1
|
Dong X, Chen Q, Du H, Qiu L. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin Suppresses Mesenchymal Cell Proliferation and Migration Through miR-214-3p in Cleft Palate. Cleft Palate Craniofac J 2024:10556656241286314. [PMID: 39314083 DOI: 10.1177/10556656241286314] [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: 09/25/2024] Open
Abstract
OBJECTIVE The aetiology of CL/P is complicated, with both genetic and environmental factors. This study aimed to investigate the association between TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) exposure and changes in the expression of miR-214-3p in the context of cleft palate. DESIGN In this study, we established a fetal mouse cleft palate model using TCDD and differentially expressed miRNAs were analysed by microarray analysis and verified by qRT-PCR. Finally, we demonstrated the effects of TCDD and microRNAs on the proliferation and migration of mesenchymal cells by using CCK8, EDU, Transwell, and wound-healing assays. RESULTS Our findings revealed significant upregulation of miRNAs such as miR-214-3p, miR-296-5p, and miR-33-5p in the TCDD intervention group, while miRNAs like miR-92a-3p, miR-126a-3p, and miR-411-5p were significantly downregulated. Notably, qRT-PCR testing confirmed a significant difference in miR-214-3P expression. Further investigations involved the overexpression of miR-214-3p, reducing cell proliferation and migration in primary mouse embryonic palatal mesenchymal (MEPM) cells. CONCLUSIONS These results are consistent with the finding that TCDD suppresses palatal mesenchymal cell proliferation and migration through miR-214-3p. In conclusion, miR-214-3p probably plays a role in TCDD-induced cleft palates in mice.
Collapse
Affiliation(s)
- Xiaobo Dong
- Department of Burn and Plastic Surgery, Children's Hospital of Chongqing Medical University, National Clinical Research Centre for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 404000, P.R. China
| | - Qiang Chen
- Department of Burn and Plastic Surgery, Children's Hospital of Chongqing Medical University, National Clinical Research Centre for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 404000, P.R. China
- Department of Paediatrics Surgery, Chongqing University Three Gorges Hospital, Chongqing 404000 P.R. China
| | - Haojuan Du
- Chongqing Key Laboratory of Pediatrics, Chongqing 404000, P.R. China
| | - Lin Qiu
- Department of Burn and Plastic Surgery, Children's Hospital of Chongqing Medical University, National Clinical Research Centre for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 404000, P.R. China
| |
Collapse
|
2
|
Wei F, Liu H, Wang Y, Li Y, Han S. Engineering macrophages and their derivatives: A new hope for antitumor therapy. Biomed Pharmacother 2024; 177:116925. [PMID: 38878637 DOI: 10.1016/j.biopha.2024.116925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
Macrophages are central to the immune system and are found in nearly all tissues. Recently, the development of therapies based on macrophages has attracted significant interest. These therapies utilize macrophages' key roles in immunity, their ability to navigate biological barriers, and their tendency to accumulate in tumors. This review explores the advancement of macrophage-based treatments. We discuss the bioengineering of macrophages for improved anti-tumor effects, the use of CAR macrophage therapy for targeting cancer cells, and macrophages as vehicles for therapeutic delivery. Additionally, we examine engineered macrophage products, like extracellular vesicles and membrane-coated nanoparticles, for their potential in precise and less toxic tumor therapy. Challenges in moving these therapies from research to clinical practice are also highlighted. The aim is to succinctly summarize the current status, challenges, and future directions of engineered macrophages in cancer therapy.
Collapse
Affiliation(s)
- Fang Wei
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Haiyang Liu
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Yuxiao Wang
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Yan Li
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China.
| | - Shuo Han
- Department of Cardiology, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China.
| |
Collapse
|
3
|
Wang Y, Liu X, Wang B, Sun H, Ren Y, Zhang H. Compounding engineered mesenchymal stem cell-derived exosomes: A potential rescue strategy for retinal degeneration. Biomed Pharmacother 2024; 173:116424. [PMID: 38471273 DOI: 10.1016/j.biopha.2024.116424] [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: 01/06/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
The prevalence of retinal degenerative diseases, including age-related macular degeneration and retinitis pigmentosa, has been increasing globally and is linked to the aging population and improved life expectancy. These diseases are characterized by chronic, progressive neuronal damage or depletion of the photoreceptor cells in the retina, and limited effective treatment options are currently available. Mesenchymal stem cell-derived exosomes (MSC-EXOs) containing cytokines, growth factors, lipids, mRNA, and miRNA, which act as mediators of intercellular communication transferring bioactive molecules to recipient cells, offer an appealing, non-cellular nanotherapeutic approach for retinal degenerative diseases. However, treatment specificity is compromised due to their high heterogeneity in size, content, functional effects, and parental cellular source. To improve this, engineered MSC-EXOs with increased drug-loading capacity, targeting ability, and resistance to bodily degradation and elimination have been developed. This review summarizes the recent advances in miRNAs of MSC-EXOs as a treatment for retinal degeneration, discussing the strategies and methods for engineering therapeutic MSC-EXOs. Notably, to address the single functional role of engineered MSC-EXOs, we propose a novel concept called "Compound Engineered MSC-EXOs (Co-E-MSC-EXOs)" along with its derived potential therapeutic approaches. The advantages and challenges of employing Co-E-MSC-EXOs for retinal degeneration in clinical applications, as well as the strategies and issues related to them, are also highlighted.
Collapse
Affiliation(s)
- Yao Wang
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China.
| | - Xianning Liu
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China
| | - Bei Wang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Hanhan Sun
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yiqian Ren
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China
| | - Hongbing Zhang
- Shaanxi Provincial Clinical Research Center for Ophthalmology Diseases, the First Affiliated Hospital of Northwest University, Xi'an No.1 hospital, Xi'an, Shaanxi, China; Shaanxi Key Laboratory of Ophthalmology, Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi 710002, China.
| |
Collapse
|
4
|
Balakrishnan R, Subbarayan R, Shrestha R, Chauhan A, Krishnamoorthy L. Exploring platelet-derived microvesicles in vascular regeneration: unraveling the intricate mechanisms and molecular mediators. Mol Biol Rep 2024; 51:393. [PMID: 38446325 DOI: 10.1007/s11033-024-09302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/30/2024] [Indexed: 03/07/2024]
Abstract
Microvesicles (MVs) serve as biomarkers and transmitters for cell communication and also act as essential contributors to diseases. Platelets release microvesicles when activated voluntarily, making them a significant source. Platelet-derived microvesicles possess a range of characteristics similar to their parent cells and were shown to exert regulatory impacts on vascular and immunological cells. MVs can alter the activity of recipient cells by transferring their internal components. Furthermore, it has been identified that microvesicles derived from platelets possess the ability to exert immunomodulatory effects on different kinds of cells. Recent research has shown that microvesicles have a bidirectional influence of harming and preventing the receptor cells. Nevertheless, the specific characteristics of the active molecules responsible for this phenomenon are still unknown. The primary focus of this review was to explore the mechanism of vascular tissue regeneration and the specific molecules that play a role in mediating various biological effects throughout this process. These molecules exert their effects by influencing autophagy, apoptosis, and inflammatory pathways.
