata J, Maeda H. Exosomes from Human Periodontal Ligament Stem Cells Promote Differentiation of Osteoblast-like Cells and Bone Healing in Rat Calvarial Bone. Biomolecules 2024; 14:1455. [PMID: 39595630 PMCID: PMC11591890 DOI: 10.3390/biom14111455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Deep caries and severe periodontitis cause bone resorption in periodontal tissue, and severe bone resorption leads to tooth loss. Periodontal ligament stem cells (PDLSCs) are important for the healing of defective periodontal tissue. It is increasingly understood that healing of periodontal tissue is mediated through the secretion of trophic factors, particularly exosomes. This study investigated the effects of exosomes from human PDLSCs (HPDLSCs-Exo) on human osteoblast-like cells in vitro and on the healing of rat calvarial bone defects in vivo. HPDLSCs-Exo were isolated and characterized by their particle shape, size (133 ± 6.4 nm), and expression of surface markers (CD9, CD63, and CD81). In vitro results showed that HPDLSCs-Exo promoted the migration, mineralization, and expression of bone-related genes such as alkaline phosphatase (ALP), bone morphogenetic protein 2 (BMP2), osteocalcin (OCN), and osteopontin (OPN) in human osteoblast-like cells. Furthermore, in vivo results showed that more newly formed bone was observed in the HPDLSCs-Exo-treated group than in the non-treated group at the defect sites in rats. These results indicated that HPDLSCs-Exo could promote osteogenesis in vitro and in vivo, and this suggests that HPDLSCs-Exo may be an attractive treatment tool for bone healing in defective periodontal tissue.
Collapse
Affiliation(s)
- Mhd Safwan Albougha
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Hideki Sugii
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Orie Adachi
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Bara Mardini
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Serina Soeno
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Sayuri Hamano
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Daigaku Hasegawa
- Department of Endodontology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shinichiro Yoshida
- Department of Endodontology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomohiro Itoyama
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Junko Obata
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
| | - Hidefumi Maeda
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (M.S.A.)
- Department of Endodontology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
3
|
Yu T, Zhao IS, Pan H, Yang J, Wang H, Deng Y, Zhang Y. Extracellular vesicle-functionalized bioactive scaffolds for bone regeneration. Asian J Pharm Sci 2024; 19:100945. [PMID: 39483718 PMCID: PMC11525715 DOI: 10.1016/j.ajps.2024.100945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 11/03/2024] Open
Abstract
The clinical need for effective bone regeneration in compromised conditions continues to drive demand for innovative solutions. Among emerging strategies, extracellular vesicles (EVs) have shown promise as an acellular approach for bone regeneration. However, their efficacy is hindered by rapid sequestration and clearance when administered via bolus injection. To address this challenge, EV-functionalized scaffolds have recently been proposed as an alternative delivery strategy to enhance EV retention and subsequent healing efficacy. This review aims to consolidate recent advancements in the development of EV-functionalized scaffolds for augmenting bone regeneration. It explores various sources of EVs and different strategies for integrating them into biomaterials. Furthermore, the mechanisms underlying their therapeutic effects in bone regeneration are elucidated. Current limitations in clinical translation and perspectives on the design of more efficient EVs for improved therapeutic efficacy are also presented. Overall, this review can provide inspiration for the development of novel EV-assisted grafts with superior bone regeneration potential.
Collapse
Affiliation(s)
- Taozhao Yu
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
| | - Irene Shuping Zhao
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| | - Hongguang Pan
- Department of Otolaryngology, Shenzhen Children Hospital, Shenzhen 518034, China
| | - Jianhua Yang
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Yongqiang Deng
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen 518055, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
4
|
Wang S, Jia Z, Dai M, Feng X, Tang C, Liu L, Cao L. Advances in natural and synthetic macromolecules with stem cells and extracellular vesicles for orthopedic disease treatment. Int J Biol Macromol 2024; 268:131874. [PMID: 38692547 DOI: 10.1016/j.ijbiomac.2024.131874] [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: 10/15/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Serious orthopedic disorders resulting from myriad diseases and impairments continue to pose a considerable challenge to contemporary clinical care. Owing to its limited regenerative capacity, achieving complete bone tissue regeneration and complete functional restoration has proven challenging with existing treatments. By virtue of cellular regenerative and paracrine pathways, stem cells are extensively utilized in the restoration and regeneration of bone tissue; however, low survival and retention after transplantation severely limit their therapeutic effect. Meanwhile, biomolecule materials provide a delivery platform that improves stem cell survival, increases retention, and enhances therapeutic efficacy. In this review, we present the basic concepts of stem cells and extracellular vesicles from different sources, emphasizing the importance of using appropriate expansion methods and modification strategies. We then review different types of biomolecule materials, focusing on their design strategies. Moreover, we summarize several forms of biomaterial preparation and application strategies as well as current research on biomacromolecule materials loaded with stem cells and extracellular vesicles. Finally, we present the challenges currently impeding their clinical application for the treatment of orthopedic diseases. The article aims to provide researchers with new insights for subsequent investigations.
Collapse
Affiliation(s)
- Supeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China; Ningxia Medical University, Ningxia 750004, China
| | - Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xujun Feng
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China.
| |
Collapse
|
5
|
Yang H, Zhao A, Chen Y, Cheng T, Zhou J, Li Z. Exploring the potential link between MitoEVs and the immune microenvironment of periodontitis based on machine learning and bioinformatics methods. BMC Oral Health 2024; 24:169. [PMID: 38308306 PMCID: PMC10838001 DOI: 10.1186/s12903-024-03912-8] [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: 09/05/2023] [Accepted: 01/18/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Periodontitis is a chronic inflammatory condition triggered by immune system malfunction. Mitochondrial extracellular vesicles (MitoEVs) are a group of highly heterogeneous extracellular vesicles (EVs) enriched in mitochondrial fractions. The objective of this research was to examine the correlation between MitoEVs and the immune microenvironment of periodontitis. METHODS Data from MitoCarta 3.0, GeneCards, and GEO databases were utilized to identify differentially expressed MitoEV-related genes (MERGs) and conduct functional enrichment and pathway analyses. The random forest and LASSO algorithms were employed to identify hub MERGs. Infiltration levels of immune cells in periodontitis and healthy groups were estimated using the CIBERSORT algorithm, and phenotypic subgroups of periodontitis based on hub MERG expression levels were explored using a consensus clustering method. RESULTS A total of 44 differentially expressed MERGs were identified. The random forest and LASSO algorithms identified 9 hub MERGs (BCL2L11, GLDC, CYP24A1, COQ2, MTPAP, NIPSNAP3A, FAM162A, MYO19, and NDUFS1). ROC curve analysis showed that the hub gene and logistic regression model presented excellent diagnostic and discriminating abilities. Immune infiltration and consensus clustering analysis indicated that hub MERGs were highly correlated with various types of immune cells, and there were significant differences in immune cells and hub MERGs among different periodontitis subtypes. CONCLUSION The periodontitis classification model based on MERGs shows excellent performance and can offer novel perspectives into the pathogenesis of periodontitis. The high correlation between MERGs and various immune cells and the significant differences between immune cells and MERGs in different periodontitis subtypes can clarify the regulatory roles of MitoEVs in the immune microenvironment of periodontitis. Future research should focus on elucidating the functional mechanisms of hub MERGs and exploring potential therapeutic interventions based on these findings.
Collapse
Affiliation(s)
- Haoran Yang
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Anna Zhao
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Yuxiang Chen
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Tingting Cheng
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China
| | | | - Ziliang Li
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China.
- Yunnan Provincial Key Laboratory of Stomatology, Kunming, Yunnan, China.
| |
Collapse
|
6
|
Wang W, Xu Z, Liu M, Cai M, Liu X. Prospective applications of extracellular vesicle-based therapies in regenerative medicine: implications for the use of dental stem cell-derived extracellular vesicles. Front Bioeng Biotechnol 2023; 11:1278124. [PMID: 37936823 PMCID: PMC10627172 DOI: 10.3389/fbioe.2023.1278124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
In the 21st century, research on extracellular vesicles (EVs) has made remarkable advancements. Recently, researchers have uncovered the exceptional biological features of EVs, highlighting their prospective use as therapeutic targets, biomarkers, innovative drug delivery systems, and standalone therapeutic agents. Currently, mesenchymal stem cells stand out as the most potent source of EVs for clinical applications in tissue engineering and regenerative medicine. Owing to their accessibility and capability of undergoing numerous differentiation inductions, dental stem cell-derived EVs (DSC-EVs) offer distinct advantages in the field of tissue regeneration. Nonetheless, it is essential to note that unmodified EVs are currently unsuitable for use in the majority of clinical therapeutic scenarios. Considering the high feasibility of engineering EVs, it is imperative to modify these EVs to facilitate the swift translation of theoretical knowledge into clinical practice. The review succinctly presents the known biotherapeutic effects of odontogenic EVs and the underlying mechanisms. Subsequently, the current state of functional cargo loading for engineered EVs is critically discussed. For enhancing EV targeting and in vivo circulation time, the review highlights cutting-edge engineering solutions that may help overcome key obstacles in the clinical application of EV therapeutics. By presenting innovative concepts and strategies, this review aims to pave the way for the adaptation of DSC-EVs in regenerative medicine within clinical settings.
Collapse
Affiliation(s)
- Wenhao Wang
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zinan Xu
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Minyi Liu
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Research Platform for Interdiscipline, Jinan University, Guangzhou, China
| | - Mingxiang Cai
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiangning Liu
- School of Stomatology, Jinan University, Guangzhou, China
- Center of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Research Platform for Interdiscipline, Jinan University, Guangzhou, China
| |
Collapse
|
7
|
Wei X, Liu S, Cao Y, Wang Z, Chen S. Polymers in Engineering Extracellular Vesicle Mimetics: Current Status and Prospective. Pharmaceutics 2023; 15:pharmaceutics15051496. [PMID: 37242738 DOI: 10.3390/pharmaceutics15051496] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
The maintenance of a high delivery efficiency by traditional nanomedicines during cancer treatment is a challenging task. As a natural mediator for short-distance intercellular communication, extracellular vesicles (EVs) have garnered significant attention owing to their low immunogenicity and high targeting ability. They can load a variety of major drugs, thus offering immense potential. In order to overcome the limitations of EVs and establish them as an ideal drug delivery system, polymer-engineered extracellular vesicle mimics (EVMs) have been developed and applied in cancer therapy. In this review, we discuss the current status of polymer-based extracellular vesicle mimics in drug delivery, and analyze their structural and functional properties based on the design of an ideal drug carrier. We anticipate that this review will facilitate a deeper understanding of the extracellular vesicular mimetic drug delivery system, and stimulate the progress and advancement of this field.