Collapse
Affiliation(s)
- Ranjith Balakrishnan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India.
| | | | - Ankush Chauhan
- Faculty of Allied Health Sciences, Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Loganathan Krishnamoorthy
- FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
5
|
Thomas J, Sun J, Montclare JK. Constructing Nucleic Acid Delivering Lipoproteoplexes from Coiled-Coil Supercharged Protein and Cationic Liposomes. Methods Mol Biol 2024; 2720:191-207. [PMID: 37775667 DOI: 10.1007/978-1-0716-3469-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
The safe and efficient delivery of nucleic acids is crucial for both clinical applications of gene therapy and pre-clinical laboratory research. Such delivery strategies rely on vectors to condense nucleic acid payloads and escort them into the cell without being degraded in the extracellular environment; however, the construction and utilization of these vectors can be difficult and time-consuming. Here, we detail the steps involved in the rapid, laboratory-scale production and assessment of a versatile, nucleic acid delivery vehicle, known as the lipoproteoplex. In this chapter, we outline: (1) the recombinant synthesis and subsequent purification of the supercharged coiled-coil protein component known as N8; (2) the synthesis of cationic liposomes from dioleoyl-3-trimethylammonium propane (DOTAP) and sodium cholate; (3) and finally a protocol for the delivery of a model siRNA cargo into a cultured cell line.
Collapse
Affiliation(s)
- Joseph Thomas
- Department of Biomedical Engineering, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY, USA
| | - Jonathan Sun
- Department of Chemistry, New York University, New York, NY, USA
- Department of Radiology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY, USA.
- Department of Chemistry, New York University, New York, NY, USA.
- Department of Radiology, State University of New York Downstate Medical Center, Brooklyn, NY, USA.
- Department of Biomaterials, New York University College of Dentistry, New York, NY, USA.
| |
Collapse
|
6
|
Zhao X, Peng X, Wang Z, Zheng X, Wang X, Wang Y, Chen J, Yuan D, Liu Y, Du J. MicroRNAs in Small Extracellular Vesicles from Amniotic Fluid and Maternal Plasma Associated with Fetal Palate Development in Mice. Int J Mol Sci 2023; 24:17173. [PMID: 38139002 PMCID: PMC10743272 DOI: 10.3390/ijms242417173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/25/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Cleft palate (CP) is a common congenital birth defect. Cellular and morphological processes change dynamically during palatogenesis, and any disturbance in this process could result in CP. However, the molecular mechanisms steering this fundamental phase remain unclear. One study suggesting a role for miRNAs in palate development via maternal small extracellular vesicles (SEVs) drew our attention to their potential involvement in palatogenesis. In this study, we used an in vitro model to determine how SEVs derived from amniotic fluid (ASVs) and maternal plasma (MSVs) influence the biological behaviors of mouse embryonic palatal mesenchyme (MEPM) cells and medial edge epithelial (MEE) cells; we also compared time-dependent differential expression (DE) miRNAs in ASVs and MSVs with the DE mRNAs in palate tissue from E13.5 to E15.5 to study the dynamic co-regulation of miRNAs and mRNAs during palatogenesis in vivo. Our results demonstrate that some pivotal biological activities, such as MEPM proliferation, migration, osteogenesis, and MEE apoptosis, might be directed, in part, by stage-specific MSVs and ASVs. We further identified interconnected networks and key miRNAs such as miR-744-5p, miR-323-5p, and miR-3102-5p, offering a roadmap for mechanistic investigations and the identification of early CP biomarkers.
Collapse
Affiliation(s)
- Xige Zhao
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Xia Peng
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Zhiwei Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Xiaoyu Zheng
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Xiaotong Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Yijia Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Jing Chen
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Dong Yuan
- Department of Geriatric Dentistry, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China;
| | - Ying Liu
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Juan Du
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
- Department of Geriatric Dentistry, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China;
| |
Collapse
|
7
|
Huang L, Wu E, Liao J, Wei Z, Wang J, Chen Z. Research Advances of Engineered Exosomes as Drug Delivery Carrier. ACS OMEGA 2023; 8:43374-43387. [PMID: 38027310 PMCID: PMC10666244 DOI: 10.1021/acsomega.3c04479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/05/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Exosomes are nanoscale vesicles secreted by living cells that have similar membrane composition to parental cells and carry a variety of proteins, lipids, and nucleic acids. Therefore, exosomes have certain biological activities and play an important role in intercellular communication. On the basis of its potential as a carrier for drug delivery systems, exosomes have been engineered to compensate for the shortage of natural exosomes through various engineering strategies for improving drug delivery efficiency, enhancing targeting to tissues and organs, and extending the circulating half-life of exosomes. This review focuses on the engineered exosomes loading drugs through different strategies, discussions on exosome surface modification strategies, and summarizes the advantages and disadvantages of different strategies. In addition, this review provides an overview of the recent applications of engineered exosomes in a number of refractory and relapsable diseases. This review has the potential to provide a reference for further research and development of engineered exosomes.
Collapse
Affiliation(s)
- Lianghui Huang
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Enguang Wu
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Jiawei Liao
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Zongyi Wei
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Jin Wang
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Zhenhua Chen
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| |
Collapse
|
8
|
Yao J, Zou P, Cui Y, Quan L, Gao C, Li Z, Gong W, Yang M. Recent Advances in Strategies to Combat Bacterial Drug Resistance: Antimicrobial Materials and Drug Delivery Systems. Pharmaceutics 2023; 15:pharmaceutics15041188. [PMID: 37111673 PMCID: PMC10141387 DOI: 10.3390/pharmaceutics15041188] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Bacterial infection is a common clinical disease. Antibiotics have saved countless lives since their discovery and are a powerful weapon in the fight against bacteria. However, with the widespread use of antibiotics, the problem of drug resistance now poses a great threat to human health. In recent years, studies have investigated approaches to combat bacterial resistance. Several antimicrobial materials and drug delivery systems have emerged as promising strategies. Nano-drug delivery systems for antibiotics can reduce the resistance to antibiotics and extend the lifespan of novel antibiotics, and they allow targeting drug delivery compared to conventional antibiotics. This review highlights the mechanistic insights of using different strategies to combat drug-resistant bacteria and summarizes the recent advancements in antimicrobial materials and drug delivery systems for different carriers. Furthermore, the fundamental properties of combating antimicrobial resistance are discussed, and the current challenges and future perspectives in this field are proposed.
Collapse
Affiliation(s)
- Jiaxin Yao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Pengfei Zou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yanan Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liangzhu Quan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meiyan Yang
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
9
|
Liu F, Sun T, An Y, Ming L, Li Y, Zhou Z, Shang F. The potential therapeutic role of extracellular vesicles in critical-size bone defects: Spring of cell-free regenerative medicine is coming. Front Bioeng Biotechnol 2023; 11:1050916. [PMID: 36733961 PMCID: PMC9887316 DOI: 10.3389/fbioe.2023.1050916] [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: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
In recent years, the incidence of critical-size bone defects has significantly increased. Critical-size bone defects seriously affect patients' motor functions and quality of life and increase the need for additional clinical treatments. Bone tissue engineering (BTE) has made great progress in repairing critical-size bone defects. As one of the main components of bone tissue engineering, stem cell-based therapy is considered a potential effective strategy to regenerate bone tissues. However, there are some disadvantages including phenotypic changes, immune rejection, potential tumorigenicity, low homing efficiency and cell survival rate that restrict its wider clinical applications. Evidence has shown that the positive biological effects of stem cells on tissue repair are largely mediated through paracrine action by nanostructured extracellular vesicles (EVs), which may overcome the limitations of traditional stem cell-based treatments. In addition to stem cell-derived extracellular vesicles, the potential therapeutic roles of nonstem cell-derived extracellular vesicles in critical-size bone defect repair have also attracted attention from scholars in recent years. Currently, the development of extracellular vesicles-mediated cell-free regenerative medicine is still in the preliminary stage, and the specific mechanisms remain elusive. Herein, the authors first review the research progress and possible mechanisms of extracellular vesicles combined with bone tissue engineering scaffolds to promote bone regeneration via bioactive molecules. Engineering modified extracellular vesicles is an emerging component of bone tissue engineering and its main progression and clinical applications will be discussed. Finally, future perspectives and challenges of developing extracellular vesicle-based regenerative medicine will be given. This review may provide a theoretical basis for the future development of extracellular vesicle-based biomedicine and provide clinical references for promoting the repair of critical-size bone defects.
Collapse
Affiliation(s)
- Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen, Guangdong, China
| | - Tianyu Sun
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying An
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture and Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Leiguo Ming
- Department of Research and Development, Shaanxi Zhonghong Institute of Regenerative Medicine, Xi’an, Shaanxi, China
| | - Yinghui Li
- Department of Orthodontics, Stomatological Hospital, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, Tibet, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| | - Fengqing Shang
- Department of Stomatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| |
Collapse
|
10
|
Tian T, Qiao S, Tannous BA. Nanotechnology-Inspired Extracellular Vesicles Theranostics for Diagnosis and Therapy of Central Nervous System Diseases. ACS APPLIED MATERIALS & INTERFACES 2023; 15:182-199. [PMID: 35929960 DOI: 10.1021/acsami.2c07981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Shuttling various bioactive substances across the blood-brain barrier (BBB) bidirectionally, extracellular vesicles (EVs) have been opening new frontiers for the diagnosis and therapy of central nervous system (CNS) diseases. However, clinical translation of EV-based theranostics remains challenging due to difficulties in effective EV engineering for superior imaging/therapeutic potential, ultrasensitive EV detection for small sample volume, as well as scale-up and standardized EV production. In the past decade, continuous advancement in nanotechnology provided extensive concepts and strategies for EV engineering and analysis, which inspired the application of EVs for CNS diseases. Here we will review the existing types of EV-nanomaterial hybrid systems with improved diagnostic and therapeutic efficacy for CNS diseases. A summary of recent progress in the incorporation of nanomaterials and nanostructures in EV production, separation, and analysis will also be provided. Moreover, the convergence between nanotechnology and microfluidics for integrated EV engineering and liquid biopsy of CNS diseases will be discussed.
Collapse
Affiliation(s)
- Tian Tian
- Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts 02129, United States
- Neuroscience Program, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Shuya Qiao
- Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts 02129, United States
- Neuroscience Program, Harvard Medical School, Boston, Massachusetts 02129, United States
| |
Collapse
|
11
|
Chiozzini C, Ridolfi B, Federico M. Extracellular Vesicles and Their Use as Vehicles of Immunogens. Methods Mol Biol 2022; 2504:177-198. [PMID: 35467287 DOI: 10.1007/978-1-0716-2341-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Healthy cells constitutively release lipid bilayered vesicles of different sizes and recognizing different biogenesis, collectively referred to as extracellular vesicles (EVs). EVs can be distinguished in exosomes and microvesicles. Biological and biomedical research on EVs is an emerging field that is rapidly growing. Many EV features including biogenesis, cell uptake, and functions still require unambiguous elucidation. Nevertheless, it has been well established that EVs are involved in communication among cells, tissues, and organs under both healthy and disease conditions by virtue of their ability to deliver macromolecules to target cells. Here, we summarize most recent findings regarding biogenesis, structure, and functions of both exosomes and microvesicles. In addition, the use of EVs as delivery tools to induce CD8+ T cell immunity is addressed compared to current designs exploiting enveloped viral vectors and virus-like particles. Finally, we describe a both safe and original approach conceived for the induction of strong CTL immunity against antigens uploaded in EVs constitutively released by muscle cells.
Collapse
Affiliation(s)
- Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy.
| | - Barbara Ridolfi
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
12
|
Potential Applications and Functional Roles of Exosomes in Cardiometabolic Disease. Pharmaceutics 2021; 13:pharmaceutics13122056. [PMID: 34959338 PMCID: PMC8703910 DOI: 10.3390/pharmaceutics13122056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Despite diagnostic and therapeutic advances, cardiometabolic disease remains the leading cause of death worldwide. Extracellular vesicles (EVs), which include exosomes and microvesicles, have gained particular interest because of their role in metabolic homeostasis and cardiovascular physiology. Indeed, EVs are recognized as critical mediators of intercellular communication in the cardiovascular system. Exosomes are naturally occurring nanocarriers that transfer biological information in the setting of metabolic abnormalities and cardiac dysfunction. The study of these EVs can increase our knowledge on the pathophysiological mechanisms of metabolic disorders and their cardiovascular complications. Because of their inherent properties and composition, exosomes have been proposed as diagnostic and prognostic biomarkers and therapeutics for specific targeting and drug delivery. Emerging fields of study explore the use exosomes as tools for gene therapy and as a cell-free alternative for regenerative medicine. Furthermore, innovative biomaterials can incorporate exosomes to enhance tissue regeneration and engineering. In this work, we summarize the most recent knowledge on the role of exosomes in cardiometabolic pathophysiology while highlighting their potential therapeutic applications.