Collapse
Affiliation(s)
- Xinyue Wei
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Sihang Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Advanced Optical Communication Systems and Networks, Key Laboratory for Thin Film and Microfabrication of the Ministry of Education, UM-SJTU Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yifeng Cao
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- Department of Electronic Chemicals, Institute of Zhejiang University-Quzhou, Quzhou 324000, China
| | - Zhen Wang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- Zhejiang Sundoc Pharmaceutical Science and Tech Co., Ltd., Hangzhou 310051, China
| | - Shengfu Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
8
|
Huang X, Li Y, Liao H, Luo X, Zhao Y, Huang Y, Zhou Z, Xiang Q. Research Advances on Stem Cell-Derived Extracellular Vesicles Promoting the Reconstruction of Alveolar Bone through RANKL/RANK/OPG Pathway. J Funct Biomater 2023; 14:jfb14040193. [PMID: 37103283 PMCID: PMC10145790 DOI: 10.3390/jfb14040193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Periodontal bone tissue defects and bone shortages are the most familiar and troublesome clinical problems in the oral cavity. Stem cell-derived extracellular vesicles (SC-EVs) have biological properties similar to their sources, and they could be a promising acellular therapy to assist with periodontal osteogenesis. In the course of alveolar bone remodeling, the RANKL/RANK/OPG signaling pathway is an important pathway involved in bone metabolism. This article summarizes the experimental studies of SC-EVs applied for the therapy of periodontal osteogenesis recently and explores the role of the RANKL/RANK/OPG pathway in their mechanism of action. Their unique patterns will open a new field of vision for people, and they will help to advance a possible future clinical treatment.
Collapse
Affiliation(s)
- Xia Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
- Department of Orthodontics, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Yuxiao Li
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Hui Liao
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Xin Luo
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Yueping Zhao
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Zhiying Zhou
- School of Stomatology, Jinan University, Guangzhou 510632, China
- Department of Orthodontics, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
9
|
Song X, Xu L, Zhang W. Biomimetic synthesis and optimization of extracellular vesicles for bone regeneration. J Control Release 2023; 355:18-41. [PMID: 36706840 DOI: 10.1016/j.jconrel.2023.01.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/29/2023]
Abstract
Critical-size bone defect repair is in high demand but is difficult to treat. Modern therapies, such as autograft and cell-based treatments, face limitations, including potential immunological rejection and tumorigenesis. Therefore, extracellular vesicle (EV)-based strategies have been proposed as a novel approach for tissue regeneration owing to EVs' complex composition of lipids, proteins, and nucleic acids, as well as their low immunogenicity and congenital cell-targeting features. Despite these remarkable features of EVs, biomimetic synthesis and optimization of natural EVs can lead to enhanced bioactivity, increased cellular uptake, and specific cell targeting, aiming to achieve optimal therapeutic efficacy. To maximize their function, these nanoparticles can be integrated into bone graft biomaterials for superior bone regeneration. Herein, we summarize the role of naturally occurring EVs from distinct cell types in bone regeneration, the current strategies for optimizing biomimetic synthetic EVs in bone regeneration, and discuss the recent advances in applying bone graft biomaterials for the delivery of EVs to bone defect repair. We focused on distinct strategies for optimizing EVs with different functions and the most recent research on achieving time-controlled release of nanoparticles from EV-loaded biomaterials. Furthermore, we thoroughly discuss several current challenges and proposed solutions, aiming to provide insight into current progress, inspiration for future development directions, and incentives for clinical application in this field.
Collapse
Affiliation(s)
- Xinyu Song
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Ling Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| |
Collapse
|
10
|
Xu J, Lin Y, Tian M, Li X, Yin Y, Li Q, Li Z, Zhou J, Jiang X, Li Y, Chen S. Periodontal Ligament Stem Cell-Derived Extracellular Vesicles Enhance Tension-Induced Osteogenesis. ACS Biomater Sci Eng 2023; 9:388-398. [PMID: 36538768 DOI: 10.1021/acsbiomaterials.2c00717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tension-induced osteogenesis has great significance in maintaining bone homeostasis and ensuring the efficiency and stability of orthodontic treatment. Recently, extracellular vesicles (EVs) have shown great potential in regulating bone remodeling. Here, we aimed to explore the effects of periodontal ligament stem cell (PDLSC)-derived EVs on tension-induced osteogenesis and the potential mechanism. PDLSC-derived EVs were extracted by ultracentrifugation. In vitro, PDLSC-derived EVs of 10 μg/mL significantly improved the proliferation of MC3T3-E1 cells and enhanced the osteogenic differentiation of osteoblasts under a tensile strain of 2000 uε. Next, a mouse model of orthodontic tooth movement (OTM) was established and treated with subperiosteal injection of PDLSC-derived EVs (1 mg/kg) on the tension side. The results showed that treatment with PDLSC-derived EVs effectively enhanced OTM and promoted osteogenesis on the tension side, including increasing trabecular bone parameters and promoting the expression of osteogenic-related biomarkers (OCN and OPN). More interestingly, we identified several mechano-sensitive miRNAs enriched in PDLSC-derived EVs by high-throughput miRNA sequencing. Bioinformatics analysis indicated that they were related to various osteogenesis-related signaling pathways. Therefore, PDLSC-derived EVs could improve the efficiency of OTM by enhancing tension-induced osteogenesis of osteoblasts. Our study may provide potential evidence for the promoting effects of PDLSC-derived EVs on osteogenesis and offer new insights into the development of treatment strategies for enhancing osteogenesis in orthodontic treatment and other metabolic bone diseases.
Collapse
Affiliation(s)
- Jingchen Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Yao Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Mi Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Yuanyuan Yin
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Qiming Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Ziyu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Jialiang Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Xiaoge Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Yulin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, Ren Min S Rd, Chengdu, Sichuan 610041, China
| |
Collapse
|
11
|
Cai R, Wang L, Zhang W, Liu B, Wu Y, Pang J, Ma C. The role of extracellular vesicles in periodontitis: pathogenesis, diagnosis, and therapy. Front Immunol 2023; 14:1151322. [PMID: 37114060 PMCID: PMC10126335 DOI: 10.3389/fimmu.2023.1151322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Periodontitis is a prevalent disease and one of the leading causes of tooth loss. Biofilms are initiating factor of periodontitis, which can destroy periodontal tissue by producing virulence factors. The overactivated host immune response is the primary cause of periodontitis. The clinical examination of periodontal tissues and the patient's medical history are the mainstays of periodontitis diagnosis. However, there is a lack of molecular biomarkers that can be used to identify and predict periodontitis activity precisely. Non-surgical and surgical treatments are currently available for periodontitis, although both have drawbacks. In clinical practice, achieving the ideal therapeutic effect remains a challenge. Studies have revealed that bacteria produce extracellular vesicles (EVs) to export virulence proteins to host cells. Meanwhile, periodontal tissue cells and immune cells produce EVs that have pro- or anti-inflammatory effects. Accordingly, EVs play a critical role in the pathogenesis of periodontitis. Recent studies have also presented that the content and composition of EVs in saliva and gingival crevicular fluid (GCF) can serve as possible periodontitis diagnostic indicators. In addition, studies have indicated that stem cell EVs may encourage periodontal regeneration. In this article, we mainly review the role of EVs in the pathogenesis of periodontitis and discuss their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Rong Cai
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Zhang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Bing Liu
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianliang Pang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| | - Chufan Ma
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| |
Collapse
|
12
|
Yang P, Shi F, Zhang Y. Baricitinib alleviates lipopolysaccharide‑induced human periodontal ligament stem cell injury and promotes osteogenic differentiation by inhibiting JAK/STAT signaling. Exp Ther Med 2022; 25:74. [PMID: 36684656 PMCID: PMC9842944 DOI: 10.3892/etm.2022.11773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Periodontitis is the chronic inflammation of the periodontal tissue. The present study aimed to investigate the role of baricitinib, a Janus kinase (JAK)1/2 inhibitor, in periodontitis by using a lipopolysaccharide (LPS)-induced human periodontal ligament stem cell (PDLSC) model. The viability of PDLSCs stimulated by LPS was assessed in the presence of baricitinib by Cell Counting Kit-8 assay. The induction of oxidative stress was evaluated by detecting the intracellular reactive oxygen species (ROS) levels, superoxide dismutase (SOD) activity and glutathione (GSH) content. ELISA and reverse transcription-quantitative PCR were used to determine the levels of inflammatory factors TNF-α, IL-1β and IL-6. Alkaline phosphatase (ALP) activity and alizarin red staining were used to assess the osteogenic differentiation of PDLSCs. The expression levels of osteogenic differentiation- and JAK/signal transducer and activator of transcription (STAT) signaling-associated proteins were estimated with western blotting. RO8191, an agonist of the JAK/STAT pathway, was used to treat PDLSCs to investigate the regulatory mechanism of baricitinib. The results indicated that baricitinib elevated the LPS-induced decrease in cell viability. LPS-triggered oxidative stress and inflammation were inhibited by baricitinib, as demonstrated by the decreased levels of ROS, TNF-α, IL-1β, IL-6 and increased levels of SOD and GSH. In addition, baricitinib caused a marked elevation in ALP activity and mineralization ability of PDLSCs, as determined by the upregulated osteocalcin and Runt-related transcription factor 2 expression. Moreover, the expression levels of phosphorylated (p)-JAK1, p-JAK2 and p-STAT3 were downregulated by baricitinib in a dose-dependent manner. Furthermore, addition of RO8191 restored the effect of baricitinib on the induction of oxidative stress, inflammation and osteogenic differentiation of PDLSCs exposed to LPS. Collectively, these findings suggested that baricitinib alleviated oxidative stress and inflammation and promoted osteogenic differentiation of LPS-induced PDLSCs by inhibiting JAK/STAT signaling.
Collapse
Affiliation(s)
- Ping Yang
- Department of Stomatology, AnTing Campus, The Third Affiliated Hospital of Naval Military Medical University, Shanghai 200438, P.R. China
| | - Fenghua Shi
- Department of Radiotherapy, AnTing Campus, The Third Affiliated Hospital of Naval Military Medical University, Shanghai 201805, P.R. China
| | - Yanli Zhang
- Outpatient Department, ChangHai Road Campus, The Third Affiliated Hospital of Naval Military Medical University, Shanghai 200438, P.R. China,Correspondence to: Dr Yanli Zhang, Outpatient Department, ChangHai Road Campus, The Third Affiliated Hospital of Naval Military Medical University, 225 Changhai Road, Yangpu, Shanghai 200438, P.R. China
| |
Collapse
|
13
|
Liu C, Li Y, Han G. Advances of Mesenchymal Stem Cells Released Extracellular Vesicles in Periodontal Bone Remodeling. DNA Cell Biol 2022; 41:935-950. [PMID: 36315196 DOI: 10.1089/dna.2022.0359] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles that include exosomes, microvesicles, and apoptotic bodies; they interact with target cell surface receptors and transport contents, including mRNA, proteins, and enzymes into the cytoplasm of target cells to function. The biological fingerprints of EVs practically mirror those of the parental cells they originated from. In the bone remodeling microenvironment, EVs could act on osteoblasts to regulate the bone formation, promote osteoclast differentiation, and regulate bone resorption. Therefore, there have been many attempts wherein EVs were used to achieve targeted therapy in bone-related diseases. Periodontitis, a common bacterial infectious disease, could cause severe alveolar bone resorption, resulting in tooth loss, whereas research on periodontal bone regeneration is also an urgent question. Therefore, EVs-related studies are important for periodontal bone remodeling. In this review, we summarize the current knowledge of mesenchymal stem cell-EVs involved in periodontal bone remodeling and explore the functional gene expression through a comparative analysis of transcriptomic content.
Collapse
Affiliation(s)
- Chaoran Liu
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Yanan Li
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Guanghong Han
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
14
|
A Narrative Review on the Effectiveness of Bone Regeneration Procedures with OsteoBiol® Collagenated Porcine Grafts: The Translational Research Experience over 20 Years. J Funct Biomater 2022; 13:jfb13030121. [PMID: 35997459 PMCID: PMC9397035 DOI: 10.3390/jfb13030121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Over the years, several bone regeneration procedures have been proposed using natural (autografts, allografts, and xenografts) and synthetic (i.e., metals, ceramics, and polymers) bone grafts. In particular, numerous in vitro and human and animal in vivo studies have been focused on the discovery of innovative and suitable biomaterials for oral and maxillofacial applications in the treatment of severely atrophied jaws. On this basis, the main objective of the present narrative review was to investigate the efficacy of innovative collagenated porcine bone grafts (OsteoBiol®, Tecnoss®, Giaveno, Italy), designed to be as similar as possible to the autologous bone, in several bone regeneration procedures. The scientific publications were screened by means of electronic databases, such as PubMed, Scopus, and Embase, finally selecting only papers that dealt with bone substitutes and scaffolds for bone and soft tissue regeneration. A total of 201 papers have been detected, including in vitro, in vivo, and clinical studies. The effectiveness of over 20 years of translational research demonstrated that these specific porcine bone substitutes are safe and able to improve the biological response and the predictability of the regenerative protocols for the treatment of alveolar and maxillofacial defects.
Collapse
|
15
|
Chouaib B, Cuisinier F, Collart-Dutilleul PY. Dental stem cell-conditioned medium for tissue regeneration: Optimization of production and storage. World J Stem Cells 2022; 14:287-302. [PMID: 35662860 PMCID: PMC9136565 DOI: 10.4252/wjsc.v14.i4.287] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/19/2021] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) effects on tissue regeneration are mainly mediated by their secreted substances (secretome), inducing their paracrine activity. This Conditioned medium (CM), including soluble factors (proteins, nucleic acids, lipids) and extracellular vesicles is emerging as a potential alternative to cell therapy. However, the manufacturing of CM suffers from variable procedures and protocols leading to varying results between studies. Besides, there is no well-defined optimized procedure targeting specific applications in regenerative medicine.
AIM To focus on conditioned medium produced from dental MSC (DMSC-CM), we reviewed the current parameters and manufacturing protocols, in order to propose a standardization and optimization of these manufacturing procedures.
METHODS We have selected all publications investigating the effects of dental MSC secretome in in vitro and in vivo models of tissue regeneration, in accordance with the PRISMA guidelines.
RESULTS A total of 351 results were identified. And based on the inclusion criteria described above, 118 unique articles were included in the systematic review. DMSC-CM production was considered at three stages: before CM recovery (cell sources for CM), during CM production (culture conditions) and after production (CM treatment).
CONCLUSION No clear consensus could be recovered as evidence-based methods, but we were able to describe the most commonly used protocols: donors under 30 years of age, dental pulp stem cells and exfoliated deciduous tooth stem cells with cell passage between 1 and 5, at a confluence of 70% to 80%. CM were often collected during 48 h, and stored at -80 °C. It is important to point out that the preconditioning environment had a significant impact on DMSC-CM content and efficiency.
Collapse
Affiliation(s)
- Batoul Chouaib
- Laboratory Bioengineering and Nanosciences UR_UM104, University of Montpellier, Montpellier 34000, France
| | - Frédéric Cuisinier
- Laboratory Bioengineering and Nanosciences UR_UM104, University of Montpellier, Montpellier 34000, France
| | | |
Collapse
|
16
|
Solakoglu Ö, Steinbach B, Götz W, Heydecke G, Schwarzenbach H. Characterization of circulating molecules and activities in plasma of patients after allogeneic and autologous intraoral bone grafting procedures: a prospective randomized controlled clinical trial in humans. BMC Oral Health 2022; 22:24. [PMID: 35094679 PMCID: PMC8802434 DOI: 10.1186/s12903-021-02036-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background The objective was to assess whether intraoral bone augmentation procedures have an impact on the patient’s plasma levels of circulating nucleic acids, exosomes, miRNA levels and caspase activities. The null hypothesis was tested, that no significant differences between the two groups will be found. Methods In this prospective randomized controlled clinical trial 35 systemically healthy non-smoking participants were randomly allocated using sealed envelopes by a blinded clinician not involved in the clinical setting. Plasma samples were collected preoperatively and 3 times postoperatively (immediately, 5 weeks and 4 months postoperatively). The test group consisted of twenty-five patients who received allogeneic bone grafting material and the control group of ten patients who received autologous bone grafts. Levels of cell-free DNA (cfDNA) and microRNAs (miR-21, miR-27a, miR-218) were quantified by real-time PCR, caspase activities and exosome concentrations were determined by ELISA. Results Statistical evaluation reveled a significantly higher exosome level before surgery (p = 0.013) and the first postsurgical sample (p = 0.017) in the control group compared to the test group. The levels of miR-27a and miR-218 significantly differed between the plasma samples before surgery and after surgery in both groups. The levels of miR-21 only significantly differed between the pre- and postsurgical plasma samples in the test group, but not in the control group. All patients completed the study, no adverse events were recorded. Conclusions Our data show the diagnostic potential of the plasma levels of miR-27a, miR-218 and miR-21 in detecting changes in bone metabolism after alveolar bone augmentation. Our very promising results indicate that there might be a high diagnostic potential in evaluating the plasma levels of the before mentioned miRNAs in order to detect bone resorption activities before they become clinically relevant. Trial registration Ethical commission of the Ärztekammer Hamburg, Germany (PV5211) on 11/03/2016 as well as by the German Registry of Clinical Studies (DRKS 00,013,010) on 30/07/2018 (http://apps.who.int/trialsearch/). Supplementary Information The online version contains supplementary material available at 10.1186/s12903-021-02036-7.
Collapse
|
17
|
Petrillo S, Genova T, Chinigò G, Roato I, Scarpellino G, Kopecka J, Altruda F, Tolosano E, Riganti C, Mussano F, Munaron L. Endothelial Cells Promote Osteogenesis by Establishing a Functional and Metabolic Coupling With Human Mesenchymal Stem Cells. Front Physiol 2022; 12:813547. [PMID: 35087424 PMCID: PMC8787057 DOI: 10.3389/fphys.2021.813547] [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: 11/11/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Bone formation involves a complex crosstalk between endothelial cells (EC) and osteodifferentiating stem cells. This functional interplay is greatly mediated by the paracrine and autocrine action of soluble factors released at the vasculature-bone interface. This study elucidates the molecular and functional responses triggered by this intimate interaction. In this study, we showed that human dermal microvascular endothelial cells (HMEC) induced the expression of pro-angiogenic factors in stem cells from human exfoliated deciduous teeth (SHED) and sustain their osteo-differentiation at the same time. In contrast, osteodifferentiating SHED increased EC recruitment and promoted the formation of complex vascular networks. Moreover, HMEC enhanced anaerobic glycolysis in proliferating SHED without compromising their ability to undergo the oxidative metabolic shift required for adequate osteo-differentiation. Taken together, these findings provide novel insights into the molecular mechanism underlying the synergistic cooperation between EC and stem cells during bone tissue renewal.