Collapse
|
13
|
Abstract
Exosomes are extracellular vesicles (EVs) released from cells that are a part of many biological and pathological processes, especially in intercellular communication. These vesicles are involved cell signaling, influence tissue and immune response, and serve as biomarkers for diseases. Most interesting are the exosomes that are released from mesenchymal stem cells (MSCs) for inflammation in joint diseases. Preliminary studies have demonstrated the advantages of using EVs rather than MSCs for cell free therapy. Research on exosomes have shown promising results as biomarkers for tracking the pathogenesis and prognosis of inflammatory arthritis. Therapeutically, animal studies have demonstrated immunosuppression, reversing inflammation, increasing chondrocyte proliferation, and drug delivery properties. The field of exosomes continues to develop and more basic science and clinical studies with safety and efficacy studies are needed.
Collapse
Affiliation(s)
- William Fang
- Department of Orthopaedic Surgery, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - C. Thomas Vangsness
- Department of Orthopaedic Surgery, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA,C. Thomas Vangsness Jr., Department of
Orthopaedic Surgery, Keck School of Medicine, University of Southern California,
1520 San Pablo Street 2000, Los Angeles, CA 90033, USA.
| |
Collapse
|
14
|
Chen M, Xie Y, Luo Y, Xie Y, Wu N, Peng S, Chen Q. Exosomes-a potential indicator and mediator of cleft lip and palate: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1485. [PMID: 34734037 PMCID: PMC8506753 DOI: 10.21037/atm-21-4198] [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: 07/23/2021] [Accepted: 09/07/2021] [Indexed: 11/28/2022]
Abstract
Objective This article summarizes the recent literature on noncoding ribonucleic acids (ncRNAs) in relation to cleft lip with or without palate and exosomes and their usage in craniofacial diseases. Background Cleft lip with or without cleft palate (CL/P) is a common congenital malformation with genetic and environmental risk factors that affects numerous children and families. Surgical procedures can correct deformations; however, residual sequelae remain after surgery. Studies exploring the pathogenesis of CL/P are crucial for its early diagnosis and treatment and can inform treatment strategy decisions, etiology searches, and treatment during pregnancy. Recently, research has shown that most disease-related genes are ncRNAs, which are important transcripts in the human transcriptome. ncRNAs include microRNAs, long noncoding RNAs, and circular RNAs. These ncRNAs play essential roles in various pathophysiological processes, including cell proliferation, migration, apoptosis, and epithelial-mesenchymal transition. Previous studies on protein-coding genes have identified a number of genes related to CL/P; however, the pathogenesis of CL/P has not yet been thoroughly explained. Exosomes are vehicles that transfer various bioactive molecules between cells and represent a new method of intercellular communication. Research has shown that exosomes are related to some craniofacial diseases. Methods We searched the PubMed database for recently published English-language articles using the following keywords: “cleft lip with or without palate,” “noncoding RNA,” “exosomes,” and “craniofacial diseases”. We then reviewed the retrieved articles. Conclusions As exosomes serve as cellular communicators and the palate consists of epithelial and mesenchymal cells, communication between the two cell types may affect its formation. Thus, exosomes could represent a new indicator and mediator of CL/P.
Collapse
Affiliation(s)
- Meng Chen
- Department of Paediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yue Xie
- Department of Burn and Plastic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yarui Luo
- Department of Paediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China.,Department of Outpatient, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yimin Xie
- Department of Paediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Na Wu
- Department of Paediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Shulei Peng
- Department of Paediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China.,Department of Sleep Medicine Centre, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qiang Chen
- Department of Paediatric Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| |
Collapse
|
15
|
Kodam SP, Ullah M. Diagnostic and Therapeutic Potential of Extracellular Vesicles. Technol Cancer Res Treat 2021; 20:15330338211041203. [PMID: 34632862 PMCID: PMC8504225 DOI: 10.1177/15330338211041203] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are naturally phospholipid enclosed nanovesicles released by many cells in the body. They are stable in circulation, have low immunogenicity, and act as carriers for functionally active biological molecules. They interact with target organs and bind to the receptors. Their target specificity is important to use EVs as noninvasive diagnostic and prognostic tools. EVs play a vital role in normal physiology and cellular communication. They are known to protect their cargo from degradation, which makes them important drug carriers for targeted drug delivery. Using EVs with markers and tracking their path in systemic circulation can be revolutionary in using them as diagnostic tools. We will discuss the scope of this in this paper. Although there are limitations in EVs isolation and storage, their high biocompatibility will fuel more innovations to overcome these challenges.
Collapse
Affiliation(s)
- Sai Priyanka Kodam
- Institute for Immunity and Transplantation, 158423Stem Cell Biology and Regenerative Medicine, School of Medicine, 6429Stanford University, Palo Alto, California, USA.,School of Medicine, 6429Stanford University, Palo Alto, California, USA
| | - Mujib Ullah
- Institute for Immunity and Transplantation, 158423Stem Cell Biology and Regenerative Medicine, School of Medicine, 6429Stanford University, Palo Alto, California, USA.,School of Medicine, 6429Stanford University, Palo Alto, California, USA
| |
Collapse
|
16
|
Ullah M, Qian NPM, Yannarelli G. Advances in innovative exosome-technology for real time monitoring of viable drugs in clinical translation, prognosis and treatment response. Oncotarget 2021; 12:1029-1031. [PMID: 34084276 PMCID: PMC8169069 DOI: 10.18632/oncotarget.27927] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 02/07/2023] Open
|
17
|
Long-Term Antitumor CD8 + T Cell Immunity Induced by Endogenously Engineered Extracellular Vesicles. Cancers (Basel) 2021; 13:cancers13092263. [PMID: 34066801 PMCID: PMC8125873 DOI: 10.3390/cancers13092263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The induction of an effective immune response against tumor cells is of a great benefit in the battle against cancers. We recently characterized a novel, safe, and cost-effective strategy to induce an efficient CD8+ T cell immune response against potentially whatever antigen. This technique is based on in vivo engineering of exosomes/extracellular vesicles (EVs), i.e., nanovesicles constitutively released by all healthy cells. Immunogenic EVs are generated by intramuscular injection of a DNA vector expressing an EV-anchoring protein fused with the antigen of interest. In this paper, we applied our vaccine platform to counteract the growth of tumors expressing antigens of Human Papilloma Virus (HPV). We demonstrated that this method is instrumental in curing mice already developing HPV-related tumors. In addition, cured mice were shown to resist a second tumor cell implantation. These results could be of relevance for a possible translation into the clinic of our technology. Abstract We developed an innovative method to induce antigen-specific CD8+ T cytotoxic lymphocyte (CTL) immunity based on in vivo engineering of extracellular vesicles (EVs). This approach employs a DNA vector expressing a mutated HIV-1 Nef protein (Nefmut) deprived of the anti-cellular effects typical of the wild-type isoform, meanwhile showing an unusual efficiency of incorporation into EVs. This function persists even when foreign antigens are fused to its C-terminus. In this way, Nefmut traffics large amounts of antigens fused to it into EVs spontaneously released by the recipient cells. We previously provided evidence that mice injected with a DNA vector expressing the Nefmut/HPV16-E7 fusion protein developed an E7-specific CTL immune response as detected 2 weeks after the second immunization. Here, we extended and optimized the anti-HPV16 CD8+ T cell immune response induced by the endogenously engineered EVs, and evaluated the therapeutic antitumor efficacy over time. We found that the co-injection of DNA vectors expressing Nefmut fused with E6 and E7 generated a stronger anti-HPV16 immune response compared to that observed in mice injected with the single vectors. When HPV16-E6 and -E7 co-expressing tumor cells were implanted before immunization, all mice survived at day 44, whereas no mice injected with either void or Nefmut-expressing vectors survived until day 32 after tumor implantation. A substantial part of immunized mice (7 out of 12) cleared the tumor. When the cured mice were re-challenged with a second tumor cell implantation, none of them developed tumors. Both E6- and E7-specific CD8+ T immunities were still detectable at the end of the observation time. We concluded that the immunity elicited by engineered EVs, besides counteracting and curing already developed tumors, was strong enough to guarantee the resistance to additional tumor attacks. These results can be of relevance for the therapy of both metastatic and relapsing tumors.