Collapse
Affiliation(s)
- Sara Petrillo
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Ilaria Roato
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Giorgia Scarpellino
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Turin, Turin, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Chiara Riganti
- Department of Oncology, University of Turin, Turin, Italy
| | - Federico Mussano
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| |
Collapse
|
18
|
A Narrative Review of Cell-Based Approaches for Cranial Bone Regeneration. Pharmaceutics 2022; 14:pharmaceutics14010132. [PMID: 35057028 PMCID: PMC8781797 DOI: 10.3390/pharmaceutics14010132] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 01/08/2023] Open
Abstract
Current cranial repair techniques combine the use of autologous bone grafts and biomaterials. In addition to their association with harvesting morbidity, autografts are often limited by insufficient quantity of bone stock. Biomaterials lead to better outcomes, but their effectiveness is often compromised by the unpredictable lack of integration and structural failure. Bone tissue engineering offers the promising alternative of generating constructs composed of instructive biomaterials including cells or cell-secreted products, which could enhance the outcome of reconstructive treatments. This review focuses on cell-based approaches with potential to regenerate calvarial bone defects, including human studies and preclinical research. Further, we discuss strategies to deliver extracellular matrix, conditioned media and extracellular vesicles derived from cell cultures. Recent advances in 3D printing and bioprinting techniques that appear to be promising for cranial reconstruction are also discussed. Finally, we review cell-based gene therapy approaches, covering both unregulated and regulated gene switches that can create spatiotemporal patterns of transgenic therapeutic molecules. In summary, this review provides an overview of the current developments in cell-based strategies with potential to enhance the surgical armamentarium for regenerating cranial vault defects.
Collapse
|
19
|
Dental stem cell-derived extracellular vesicles as promising therapeutic agents in the treatment of diseases. Int J Oral Sci 2022; 14:2. [PMID: 34980877 PMCID: PMC8724288 DOI: 10.1038/s41368-021-00152-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023] Open
Abstract
Dental stem cells (DSCs), an important source of mesenchymal stem cells (MSCs), can be easily obtained by minimally invasive procedures and have been used for the treatment of various diseases. Classic paradigm attributed the mechanism of their therapeutic action to direct cell differentiation after targeted migration, while contemporary insights into indirect paracrine effect opened new avenues for the mystery of their actual low engraftment and differentiation ability in vivo. As critical paracrine effectors, DSC-derived extracellular vesicles (DSC-EVs) are being increasingly linked to the positive effects of DSCs by an evolving body of in vivo studies. Carrying bioactive contents and presenting therapeutic potential in certain diseases, DSC-EVs have been introduced as promising treatments. Here, we systematically review the latest in vivo evidence that supports the therapeutic effects of DSC-EVs with mechanistic studies. In addition, current challenges and future directions for the clinical translation of DSC-EVs are also highlighted to call for more attentions to the (I) distinguishing features of DSC-EVs compared with other types of MSC-EVs, (II) heterogeneity among different subtypes of DSC-derived EVs, (III) action modes of DSC-EVs, (IV) standardization for eligible DSC-EVs and (V) safety guarantee for the clinical application of DSC-EVs. The present review would provide valuable insights into the emerging opportunities of DSC-EVs in future clinical applications.
Collapse
|
20
|
Kouhestani F, Aghandeh P, Isamorad F, Akbari S, Tanbakuchi B, Motamedian S. Efficacy of Application of Periodontal Ligament Stem Cells in Bone Regeneration: A Systematic Review of Animal Studies. DENTAL HYPOTHESES 2022. [DOI: 10.4103/denthyp.denthyp_136_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Sun H, Cao X, Gong A, Huang Y, Xu Y, Zhang J, Sun J, Lv B, Li Z, Guan S, Lu L, Yin G. Extracellular vesicles derived from astrocytes facilitated neurite elongation by activating the Hippo pathway. Exp Cell Res 2021; 411:112937. [PMID: 34863709 DOI: 10.1016/j.yexcr.2021.112937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023]
Abstract
Spinal cord injury (SCI) often causes severe neurological dysfunction, and facilitating neurite elongation is particularly important in its treatment. Astrocytes (AS) play an important role in the central nervous system (CNS), and their high plasticity and versatility provide a feasible entry point for relevant research. Our purpose was to explore whether extracellular vesicles (EVs) from astrocytes (AS-EVs) and lipopolysaccharide (LPS)-preactivated astrocytes (LPAS-EVs) facilitate neurite elongation, to explore the underlying mechanism, and to verify whether these EVs promote locomotor recovery in rats. We used LPS to preactivate astrocytes and cocultured them with PC12 cells to observe neurite changes, then extracted and identified AS-EVs and LPAS-EVs and the role and mechanism of these EVs in facilitating neurite elongation was examined in vivo and vitro. We demonstrated that AS-EVs and LPAS-EVs facilitated the elongation of neurites and the recovery of rats with SCI. LPAS-EVs had a stronger effect than AS-EVs, by activating the Hippo pathway, promoting monopole spindle binding protein 1 (MOB1) expression, and reducing Yes-associated protein (YAP) levels. The data also suggest a feedback regulation between MOB1 and p-YAP/YAP. In sum, AS-EVs and LPAS-EVs can play an active role in facilitating neurite elongation by activating the Hippo pathway. These findings provide a new strategy for treating SCI and other CNS-related injuries.
Collapse
Affiliation(s)
- Haitao Sun
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xingbing Cao
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aihua Gong
- Medical College, Jiangsu University, Zhenjiang City, Jiangsu, China
| | - Yonghui Huang
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yi Xu
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jinglong Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jifu Sun
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bin Lv
- Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhen Li
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shihao Guan
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ling Lu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Guoyong Yin
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
22
|
Chabria Y, Duffy GP, Lowery AJ, Dwyer RM. Hydrogels: 3D Drug Delivery Systems for Nanoparticles and Extracellular Vesicles. Biomedicines 2021; 9:1694. [PMID: 34829923 PMCID: PMC8615452 DOI: 10.3390/biomedicines9111694] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 12/16/2022] Open
Abstract
Synthetic and naturally occurring nano-sized particles present versatile vehicles for the delivery of therapy in a range of clinical settings. Their small size and modifiable physicochemical properties support refinement of targeting capabilities, immune response, and therapeutic cargo, but rapid clearance from the body and limited efficacy remain a major challenge. This highlights the need for a local sustained delivery system for nanoparticles (NPs) and extracellular vesicles (EVs) at the target site that will ensure prolonged exposure, maximum efficacy and dose, and minimal toxicity. Biocompatible hydrogels loaded with therapeutic NPs/EVs hold immense promise as cell-free sustained and targeted delivery systems in a range of disease settings. These bioscaffolds ensure retention of the nano-sized particles at the target site and can also act as controlled release systems for therapeutics over a prolonged period of time. The encapsulation of stimuli sensitive components into hydrogels supports the release of the content on-demand. In this review, we highlight the prospect of the sustained and prolonged delivery of these nano-sized therapeutic entities from hydrogels for broad applications spanning tissue regeneration and cancer treatment. Further understanding of the parameters controlling the release rate of these particles and efficient transfer of cargo to target cells will be fundamental to success.
Collapse
Affiliation(s)
- Yashna Chabria
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland Galway, H91 V4AY Galway, Ireland; (Y.C.); (A.J.L.)
- CÚRAM, The SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY Galway, Ireland;
| | - Garry P. Duffy
- CÚRAM, The SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY Galway, Ireland;
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Aoife J Lowery
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland Galway, H91 V4AY Galway, Ireland; (Y.C.); (A.J.L.)
| | - Róisín M. Dwyer
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland Galway, H91 V4AY Galway, Ireland; (Y.C.); (A.J.L.)
- CÚRAM, The SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY Galway, Ireland;
| |
Collapse
|
23
|
Bar JK, Lis-Nawara A, Grelewski PG. Dental Pulp Stem Cell-Derived Secretome and Its Regenerative Potential. Int J Mol Sci 2021; 22:ijms222112018. [PMID: 34769446 PMCID: PMC8584775 DOI: 10.3390/ijms222112018] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
The therapeutic potential of the dental pulp stem (DSC) cell-derived secretome, consisting of various biomolecules, is undergoing intense research. Despite promising in vitro and in vivo studies, most DSC secretome-based therapies have not been implemented in human medicine because the paracrine effect of the bioactive factors secreted by human dental pulp stem cells (hDPSCs) and human exfoliated deciduous teeth (SHEDs) is not completely understood. In this review, we outline the current data on the hDPSC- and SHED-derived secretome as a potential candidate in the regeneration of bone, cartilage, and nerve tissue. Published reports demonstrate that the dental MSC-derived secretome/conditional medium may be effective in treating neurodegenerative diseases, neural injuries, cartilage defects, and repairing bone by regulating neuroprotective, anti-inflammatory, antiapoptotic, and angiogenic processes through secretome paracrine mechanisms. Dental MSC-secretomes, similarly to the bone marrow MSC-secretome activate molecular and cellular mechanisms, which determine the effectiveness of cell-free therapy. Many reports emphasize that dental MSC-derived secretomes have potential application in tissue-regenerating therapy due to their multidirectional paracrine effect observed in the therapy of many different injured tissues.
Collapse
|
24
|
Comparison of Surface Functionalization of PLGA Composite to Immobilize Extracellular Vesicles. Polymers (Basel) 2021; 13:polym13213643. [PMID: 34771200 PMCID: PMC8587822 DOI: 10.3390/polym13213643] [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: 09/29/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelialization by materials provides a promising approach for the rapid re-endothelialization of a cardiovascular implantation. Although previous studies have focused on improving endothelialization through the immobilization of bioactive molecules onto the surface of biodegradable implants, comparative studies of effective surface modification have not yet been reported. Here, we conducted a comparative study on the surface modification of poly(lactide-co-glycolide) (PLGA)-based composites to graft mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) using three different materials, fibronectin (FN), polyethylenimine (PEI), and polydopamine (PDA), which have different bond strengths of ligand–receptor interaction, ionic bond, and covalent bond, respectively. Further in vitro analysis exhibited that MSC-EVs released from all modified films sustainably, but the MSC-EVs grafted onto the surface coated with PEI are more effective than other groups in increasing angiogenesis and reducing the inflammatory responses in endothelial cells. Therefore, the overall results demonstrated that PEI is a desirable coating reagent for the immobilization of MSC-EVs on the surface of biodegradable implants.
Collapse
|
25
|
Transcriptomic analysis revealed increased expression of genes involved in keratinization in the tears of COVID-19 patients. Sci Rep 2021; 11:19817. [PMID: 34615949 PMCID: PMC8494911 DOI: 10.1038/s41598-021-99344-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 09/23/2021] [Indexed: 01/08/2023] Open
Abstract
Recent studies have focused their attention on conjunctivitis as one of the symptoms of coronavirus disease 2019 (COVID-19). Therefore, tear samples were taken from COVID-19 patients and the presence of SARS-CoV-2 was evidenced using Real Time reverse transcription polymerase chain reaction. The main aim of this study was to analyze mRNA expression in the tears of patients with COVID-19 compared with healthy subjects using Next Generation Sequencing (NGS). The functional evaluation of the transcriptome highlighted 25 genes that differ statistically between healthy individuals and patients affected by COVID-19. In particular, the NGS analysis identified the presence of several genes involved in B cell signaling and keratinization. In particular, the genes involved in B cell signaling were downregulated in the tears of COVID-19 patients, while those involved in keratinization were upregulated. The results indicated that SARS-CoV-2 may induce a process of ocular keratinization and a defective B cell response.
Collapse
|
26
|
The relationship between transforming growth factor β superfamily members (GDF11 and BMP4) and lumbar spine bone mineral density in postmenopausal Chinese women. Arch Gynecol Obstet 2021; 305:737-747. [PMID: 34417839 DOI: 10.1007/s00404-021-06183-8] [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: 05/27/2020] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE The relationship between transforming growth factor β superfamily members (GDF11 and BMP4) and bone metabolism remains controversial. The aim of this study was to investigate the association between serum GDF11 and BMP4 levels and lumbar spine bone mineral density (LBMD) in a cohort of postmenopausal Chinese women. METHODS This was a non-prospective cross-sectional study of 350 postmenopausal women with a mean age of 63.13 ± 8.66 years who came from Shenyang, China. LBMD was measured using dual-energy X-ray absorptiometry. Serum GDF11 and BMP4 concentrations were detected using a sandwich enzyme immunoassay kit. Pearson's correlation analysis and regression analyses were carried out to investigate the relationships between LBMD and serum GDF11 and BMP4 levels. RESULTS A linear association between LBMD and serum LgGDF11 concentration was observed after adjusting for numerous confounders (P = 0.018). In addition, the osteoporosis (OP) was inversely related to LgGDF11 and the odds ratios for postmenopausal women with lumbar OP in LgGDF11 quartile group 2, group 3, and group 4 were 0.46 (95% CI 0.23-0.90, P < 0.05), 0.41 (95% CI 0.20-0.84, P < 0.05), and 0.30 (95% CI 0.14-0.63, P < 0.01), respectively (P = 0.001 for the trend), when compared to the highest quartile of LgGDF11 after adjustments for many confounding variables in this study. CONCLUSIONS This study showed that serum GDF11 levels were linearly related to LBMD, and it was also revealed that serum GDF11 levels were significantly associated with lumbar OP in postmenopausal women. However, serum BMP4 levels were not associated with LBMD and lumbar OP.
Collapse
|
27
|
Nagelkerke A, Ojansivu M, van der Koog L, Whittaker TE, Cunnane EM, Silva AM, Dekker N, Stevens MM. Extracellular vesicles for tissue repair and regeneration: Evidence, challenges and opportunities. Adv Drug Deliv Rev 2021; 175:113775. [PMID: 33872693 DOI: 10.1016/j.addr.2021.04.013] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are biological nanoparticles naturally secreted by cells, acting as delivery vehicles for molecular messages. During the last decade, EVs have been assigned multiple functions that have established their potential as therapeutic mediators for a variety of diseases and conditions. In this review paper, we report on the potential of EVs in tissue repair and regeneration. The regenerative properties that have been associated with EVs are explored, detailing the molecular cargo they carry that is capable of mediating such effects, the signaling cascades triggered in target cells and the functional outcome achieved. EV interactions and biodistribution in vivo that influence their regenerative effects are also described, particularly upon administration in combination with biomaterials. Finally, we review the progress that has been made for the successful implementation of EV regenerative therapies in a clinical setting.
Collapse
Affiliation(s)
- Anika Nagelkerke
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, P.O. Box 196, XB20, 9700 AD Groningen, the Netherlands.
| | - Miina Ojansivu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| | - Luke van der Koog
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, P.O. Box 196, XB10, 9700 AD Groningen, the Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Thomas E Whittaker
- Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Eoghan M Cunnane
- Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK.
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Molly M Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Materials, Imperial College London, London, UK; Department of Bioengineering, Imperial College London, London, UK; Institute of Biomedical Engineering, Imperial College London, London, UK.
| |
Collapse
|
28
|
Periodontal and Dental Pulp Cell-Derived Small Extracellular Vesicles: A Review of the Current Status. NANOMATERIALS 2021; 11:nano11071858. [PMID: 34361246 PMCID: PMC8308278 DOI: 10.3390/nano11071858] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are membrane-bound lipid particles that are secreted by all cell types and function as cell-to-cell communicators through their cargos of protein, nucleic acid, lipids, and metabolites, which are derived from their parent cells. There is limited information on the isolation and the emerging therapeutic role of periodontal and dental pulp cell-derived small EVs (sEVs, <200 nm, or exosome). In this review, we discuss the biogenesis of three EV subtypes (sEVs, microvesicles and apoptotic bodies) and the emerging role of sEVs from periodontal ligament (stem) cells, gingival fibroblasts (or gingival mesenchymal stem cells) and dental pulp cells, and their therapeutic potential in vitro and in vivo. A review of the relevant methodology found that precipitation-based kits and ultracentrifugation are the two most common methods to isolate periodontal (dental pulp) cell sEVs. Periodontal (and pulp) cell sEVs range in size, from 40 nm to 2 μm, due to a lack of standardized isolation protocols. Nevertheless, our review found that these EVs possess anti-inflammatory, osteo/odontogenic, angiogenic and immunomodulatory functions in vitro and in vivo, via reported EV cargos of EV–miRNAs, EV–circRNAs, EV–mRNAs and EV–lncRNAs. This review highlights the considerable therapeutic potential of periodontal and dental pulp cell-derived sEVs in various regenerative applications.
Collapse
|
29
|
Bone Regeneration Improves with Mesenchymal Stem Cell Derived Extracellular Vesicles (EVs) Combined with Scaffolds: A Systematic Review. BIOLOGY 2021; 10:biology10070579. [PMID: 34202598 PMCID: PMC8301056 DOI: 10.3390/biology10070579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023]
Abstract
Scaffolds associated with mesenchymal stem cell (MSC) derivatives, such as extracellular vesicles (EVs), represent interesting carriers for bone regeneration. This systematic review aims to analyze in vitro and in vivo studies that report the effects of EVs combined with scaffolds in bone regeneration. A methodical review of the literature was performed from PubMed and Embase from 2012 to 2020. Sixteen papers were analyzed; of these, one study was in vitro, eleven were in vivo, and four were both in vitro and in vivo studies. This analysis shows a growing interest in this upcoming field, with overall positive results. In vitro results were demonstrated as both an effect on bone mineralization and proangiogenic ability. The interesting in vitro outcomes were confirmed in vivo. Particularly, these studies showed positive effects on bone regeneration and mineralization, activation of the pathway for bone regeneration, induction of vascularization, and modulation of inflammation. However, several aspects remain to be elucidated, such as the concentration of EVs to use in clinic for bone-related applications and the definition of the real advantages.
Collapse
|
30
|
Marconi GD, Fonticoli L, Rajan TS, Pierdomenico SD, Trubiani O, Pizzicannella J, Diomede F. Epithelial-Mesenchymal Transition (EMT): The Type-2 EMT in Wound Healing, Tissue Regeneration and Organ Fibrosis. Cells 2021; 10:cells10071587. [PMID: 34201858 PMCID: PMC8307661 DOI: 10.3390/cells10071587] [Citation(s) in RCA: 193] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/07/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
The epithelial–mesenchymal transition (EMT) is an essential event during cell development, in which epithelial cells acquire mesenchymal fibroblast-like features including reduced intercellular adhesion and increased motility. EMT also plays a key role in wound healing processes, which are mediated by inflammatory cells and fibroblasts. These cells secrete specific factors that interact with molecules of the extracellular matrix (ECM) such as collagens, laminins, elastin and tenascins. Wound healing follows four distinct and successive phases characterized by haemostasis, inflammation, cell proliferation and finally tissue remodeling. EMT is classified into three diverse subtypes: type-1 EMT, type-2 EMT and type-3 EMT. Type-1 EMT is involved in embryogenesis and organ development. Type-2 EMT is associated with wound healing, tissue regeneration and organ fibrosis. During organ fibrosis, type-2 EMT occurs as a reparative-associated process in response to ongoing inflammation and eventually leads to organ destruction. Type-3 EMT is implicated in cancer progression, which is linked to the occurrence of genetic and epigenetic alterations, in detail the ones promoting clonal outgrowth and the formation of localized tumors. The current review aimed at exploring the role of EMT process with particular focus on type-2 EMT in wound healing, fibrosis and tissue regeneration, as well as some recent progresses in the EMT and tissue regeneration field, including the modulation of EMT by biomaterials.
Collapse
Affiliation(s)
- Guya D. Marconi
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy;
| | - Luigia Fonticoli
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (L.F.); (S.D.P.)
| | - Thangavelu Soundara Rajan
- Department of Biotechnology, School of Life Sciences, Karpagam Academy of Higher Education, Coimbatore 641021, India;
| | - Sante D. Pierdomenico
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (L.F.); (S.D.P.)
| | - Oriana Trubiani
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (L.F.); (S.D.P.)
- Correspondence: (O.T.); (F.D.); Tel.: +39-08713554097 (O.T.); +39-08713554080 (F.D.)
| | | | - Francesca Diomede
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (L.F.); (S.D.P.)