Collapse
|
18
|
Babaei M, Rezaie J. Application of stem cell-derived exosomes in ischemic diseases: opportunity and limitations. J Transl Med 2021; 19:196. [PMID: 33964940 PMCID: PMC8106139 DOI: 10.1186/s12967-021-02863-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic diseases characterized by an insufficient blood flow that leads to a decrease in oxygen and nutrient uptake by cells have emerged as an important contributor to both disability and death worldwide. Up-regulation of angiogenesis may be a key factor for the improvement of ischemic diseases. This article searched articles in PubMed with the following keywords: stem cells, exosomes, angiogenesis, ischemic diseases either alone or in grouping form. The most relevant selected items were stem cell-derived exosomes and ischemic diseases. A growing body of evidence indicates that stem cells produce exosomes, which is the novel emerging approach to cell-to-cell communication and offers a new standpoint on known therapeutic strategies of ischemic diseases. Exosomes transport biological molecules such as many types of proteins, RNAs, DNA fragments, signaling molecules, and lipids between cells. Different stem cells release exosomes representing beneficial effects on ischemic diseases as they promote angiogenesis both in vitro and in vivo experiments. Application of exosomes for therapeutic angiogenesis opened new opportunities in the regenerative medicine, however, some limitations regarding exosomes isolation and application remain concerned. In addition, most of the experiments were conducted in preclinical and therefore translation of these results from bench to bed requires more effort in this field. Exosomes from stem cells are a promising tool for the treatment of ischemic diseases. In addition, translation of pre-clinic results into clinic needs further studies in this field.
Collapse
Affiliation(s)
- Majid Babaei
- Social Determinants of Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, 57147, Urmia, Iran.
| |
Collapse
|
19
|
Ferrantelli F, Chiozzini C, Manfredi F, Giovannelli A, Leone P, Federico M. Simultaneous CD8 + T-Cell Immune Response against SARS-Cov-2 S, M, and N Induced by Endogenously Engineered Extracellular Vesicles in Both Spleen and Lungs. Vaccines (Basel) 2021; 9:240. [PMID: 33801926 PMCID: PMC7999804 DOI: 10.3390/vaccines9030240] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/29/2022] Open
Abstract
Most advanced vaccines against severe acute respiratory syndrome coronavirus (SARS-CoV)-2 are designed to induce antibodies against spike (S) protein. Differently, we developed an original strategy to induce CD8+ T cytotoxic lymphocyte (CTL) immunity based on in vivo engineering of extracellular vesicles (EVs). This is a new vaccination approach based on intramuscular injection of DNA expression vectors coding for a biologically inactive HIV-1 Nef protein (Nefmut) with an unusually high efficiency of incorporation into EVs, even when foreign polypeptides are fused to its C-terminus. Nanovesicles containing Nefmut-fused antigens released by muscle cells can freely circulate into the body and are internalized by antigen-presenting cells. Therefore, EV-associated antigens can be cross-presented to prime antigen-specific CD8+ T-cells. To apply this technology to a strategy of anti-SARS-CoV-2 vaccine, we designed DNA vectors expressing the products of fusion between Nefmut and different viral antigens, namely N- and C-terminal moieties of S (referred to as S1 and S2), M, and N. We provided evidence that all fusion products are efficiently uploaded in EVs. When the respective DNA vectors were injected in mice, a strong antigen-specific CD8+ T cell immunity became detectable in spleens and, most important, in lung airways. Co-injection of DNA vectors expressing the diverse SARS-CoV-2 antigens resulted in additive immune responses in both spleen and lungs. Hence, DNA vectors expressing Nefmut-based fusion proteins can be proposed for new anti-SARS-CoV-2 vaccine strategies.
Collapse
Affiliation(s)
- Flavia Ferrantelli
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Francesco Manfredi
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Andrea Giovannelli
- National Center for Animal Experimentation and Welfare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Patrizia Leone
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| |
Collapse
|
20
|
Yang X, Xie B, Peng H, Shi G, Sreenivas B, Guo J, Wang C, He Y. Eradicating intracellular MRSA via targeted delivery of lysostaphin and vancomycin with mannose-modified exosomes. J Control Release 2020; 329:454-467. [PMID: 33253805 DOI: 10.1016/j.jconrel.2020.11.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 10/17/2020] [Accepted: 11/23/2020] [Indexed: 02/08/2023]
Abstract
Intracellular methicillin-resistant Staphylococcus aureus (MRSA) is extremely difficult to remove by common antibiotics, leading to infection recurrence and resistance. Herein we report a novel exosome-based antibiotic delivery platform for eradicating intracellular MRSA, where mannosylated exosome (MExos) is employed as the drug carrier and preferentially taken up by macrophages, delivering lysostaphin (MExoL) and vancomycin (MExoV) to intracellular pathogens. Combination of MExoL and MExoV eradicated intracellular quiescent MRSA. Moreover, MExos rapidly accumulated in mouse liver and spleen, the target organs of intracellular MRSA, after intravenous (IV) administration. Thus, the MExos antibiotic delivery platform is a promising strategy for combating intracellular infection.