- Correspondence: (O.T.); (F.D.); Tel.: +39-08713554097 (O.T.); +39-08713554080 (F.D.)
| |
Collapse
|
31
|
Murali VP, Holmes CA. Mesenchymal stromal cell-derived extracellular vesicles for bone regeneration therapy. Bone Rep 2021; 14:101093. [PMID: 34095360 PMCID: PMC8166743 DOI: 10.1016/j.bonr.2021.101093] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose To analyze preclinical bone regeneration studies employing mesenchymal stromal cell (MSC)- derived extracellular vesicles (EVs) and highlight any commonalities in EV biomarker expression, miRNA cargo(s) or pathway activation that will aid in understanding the underlying therapeutic mechanisms. Methods Articles employing EVs derived from either MSCs or MSC-like osteogenic stromal cells in preclinical bone regeneration studies are included in this review. Results EVs derived from a variety of MSC types were able to successfully induce bone formation in preclinical models. Many studies failed to perform in-depth EV characterization. The studies with detailed EV characterization data report very different miRNA cargos, even in EVs isolated from the same species and cell types. Few preclinical studies have analyzed the underlying mechanisms of MSC-EV therapeutic action. Conclusion There is a critical need for mechanistic preclinical studies with thorough EV characterization to determine the best therapeutic MSC-EV source for bone regeneration therapies. Issues including controlled EV delivery, large scale production, and proper storage also need to be addressed before EV-based bone regeneration therapies can be translated for clinical bone repair. EVs from different MSC sources successfully regenerate bone in preclinical models. Studies were reviewed to find commonalities in EV cargo(s)/pathways activated in MSC-EV-based bone regeneration therapies. Issues that need to be overcome to enable clinical translation of EV-based therapies were addressed.
Collapse
Affiliation(s)
- Vishnu Priya Murali
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA
| | - Christina A Holmes
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA
| |
Collapse
|
32
|
Oral Bone Tissue Regeneration: Mesenchymal Stem Cells, Secretome, and Biomaterials. Int J Mol Sci 2021; 22:ijms22105236. [PMID: 34063438 PMCID: PMC8156243 DOI: 10.3390/ijms22105236] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, tissue engineering has become one of the most studied medical fields. Even if bone shows self-remodeling properties, in some cases, due to injuries or anomalies, bone regeneration can be required. In particular, oral bone regeneration is needed in the dentistry field, where the functional restoration of tissues near the tooth represents a limit for many dental implants. In this context, the application of biomaterials and mesenchymal stem cells (MSCs) appears promising for bone regeneration. This review focused on in vivo studies that evaluated bone regeneration using biomaterials with MSCs. Different biocompatible biomaterials were enriched with MSCs from different sources. These constructs showed an enhanced bone regenerative power in in vivo models. However, we discussed also a future perspective in tissue engineering using the MSC secretome, namely the conditioned medium and extracellular vesicles. This new approach has already shown promising results for bone tissue regeneration in experimental models.
Collapse
|
33
|
Tan L, Cao Z, Chen H, Xie Y, Yu L, Fu C, Zhao W, Wang Y. Curcumin reduces apoptosis and promotes osteogenesis of human periodontal ligament stem cells under oxidative stress in vitro and in vivo. Life Sci 2021; 270:119125. [PMID: 33513394 DOI: 10.1016/j.lfs.2021.119125] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022]
Abstract
AIMS Human periodontal ligament stem cells (hPDLSCs) tether the teeth to the surrounding bone and are considered as major functional stem cells responsible for regeneration of the alveolar bone and periodontal ligament tissue. However, the outcome of stem cell regenerative therapy is affected by the survival rate and their differentiation potential of transplanted cells. This is primarily because of local oxidative stress and chronic inflammation at the transplantation site. Therefore, our study aimed to explore whether a natural antioxidant, curcumin could increase the tissue regeneration ability of transplanted hPDLSCs. MAIN METHODS A hydrogen peroxide environment and a rat cranial bone defect model were built to mimic the oxidative stress conditions in vitro and in vivo, respectively. We evaluated the effect of curcumin on oxidative status, apoptosis, mitochondrial function and osteogenic differentiation of H2O2-stimulated hPDLSCs in vitro. We also measured the effect of curcumin on cell viability and bone repair ability of transplanted hPDLSCs in vivo. KEY FINDINGS Our data showed that curcumin enhanced cell proliferation, reduced the reactive oxygen species (ROS) levels and apoptosis, maintained the standard mitochondrial structure and function, and promoted osteogenic differentiation of H2O2-stimulated hPDLSCs. The extracellular regulated protein kinases 1/2 (Erk1/2) signaling pathway was determined to be involved in the osteogenic differentiation of the H2O2-stimulated hPDLSCs. Moreover, curcumin enhanced the viability and the bone repair ability of hPDLSCs in vivo. SIGNIFICANCE Curcumin reduced apoptosis and promoted osteogenesis of the hPDLSCs under oxidative stress, and might therefore have a potential clinical use with respect to tissue regeneration.
Collapse
Affiliation(s)
- Lingping Tan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Zeyuan Cao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Huan Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Yunyi Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Le Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Chuanqiang Fu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China.
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, 56 Lingyuanxi Road, Guangzhou 510055, China.
| |
Collapse
|
34
|
Li N, Li Z, Wang Y, Chen Y, Ge X, Lu J, Bian M, Wu J, Yu J. CTP-CM enhances osteogenic differentiation of hPDLSCs via NF-κB pathway. Oral Dis 2021; 27:577-588. [PMID: 32691476 DOI: 10.1111/odi.13567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 04/02/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The conditioned medium of calcined tooth powder (CTP-CM) is a type of biomimetic mineralized material and well contributing to bone healing and bone formation in vivo. However, little is known about the effect of CTP-CM on human periodontal ligament stem cells (hPDLSCs) as well as the underlying mechanisms. METHODS ALP activity assay was conducted to select the concentration with the highest ALP level, which was used for the following experiments. Cell proliferation was measured by cell counting kit-8 assay and flow cytometry analysis. Expression levels of osteogenic markers in CTP-CM-induced hPDLSCs were evaluated with real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), immunofluorescence staining, and Western blot. Mineralization of CTP-CM-induced hPDLSCs was evaluated by alizarin red staining. Furthermore, the involvement of NF-κB pathway was examined by immunofluorescence staining and Western blot. RESULTS 20 μg/ml was selected for the further experiments. Functional studies demonstrated that CTP-CM exerted almost no influence on the proliferation of hPDLSCs and CTP-CM increased the osteogenic differentiation of hPDLSCs. Mechanistically, CTP-CM leads to activation of NF-κB signaling pathway. When treated with BMS345541, the osteogenic differentiation of CTP-CM-treated hPDLSCs was significantly attenuated. CONCLUSION CTP-CM can promote the osteogenic differentiation of hPDLSCs via activating NF-κB pathway.
Collapse
Affiliation(s)
- Na Li
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zehan Li
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
- Stem Cells & Regenerative Medicine Laboratory, Peninsula Dental School, Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Yanqiu Wang
- Endodontic Department, School of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingyun Ge
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jiamin Lu
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
| | - Minxia Bian
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jintao Wu
- Endodontic Department, School of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jinhua Yu
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
- Endodontic Department, School of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Murali VP, Holmes CA. Biomaterial-based extracellular vesicle delivery for therapeutic applications. Acta Biomater 2021; 124:88-107. [PMID: 33454381 DOI: 10.1016/j.actbio.2021.01.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicle (EV)- based therapies have been successfully tested in preclinical models for several biomedical applications, including tissue engineering, drug delivery and cancer therapy. However, EVs are most commonly delivered via local or systemic injection, which results in rapid clearance. In order to prolong the retention of EVs at target site and improve their therapeutic efficacy, biomaterial-based delivery systems are being investigated. This review discusses the various biomaterial-based systems that have been used to deliver EVs for therapeutic applications, specifically highlighting any strategies for controlled release. Further, challenges to clinical translation of biomaterial-based EV delivery systems are also discussed.
Collapse
Affiliation(s)
- Vishnu Priya Murali
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA.
| | - Christina A Holmes
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA.
| |
Collapse
|
36
|
Yi G, Ma Y, Chen Y, Yang X, Yang B, Tian W. A Review of the Functions of Matrix Vesicles in Periodontal Tissues. Stem Cells Dev 2021; 30:165-176. [PMID: 33349125 DOI: 10.1089/scd.2020.0155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Periodontal tissues consist of cementum, periodontal ligaments, and alveolar bone, which provide indispensable support for physiological activities involving mastication, swallowing, and pronunciation. The formation of periodontal tissues requires a complex process, during which a close relationship with biomineralization is noticeable. Alveolar bone and cementum are physically hard, both of which are generated from biomineralization and possess the exact mechanical properties resembling other hard tissues. However, when periodontitis, congenital abnormalities, periapical diseases, and other pathological conditions affect the organism, the most common symptom, alveolar bone defect, is always unavoidable, which results in difficulties for current clinical treatment. Thus, exploring effective therapies to improve the prognosis is important. Matrix vesicles (MVs), a special subtype of extracellular vesicles related to histogenesis, are widely produced by the stem cells of developing hard tissues. With the assistance of the enzymes and transporters contained within them, MVs can construct the extracellular matrix and an adequate microenvironment, thus promoting biomineralization and periodontal development. Presently, MVs can be effectively extracted and delivered by scaffolds and generate hard tissues in vitro and in vivo, which are expected to be translated into therapies for alveolar bone defects. In this review, we generalize recent research progress on MV morphology, molecular composition, biological mechanism, and, in particular, the biological functions in periodontal development. In addition to the above unique roles of MVs, we further describe the available MV-related biotechnologies and achievements that make them promising for coping with existing problems and improving the treatment of alveolar bone defects.
Collapse
Affiliation(s)
- Genzheng Yi
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yue Ma
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yan Chen
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xueting Yang
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Bo Yang
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Weidong Tian
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
37
|
Silvestro S, Gugliandolo A, Chiricosta L, Diomede F, Trubiani O, Bramanti P, Pizzicannella J, Mazzon E. MicroRNA Profiling of HL-1 Cardiac Cells-Derived Extracellular Vesicles. Cells 2021; 10:cells10020273. [PMID: 33573156 PMCID: PMC7912193 DOI: 10.3390/cells10020273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 11/20/2022] Open
Abstract
HL-1 is a cell line that shows a phenotype similar to adult cardiomyocytes. All major cardiac cell types release extracellular vesicles (EVs) that emerge as key mediators of intercellular communication. EVs can mediate intercellular cross-talk through the transfer of specific microRNAs (miRNAs). MiRNAs are known to play important regulatory roles during tissue differentiation and regeneration processes. Furthermore, miRNAs have recently been shown to be involved in the proliferation of adult cardiomyocytes. In this context, the purpose of this study was to analyze the transcriptomic profile of miRNAs expressed from HL-1 cardiac muscle cell-derived EVs, using next generation sequencing (NGS). Specifically, our transcriptomic analysis showed that the EVs derived from our HL-1 cells contained miRNAs that induce blood vessel formation and increase cell proliferation. Indeed, our bioinformatics analysis revealed 26 miRNAs expressed in EVs derived from our HL-1 that target genes related to cardiovascular development. In particular, their targets are enriched for the following biological processes related to cardiovascular development: heart morphogenesis, positive regulation of angiogenesis, artery development, ventricular septum development, cardiac atrium development, and myoblast differentiation. Consequently, EVs could become important in the field of regenerative medicine.
Collapse
Affiliation(s)
- Serena Silvestro
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; (F.D.); (O.T.)
| | - Oriana Trubiani
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; (F.D.); (O.T.)
| | - Placido Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
- Correspondence: ; Tel.: +39-090-6012-8172
| |
Collapse
|
38
|
Ascorbic Acid: A New Player of Epigenetic Regulation in LPS- gingivalis Treated Human Periodontal Ligament Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6679708. [PMID: 33542783 PMCID: PMC7840256 DOI: 10.1155/2021/6679708] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/21/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022]
Abstract
Periodontitis is usually sustained from microorganism of oral cavity, like Porphyromonas gingivalis (P. gingivalis). Periodontal disease is an infectious disease that afflicts a large number of people. Researches are investigating on the mesenchymal stem cells (MSCs) response to inflammatory events in combination with antioxidant substances. In particular, ascorbic acid (AA) increased cell proliferation, upregulated the cells pluripotency marker expression, provide a protection from inflammation, and induced the regeneration of periodontal ligament tissue. The purpose of the present research was to investigate the effects of AA in primary culture of human periodontal ligament stem cells (hPDLSCs) exposed to P. gingivalis lipopolysaccharide (LPS-G). The effect of AA on hPDLSCs exposed to LPS-G was determined through the cell proliferation assay. The molecules involved in the inflammatory pathway and epigenetic regulation have been identified using immunofluorescence and Western blot analyses. miR-210 level was quantified by qRT-PCR, and the ROS generation was finally studied. Cells co-treated with LPS-G and AA showed a restoration in terms of cell proliferation. The expression of NFκB, MyD88, and p300 was upregulated in LPS-G exposed cells, while the expression was attenuated in the co-treatment with AA. DNMT1 expression is attenuated in the cells exposed to the inflammatory stimulus. The level of miR-210 was reduced in stimulated cells, while the expression was evident in the hPDLSCs co-treated with LPS-G and AA. In conclusion, the AA could enhance a protective effect in in vitro periodontitis model, downregulating the inflammatory pathway and ROS generation and modulating the miR-210 level.
Collapse
|
39
|
Gholami L, Nooshabadi VT, Shahabi S, Jazayeri M, Tarzemany R, Afsartala Z, Khorsandi K. Extracellular vesicles in bone and periodontal regeneration: current and potential therapeutic applications. Cell Biosci 2021; 11:16. [PMID: 33436061 PMCID: PMC7802187 DOI: 10.1186/s13578-020-00527-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022] Open
Abstract
Oral mesenchymal stem cells (MSCs) and their secretomes are considered important factors in the field of medical tissue engineering and cell free biotherapy due to their ease of access, differentiation potential, and successful therapeutic outcomes. Extracellular vesicles (EVs) and the conditioned medium (CM) from MSCs are gaining more attraction as an alternative to cell-based therapies due to the less ethical issues involved, and their easier acquisition, preservation, long term storage, sterilization, and packaging. Bone and periodontal regenerative ability of EVs and CM have been the focus of some recent studies. In this review, we looked through currently available literature regarding MSCs' EVs or conditioned medium and their general characteristics, function, and regenerative potentials. We will also review the novel applications in regenerating bone and periodontal defects.
Collapse
Affiliation(s)
- Leila Gholami
- Department of Periodontics, Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Science, Semnan, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Shiva Shahabi
- Student Research Committee, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Jazayeri
- Student Research Committee, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rana Tarzemany
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, Canada
| | - Zohreh Afsartala
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.
| |
Collapse
|
40
|
Dental Mesenchymal Stem/Progenitor Cells: A New Prospect in Regenerative Medicine. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
41
|
Ercal P, Pekozer GG. A Current Overview of Scaffold-Based Bone Regeneration Strategies with Dental Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1288:61-85. [PMID: 32185698 DOI: 10.1007/5584_2020_505] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone defects due to trauma or diseases still pose a clinical challenge to be resolved in the current tissue engineering approaches. As an alternative to traditional methods to restore bone defects, such as autografts, bone tissue engineering aims to achieve new bone formation via novel biomaterials used in combination with multipotent stem cells and bioactive molecules. Mesenchymal stem cells (MSCs) can be successfully isolated from various dental tissues at different stages of development including dental pulp, apical papilla, dental follicle, tooth germ, deciduous teeth, periodontal ligament and gingiva. A wide range of biomaterials including polymers, ceramics and composites have been investigated for their potential as an ideal bone scaffold material. This article reviews the properties and the manufacturing methods of biomaterials used in bone tissue engineering, and provides an overview of bone tissue regeneration approaches of scaffold and dental stem cell combinations as well as their limitations.
Collapse
Affiliation(s)
- Pınar Ercal
- Faculty of Dentistry, Department of Oral Surgery, Altinbas University, Istanbul, Turkey.
| | - Gorke Gurel Pekozer
- Faculty of Electrical and Electronics Engineering, Department of Biomedical Engineering, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
42
|
Human Periodontal Ligament Stem Cell-Derived Exosomes Promote Bone Regeneration by Altering MicroRNA Profiles. Stem Cells Int 2020; 2020:8852307. [PMID: 33293963 PMCID: PMC7691010 DOI: 10.1155/2020/8852307] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
The role and underlying mechanism of exosomes derived from human periodontal ligament stem cells (PDLSC) in osteogenesis are unclear. In the present study, we identified the exosomes derived from PDLSCs and found that osteogenic induction can enhance the osteogenic ability of PDLSC-derived exosomes in promoting the osteogenic differentiation of rat bone marrow stem cells (BMSCs). To investigate the underlying mechanism, we analyzed the exosomal miRNA expression profiles of undifferentiated and osteogenic differentiated PDLSCs by RNA sequencing. The results showed that seventy-two miRNAs were upregulated and thirty-five miRNAs were downregulated after osteogenic induction. The results of Gene Ontology analysis and pathway analysis demonstrated that the target genes of differentially expressed exosomal miRNAs participate in the regulation of a variety of biological processes, such as catalytic activity, protein binding, metabolic processes, cell development, and differentiation, and are enriched in osteogenic differentiation-related pathways, such as MAPK signaling, AMPK signaling, and insulin signaling pathways. Our results reveal for the first time that the exosomal miRNAs derived from osteogenic differentiated PDLSCs may promote the osteogenic differentiation of BMSCs, which provides a basis for further research on the regulatory function of exosomal miRNA of PDLSCs during osteogenesis.
Collapse
|
43
|
Jiang M, Liu L, Liu R, Lam KS, Lane NE, Yao W. A new anabolic compound, LLP2A-Ale, reserves periodontal bone loss in mice through augmentation of bone formation. BMC Pharmacol Toxicol 2020; 21:76. [PMID: 33187558 PMCID: PMC7664094 DOI: 10.1186/s40360-020-00454-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/27/2020] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Currently, there are no effective medications to reverse periodontal disease (PD)-induced bone loss. The objective of this study was to test a new anabolic compound, LLP2A-Ale, or with the combination treatment of mesenchymal stromal cell (MSC), in the treatment of bone loss secondary to PD. METHODS PD was induced in mice by placing a ligature around the second right molar. At one week after disease induction, the mice were treated with placebo, LLP2A-Ale, MSCs, or combination of LLP2A-Ale + MSCs, and euthanized at week 4. RESULTS We found that PD induced alveolar bone loss that was associated with reduced bone formation. LLP2A-Ale alone or in combination with MSCs sustained alveolar bone formation and reversed alveolar bone loss. Additionally, PD alone caused systemic inflammation and increased the circulating levels of G-CSF, IP-10, MIP-1a, and MIP2, which were suppressed by LLP2A-Ale +/- MSCs. LLP2A-Ale +/- MSCs increased bone formation at the peripheral skeletal site (distal femur), which was otherwise suppressed by PD. CONCLUSION Our findings indicated that LLP2A-Ale treatment rescued alveolar bone loss caused by PD, primarily by increasing bone formation. LLP2A-Ale also attenuated the circulating levels of a series of inflammatory cytokines and reversed the PD-induced suppression of systemic bone formation.