Collapse
Affiliation(s)
- Xiaohong Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Shuitu Technology Development Zone, Beibei, 400714 Chongqing, China; Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Beibei Xie
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Haibo Peng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Gongming Shi
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Banne Sreenivas
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Jian Guo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Chenhui Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China.
| |
Collapse
|
21
|
Lorenc T, Chrzanowski J, Olejarz W. Current Perspectives on Clinical Use of Exosomes as a Personalized Contrast Media and Theranostics. Cancers (Basel) 2020; 12:E3386. [PMID: 33207614 PMCID: PMC7698051 DOI: 10.3390/cancers12113386] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/08/2020] [Accepted: 11/14/2020] [Indexed: 02/07/2023] Open
Abstract
An appropriate combination of biomarkers and imaging technologies will become standard practice in the future. Because the incidence of and mortality from cancers is rising, the further study of new approaches for the early detection and precise characterization of tumors is essential. Extracellular vesicles (EVs), including exosomes, prove to have great potential when it comes to diagnosis and targeted therapy. Due to their natural ability to pass through biological barriers, depending on their origin, EVs can accumulate at defined sites, including tumors, preferentially. This manuscript discusses the difficulties and simplicities of processing cell-derived materials, packaging diverse groups of agents in EVs, and activating the biological complex. Developing exosome-based diagnostic techniques to detect disease precisely and early as well as treat disease marks a new era of personalized radiology and nuclear medicine. As circulating drug delivery vehicles for novel therapeutic modalities, EVs offer a new platform for cancer theranostic.
Collapse
Affiliation(s)
- Tomasz Lorenc
- Ist Department of Clinical Radiology, Medical University of Warsaw, 5 Chalubinskiego Street, 02-004 Warsaw, Poland
| | - Julian Chrzanowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.C.); (W.O.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.C.); (W.O.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
22
|
Foot-and-mouth disease virus degrades Rab27a to suppress the exosome-mediated antiviral immune response. Vet Microbiol 2020; 251:108889. [PMID: 33223235 DOI: 10.1016/j.vetmic.2020.108889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022]
Abstract
Foot-and-mouth disease (FMD) is a highly contagious infection caused by foot-and-mouth disease virus (FMDV). Exosomes are extracellular vesicles that mediate antiviral immune responses in host cells and could be used by pathogens to evade host cell immune responses. Whether FMDV affects exosome secretion or whether exosomes derived from FMDV-infected cells mediate host cell antiviral immune responses is not yet clarified. In this study, the exosomes were identified and extracted from FMDV-infected PK-15 cells, and it was found that FMDV inhibits exosome secretion. Further investigation revealed that FMDV suppresses exosomes by degrading Rab27a via the autophagy-lysosome pathway. Also, microRNA (miRNA) differential analysis was performed in exosomes, which revealed that miRNA-136 was highly differentially expressed in exosomes and may be the key miRNA that inhibits the proliferation of FMDV. In summary, these results showed that host cells take advantage of exosomes to mediate their antiviral immune response, while FMDV evades exosome-mediated immune responses by degrading the exosome molecular switch, Rab27a.
Collapse
|
23
|
Torres-Berrío A, Nouel D, Cuesta S, Parise EM, Restrepo-Lozano JM, Larochelle P, Nestler EJ, Flores C. MiR-218: a molecular switch and potential biomarker of susceptibility to stress. Mol Psychiatry 2020; 25:951-964. [PMID: 30980043 PMCID: PMC6790160 DOI: 10.1038/s41380-019-0421-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 03/27/2019] [Accepted: 04/03/2019] [Indexed: 01/22/2023]
Abstract
Low miR-218 expression in the medial prefrontal cortex (mPFC) is a consistent trait of depression. Here we assessed whether miR-218 in the mPFC confers resilience or susceptibility to depression-like behaviors in adult mice, using the chronic social defeat stress (CSDS) model of depression. We also investigated whether stress-induced variations of miR-218 expression in the mPFC can be detected in blood. We find that downregulation of miR-218 in the mPFC increases susceptibility to a single session of social defeat, whereas overexpression of miR-218 selectively in mPFC pyramidal neurons promotes resilience to CSDS and prevents stress-induced morphological alterations to those neurons. After CSDS, susceptible mice have low levels of miR-218 in blood, as compared with control or resilient groups. We show further that upregulation and downregulation of miR-218 levels specifically in the mPFC correlate with miR-218 expression in blood. Our results suggest that miR-218 in the adult mPFC might function as a molecular switch that determines susceptibility vs. resilience to chronic stress, and that stress-induced variations in mPFC levels of miR-218 could be detected in blood. We propose that blood expression of miR-218 might serve as potential readout of vulnerability to stress and as a proxy of mPFC function.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dominique Nouel
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Santiago Cuesta
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
| | - Eric M Parise
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - José María Restrepo-Lozano
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Pier Larochelle
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, Québec, Canada.
- Department of Psychiatry, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
24
|
Maione F, Cappellano G, Bellan M, Raineri D, Chiocchetti A. Chicken-or-egg question: Which came first, extracellular vesicles or autoimmune diseases? J Leukoc Biol 2020; 108:601-616. [PMID: 32108378 PMCID: PMC7496139 DOI: 10.1002/jlb.3mr0120-232r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/21/2020] [Accepted: 02/03/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) have attracted great interest as contributors to autoimmune disease (AD) pathogenesis, owing to their immunomodulatory potential; they may also play a role in triggering tolerance disruption, by delivering auto‐antigens. EVs are released by almost all cell types, and afford paracrine or distal cell communication, functioning as biological carriers of active molecules including lipids, proteins, and nucleic acids. Depending on stimuli from the external microenvironment or on their cargo, EVs can promote or suppress immune responses. ADs are triggered by inappropriate immune‐system activation against the self, but their precise etiology is still poorly understood. Accumulating evidence indicates that lifestyle and diet have a strong impact on their clinical onset and development. However, to date the mechanisms underlying AD pathogenesis are not fully clarified, and reliable markers, which would provide early prediction and disease progression monitoring, are lacking. In this connection, EVs have recently been indicated as a promising source of AD biomarkers. Although EV isolation is currently based on differential centrifugation or density‐gradient ultracentrifugation, the resulting co‐isolation of contaminants (i.e., protein aggregates), and the pooling of all EVs in one sample, limit this approach to abundantly‐expressed EVs. Flow cytometry is one of the most promising methods for detecting EVs as biomarkers, and may have diagnostic applications. Furthermore, very recent findings describe a new method for identifying and sorting EVs by flow cytometry from freshly collected body fluids, based on specific EV surface markers.