Collapse
Affiliation(s)
- Min Jiang
- Department of Internal Medicine, University of California, Davis Medical Center, 4625 2nd Avenue, Sacramento, CA, 95817, USA
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Lixian Liu
- Department of Internal Medicine, University of California, Davis Medical Center, 4625 2nd Avenue, Sacramento, CA, 95817, USA
- Yunan Vocational and Technical College of Agriculture, Kunming, 650031, Yunan, China
| | - Ruiwu Liu
- Department of Biochemistry & Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry & Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Nancy E Lane
- Department of Internal Medicine, University of California, Davis Medical Center, 4625 2nd Avenue, Sacramento, CA, 95817, USA
| | - Wei Yao
- Department of Internal Medicine, University of California, Davis Medical Center, 4625 2nd Avenue, Sacramento, CA, 95817, USA.
| |
Collapse
|
44
|
Alqurashi H, Ortega Asencio I, Lambert DW. The Emerging Potential of Extracellular Vesicles in Cell-Free Tissue Engineering and Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:530-538. [PMID: 33126845 DOI: 10.1089/ten.teb.2020.0222] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (Evs) are membrane-enclosed vesicles secreted by all cell types that mediate cell-cell communication via their protein, lipid, carbohydrate, and nucleic acid (RNA, DNA) cargo. EVs are involved in a multitude of physiological processes, including development, cell differentiation, and angiogenesis, and have been implicated in tissue repair. Thus, they have been suggested to offer opportunities for the development of novel cell-free tissue engineering (TE) approaches. In this review, we provide an overview of current understanding and emerging applications of EVs in TE and address opportunities and challenges for clinical translation. In addition, we discuss systemic and local routes of delivery of EVs and the advantages and disadvantages of different biomaterials in providing a substrate for the sustained release of EVs in vivo. Impact statement Extracellular vesicles (EVs) are nanoscale, membrane-bound vesicles released by most, if not all, cells in the body. They are implicated in a wide range of physiological processes and diseases ranging from cancer to neurodegeneration, and hold huge potential as mediators of tissue regeneration. This has led to an explosion of interest in using EVs in a variety of tissue engineering applications. In this review, we provide an overview of current progress in the field and highlight the opportunities and challenges of harnessing the potential of EVs in regenerative medicine.
Collapse
Affiliation(s)
- Hatim Alqurashi
- School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom.,College of Dentistry, King Faisal University, Alhassa, Saudi Arabia
| | - Ilida Ortega Asencio
- School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom
| | - Daniel W Lambert
- School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
45
|
Diameters and Fluorescence Calibration for Extracellular Vesicle Analyses by Flow Cytometry. Int J Mol Sci 2020; 21:ijms21217885. [PMID: 33114229 PMCID: PMC7660682 DOI: 10.3390/ijms21217885] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in the intercellular crosstalk. Mesenchymal stem cell-derived EVs (MSC-EVs), displaying promising therapeutic roles, contribute to the strong rationale for developing EVs as an alternative therapeutic option. EV analysis still represents one of the major issues to be solved in order to translate the use of MSC-EV detection in clinical settings. Even if flow cytometry (FC) has been largely applied for EV studies, the lack of consensus on protocols for FC detection of EVs generated controversy. Standard FC procedures, based on scatter measurements, only allows the detection of the “tip of the iceberg” of all EVs. We applied an alternative FC approach based on the use of a trigger threshold on a fluorescence channel. The EV numbers obtained by the application of the fluorescence triggering resulted significantly higher in respect to them obtained from the same samples acquired by placing the threshold on the side scatter (SSC) channel. The analysis of EV concentrations carried out by three different standardized flow cytometers allowed us to achieve a high level of reproducibility (CV < 20%). By applying the here-reported method highly reproducible results in terms of EV analysis and concentration measurements were obtained.
Collapse
|
46
|
Potential Therapeutic Effects of Exosomes in Regenerative Endodontics. Arch Oral Biol 2020; 120:104946. [PMID: 33129129 DOI: 10.1016/j.archoralbio.2020.104946] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/07/2020] [Accepted: 10/04/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This review aims to describe the basic characteristics of exosomes, and summarize their possible source and potential biological effects in pulp regeneration, providing new insights into the therapeutic role of exosomes for regenerative endodontics in the future. DESIGN A comprehensive review of scientific literature related to exosomes potentially used for pulp regeneration was conducted. RESULTS Dental mesenchymal stem cells (MSCs) play an important role in dental pulp regeneration. MSC-derived exosomes, as important biotransmitters in intercellular communication, have been shown to replicate the therapeutic effects of their parental cells. These exosomes have better stability, lower immunogenicity, higher safety and clinical efficiency, making it possible to apply them in pulp regeneration. Existing research suggests that exosomes could trigger the regeneration of dentin/pulp-like tissue in vivo, which may attribute to their role in promoting pulp angiogenesis, regulating dental cell proliferation, migration and differentiation, and providing neuroprotection. CONCLUSIONS The applications of exosomes in the treatment of pulp regeneration have great potential, and exosomes may become ideal therapeutic biomaterial in regenerative endodontics.
Collapse
|
47
|
Fraxinellone alleviates inflammation and promotes osteogenic differentiation in lipopolysaccharide-stimulated periodontal ligament stem cells by regulating the bone morphogenetic protein 2/Smad pathway. Arch Oral Biol 2020; 121:104927. [PMID: 33137544 DOI: 10.1016/j.archoralbio.2020.104927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of the present study was to investigate the role of fraxinellone in periodontitis and identify its potential mechanisms. DESIGN Lipopolysaccharide-induced periodontal ligament stem cells (PDLSCs) was employed to simulate the periodontitis in vitro. The levels of inflammatory factors were evaluated. After treatment with fraxinellone, alkaline phosphatase activity was determined. Additionally, calcium nodules staining was evaluated by alizarin red staining and the expression of osteogenesis differentiation-associated proteins was detected using western blot analysis. Moreover, the levels of proteins in bone morphogenetic protein 2 (BMP2)/Smad pathway were measured. Subsequently, BMP2 was silenced by transfection with small hairpin RNA to explore the underlying mechanisms of fraxinellone in lipopolysaccharide-induced PDLSCs. RESULTS Lipopolysaccharide stimulation significantly upregulated the levels of inflammatory factors, which were inhibited by fraxinellone intervention. Moreover, fraxinellone notably promoted osteogenic differentiation and calcification shown by increasing levels of alkaline phosphatase, calcification and osteogenic marker proteins. Furthermore, the expression of BMP2, phosphorylated Smad1 and phosphorylated Smad5 was remarkably upregulated when fraxinellone exposure in lipopolysaccharide-induced PDLSCs. What's more, BMP2 silencing dramatically restored the effects of fraxinellone on inflammation and osteogenic differentiation of PDLSCs stimulated by lipopolysaccharide. CONCLUSION These data demonstrated that fraxinellone alleviates inflammation and promotes osteogenic differentiation in lipopolysaccharide-stimulated PDLSCs by regulating the BMP2/Smad pathway, providing experimental supports for the clinical application of fraxinellone in the treatment of periodontitis.
Collapse
|
48
|
Granz CL, Gorji A. Dental stem cells: The role of biomaterials and scaffolds in developing novel therapeutic strategies. World J Stem Cells 2020; 12:897-921. [PMID: 33033554 PMCID: PMC7524692 DOI: 10.4252/wjsc.v12.i9.897] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/05/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023] Open
Abstract
Dental stem cells (DSCs) are self-renewable cells that can be obtained easily from dental tissues, and are a desirable source of autologous stem cells. The use of DSCs for stem cell transplantation therapeutic approaches is attractive due to their simple isolation, high plasticity, immunomodulatory properties, and multipotential abilities. Using appropriate scaffolds loaded with favorable biomolecules, such as growth factors, and cytokines, can improve the proliferation, differentiation, migration, and functional capacity of DSCs and can optimize the cellular morphology to build tissue constructs for specific purposes. An enormous variety of scaffolds have been used for tissue engineering with DSCs. Of these, the scaffolds that particularly mimic tissue-specific micromilieu and loaded with biomolecules favorably regulate angiogenesis, cell-matrix interactions, degradation of extracellular matrix, organized matrix formation, and the mineralization abilities of DSCs in both in vitro and in vivo conditions. DSCs represent a promising cell source for tissue engineering, especially for tooth, bone, and neural tissue restoration. The purpose of the present review is to summarize the current developments in the major scaffolding approaches as crucial guidelines for tissue engineering using DSCs and compare their effects in tissue and organ regeneration.
Collapse
Affiliation(s)
- Cornelia Larissa Granz
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| | - Ali Gorji
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| |
Collapse
|
49
|
Diomede F, Marconi GD, Fonticoli L, Pizzicannella J, Trubiani O. Stem Cells Secretome from Oral Tissue Could Represent a Promising Therapeutic Approach in COVID-19-Disease? Int J Mol Sci 2020; 21:ijms21186833. [PMID: 32957696 PMCID: PMC7554818 DOI: 10.3390/ijms21186833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
At present, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has quickly become a health emergency because no specifics vaccines or drugs, at this moment, are available. Recent studies have shown that the transplantation of mesenchymal stem cells (MSCs) into Coronavirus Disease 2019 (COVID-19) patients could represent a promising strategy for the development of new therapeutic methods. We speculate and suggest that the secretome of human Oral Tissue Stem Cells (hOTSCs), for their immunomodulatory and anti-inflammatory specific properties, could exert beneficial effects on the COVID-19 patients through an innovative aerosolisation technique. This non-invasive technique can offer multiple advantages in prophylaxis, as well as the prevention and treatment of severe epidemic respiratory syndrome with minimum risk and optimal therapeutic effects. This has the potential to create a novel pathway towards immunomodulatory therapy for the treatment of COVID-19 positive patients.
Collapse
Affiliation(s)
- Francesca Diomede
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (F.D.); (G.D.M.); (L.F.)
| | - Guya D. Marconi
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (F.D.); (G.D.M.); (L.F.)
| | - Luigia Fonticoli
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (F.D.); (G.D.M.); (L.F.)
| | | | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy; (F.D.); (G.D.M.); (L.F.)
- Correspondence: ; Tel.: +39-0871-355-4097
| |
Collapse
|
50
|
Biomaterials and extracellular vesicles in cell-free therapy for bone repair and regeneration: Future line of treatment in regenerative medicine. MATERIALIA 2020. [DOI: 10.1016/j.mtla.2020.100736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|