Collapse
Affiliation(s)
- Federica Maione
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy.,Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases- IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Giuseppe Cappellano
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy.,Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases- IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Bellan
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy.,Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Davide Raineri
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy.,Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases- IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Annalisa Chiocchetti
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy.,Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases- IRCAD, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
25
|
Lara P, Palma-Florez S, Salas-Huenuleo E, Polakovicova I, Guerrero S, Lobos-Gonzalez L, Campos A, Muñoz L, Jorquera-Cordero C, Varas-Godoy M, Cancino J, Arias E, Villegas J, Cruz LJ, Albericio F, Araya E, Corvalan AH, Quest AFG, Kogan MJ. Gold nanoparticle based double-labeling of melanoma extracellular vesicles to determine the specificity of uptake by cells and preferential accumulation in small metastatic lung tumors. J Nanobiotechnology 2020; 18:20. [PMID: 31973696 PMCID: PMC6979068 DOI: 10.1186/s12951-020-0573-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Extracellular vesicles (EVs) have shown great potential for targeted therapy, as they have a natural ability to pass through biological barriers and, depending on their origin, can preferentially accumulate at defined sites, including tumors. Analyzing the potential of EVs to target specific cells remains challenging, considering the unspecific binding of lipophilic tracers to other proteins, the limitations of fluorescence for deep tissue imaging and the effect of external labeling strategies on their natural tropism. In this work, we determined the cell-type specific tropism of B16F10-EVs towards cancer cell and metastatic tumors by using fluorescence analysis and quantitative gold labeling measurements. Surface functionalization of plasmonic gold nanoparticles was used to promote indirect labeling of EVs without affecting size distribution, polydispersity, surface charge, protein markers, cell uptake or in vivo biodistribution. Double-labeled EVs with gold and fluorescent dyes were injected into animals developing metastatic lung nodules and analyzed by fluorescence/computer tomography imaging, quantitative neutron activation analysis and gold-enhanced optical microscopy. Results We determined that B16F10 cells preferentially take up their own EVs, when compared with colon adenocarcinoma, macrophage and kidney cell-derived EVs. In addition, we were able to detect the preferential accumulation of B16F10 EVs in small metastatic tumors located in lungs when compared with the rest of the organs, as well as their precise distribution between tumor vessels, alveolus and tumor nodules by histological analysis. Finally, we observed that tumor EVs can be used as effective vectors to increase gold nanoparticle delivery towards metastatic nodules. Conclusions Our findings provide a valuable tool to study the distribution and interaction of EVs in mice and a novel strategy to improve the targeting of gold nanoparticles to cancer cells and metastatic nodules by using the natural properties of malignant EVs.
Collapse
Affiliation(s)
- Pablo Lara
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile.,Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Sujey Palma-Florez
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Edison Salas-Huenuleo
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Iva Polakovicova
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Laboratory of Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago, Chile
| | - Simón Guerrero
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Instituto de investigación Interdisciplinar en Ciencias biomédicas, Universidad SEK (I3CBSEK). Facultad Ciencias de La Salud, Fernando Manterola 0789, Santiago, Chile
| | - Lorena Lobos-Gonzalez
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Centro de Medicina Regenerativa, Facultad de Medicina-Clinica Alemana, Universidad Del Desarrollo, Avenida las condes 12438, lo Barnechea, Santiago, Chile
| | - America Campos
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Luis Muñoz
- Laboratorio de Análisis Por Activación Neutrónica, Comisión Chilena de Energía Nuclear, Nueva Bilbao, 12501, Santiago, Chile
| | - Carla Jorquera-Cordero
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile
| | - Manuel Varas-Godoy
- Centro de Biología Celular Y Biomedicina (CEBICEM), Facultad de Medicina Y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
| | - Jorge Cancino
- Centro de Biología Celular Y Biomedicina (CEBICEM), Facultad de Medicina Y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
| | - Eloísa Arias
- Centro de Biología Celular Y Biomedicina (CEBICEM), Facultad de Medicina Y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
| | - Jaime Villegas
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Republica 440, Santiago, Chile
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands
| | - Fernando Albericio
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, and Department of Organic Chemistry, University of Barcelona, 08028, Barcelona, Spain
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Universidad Andres Bello, Republica 275, 8370146, Santiago, Chile
| | - Alejandro H Corvalan
- Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.,Laboratory of Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago, Chile
| | - Andrew F G Quest
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), University of Chile, Av. Independencia 1027, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.
| | - Marcelo J Kogan
- Departamento de Química Farmacológica Y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Sergio Livingstone 1007, Santiago, Chile.
| |
Collapse
|
26
|
Cooper LF, Ravindran S, Huang CC, Kang M. A Role for Exosomes in Craniofacial Tissue Engineering and Regeneration. Front Physiol 2020; 10:1569. [PMID: 32009978 PMCID: PMC6971208 DOI: 10.3389/fphys.2019.01569] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022] Open
Abstract
Tissue engineering and regenerative medicine utilize mesenchymal stem cells (MSCs) and their secretome in efforts to create or induce functional tissue replacement. Exosomes are specific extracellular vesicles (EVs) secreted by MSCs and other cells that carry informative cargo from the MSC to targeted cells that influence fundamental cellular processes including apoptosis, proliferation, migration, and lineage-specific differentiation. In this report, we review the current knowledge regarding MSC exosome biogenesis, cargo and function. This review summarizes the use of MSC exosomes to control or induce bone, cartilage, dentin, mucosa, and pulp tissue formation. The next-step engineering of exosomes provides additional avenues to enhance oral and craniofacial tissue engineering and regeneration.
Collapse
Affiliation(s)
- Lyndon F. Cooper
- College of Dentistry, The University of Illinois at Chicago, Chicago, IL, United States
| | | | | | | |
Collapse
|
27
|
Huang Y, Deng X, Liang J. Review of the Application of Nanovesicles and the Human Interstitial Fluid in Gastrointestinal Premalignant Lesion Detection, Diagnosis, Prognosis and Therapy. Int J Nanomedicine 2019; 14:9469-9482. [PMID: 31819444 PMCID: PMC6896916 DOI: 10.2147/ijn.s208559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022] Open
Abstract
Premalignant lesions arise from cells that abnormally proliferate and have a tendency to become cancerous. Developing methods to specifically target and remove these premalignant lesions is imperative to the prevention of malignant progression into gastrointestinal (GI) tumors. However, accurate detection and diagnosis of GI precancerous lesions is challenging, as these lesions show little or no structural change. Thus, this prevents early intervention and reduces the success rate of therapy. In this review, we performed a systematic analysis of the technological advancements in the combined application of nanovesicles (NVs) and the human interstitial fluid (HIF) to specifically target GI premalignant lesions. NVs, which include quantum dots (QDs), are small membranous vehicles of a nanometer diameter that are widely used as drug delivery vectors, therapeutic effectors and diagnostic sensors. HIF is the fluid that is present in human interstitial tissues (HITs) in which signaling molecules and agents travel and can be found throughout the body. HIF is exploited by tumor cells for their invasion, migration and spread. Because the HITs span the entire submucosa of the gastrointestinal tract, they have been increasingly targeted in GI tumor therapy. The challenges involved in the combined application of NVs and HIF in the detection, diagnosis, prognosis and therapy of GI premalignant lesions are also discussed.
Collapse
Affiliation(s)
- Yu Huang
- Liuzhou Traditional Chinese Medical Hospital, Liuzhou 545001, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xin Deng
- Guangxi University of Chinese Medicine, Nanning 530001, Guangxi Zhuang Autonomous Region, People's Republic of China.,Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Jian Liang
- Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
28
|
Chen SL, Chen CY, Hsieh JCH, Yu ZY, Cheng SJ, Hsieh KY, Yang JW, Kumar PV, Lin SF, Chen GY. Graphene Oxide-Based Biosensors for Liquid Biopsies in Cancer Diagnosis. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1725. [PMID: 31816919 PMCID: PMC6956293 DOI: 10.3390/nano9121725] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
Liquid biopsies use blood or urine as test samples, which are able to be continuously collected in a non-invasive manner. The analysis of cancer-related biomarkers such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), microRNA, and exosomes provides important information in early cancer diagnosis, tumor metastasis detection, and postoperative recurrence monitoring assist with clinical diagnosis. However, low concentrations of some tumor markers, such as CTCs, ctDNA, and microRNA, in the blood limit its applications in clinical detection and analysis. Nanomaterials based on graphene oxide have good physicochemical properties and are now widely used in biomedical detection technologies. These materials have properties including good hydrophilicity, mechanical flexibility, electrical conductivity, biocompatibility, and optical performance. Moreover, utilizing graphene oxide as a biosensor interface has effectively improved the sensitivity and specificity of biosensors for cancer detection. In this review, we discuss various cancer detection technologies regarding graphene oxide and discuss the prospects and challenges of this technology.
Collapse
Affiliation(s)
- Shiue-Luen Chen
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Chong-You Chen
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Jason Chia-Hsun Hsieh
- Division of Haematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital (Linkou), Taoyuan 333, Taiwan;
| | - Zih-Yu Yu
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
| | - Sheng-Jen Cheng
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Kuan Yu Hsieh
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Jia-Wei Yang
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Priyank V Kumar
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Shien-Fong Lin
- Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan; (S.-L.C.); (C.-Y.C.); (Z.-Y.Y.); (S.-J.C.); (K.Y.H.); (J.-W.Y.); (S.-F.L.)
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Guan-Yu Chen
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
29
|
Lee DS, Suh MI, Kang SY, Hwang DW. Physiologic constraints of using exosomes in vivo as systemic delivery vehicles. PRECISION NANOMEDICINE 2019. [DOI: 10.33218/prnano2(3)070819.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Systemic delivery of exosomes meets hurdles which had not been elucidated using live molecular imaging for their biodistribution. Production and uptake of endogenous exosomes are expected to be nonspecific and specific, respectively, where external stimuli of production of exosomes and their quantitative degree of productions are not understood. Despite this lack of understanding of basic physiology of in vivo behavior of exosomes including their possible paracrine or endocrine actions, many engineering efforts are taken to develop therapeutic vehicles. Especially, the fraction of exosomes’ taking the routes of waste disposal and exerting target actions are not characterized after systemic administration. Here, we reviewed the literature about in vivo distribution and disposal/excretion of exogenous or endogenous exosomes and, from these limited resources of knowledge currently available, summarized the knowledge and the uncertainties of exosomes on physiologic standpoints. An eloquent example of the investigations to understand the roles and confounders of exosomes’ action in the brain was highlighted with emphasis on the recent discovery of brain lymphatics and hypothesis of glymphatic/lymphatic clearance pathways in diseases as well as in physiologic processes. The possibility of delivering therapeutic exosomes through the systemic circulation, across blood-brain barriers and finally to target cells such as microglia, astrocytes and/or neurons is a good testbed in which the investigators can formulate problems to solve for both understanding (science) and application (engineering).
Collapse
Affiliation(s)
- Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - MInseok Suh
- 2Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University,
| | - Seo Young Kang
- Department of Nuclear Medicine, Ewha Womans University Medical Center, Seoul,
| | - Do Won Hwang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
30
|
Meldolesi J. Extracellular vesicles, news about their role in immune cells: physiology, pathology and diseases. Clin Exp Immunol 2019; 196:318-327. [PMID: 30756386 DOI: 10.1111/cei.13274] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
Two types of extracellular vesicles (EVs), exosomes and ectosomes, are generated and released by all cells, including immune cells. The two EVs appear different in many properties: size, mechanism and site of assembly, composition of their membranes and luminal cargoes, sites and processes of release. In functional terms, however, these differences are minor. Moreover, their binding to and effects on target cells appear similar, thus the two types are considered distinct only in a few cases, otherwise they are presented together as EVs. The EV physiology of the various immune cells differs as expected from their differential properties. Some properties, however, are common: EV release, taking place already at rest, is greatly increased upon cell stimulation; extracellular navigation occurs adjacent and at distance from the releasing cells; binding to and uptake by target cells are specific. EVs received from other immune or distinct cells govern many functions in target cells. Immune diseases in which EVs play multiple, often opposite (aggression and protection) effects, are numerous; inflammatory diseases; pathologies of various tissues; and brain diseases, such as multiple sclerosis. EVs also have effects on interactive immune and cancer cells. These effects are often distinct, promoting cytotoxicity or proliferation, the latter together with metastasis and angiogenesis. Diagnoses depend on the identification of EV biomarkers; therapies on various mechanisms such as (1) removal of aggression-inducing EVs; (2) EV manipulations specific for single targets, with insertion of surface peptides or luminal miRNAs; and (3) removal or re-expression of molecules from target cells.
Collapse
Affiliation(s)
- J Meldolesi
- Division of Neuroscience, Unit of Molecular and Cellular Neuroscience, San Raffaele Scientific Institute and San Raffaele University, Milan, Italy
| |
Collapse
|
31
|
Shanmuganathan M, Vughs J, Noseda M, Emanueli C. Exosomes: Basic Biology and Technological Advancements Suggesting Their Potential as Ischemic Heart Disease Therapeutics. Front Physiol 2018; 9:1159. [PMID: 30524292 PMCID: PMC6262308 DOI: 10.3389/fphys.2018.01159] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
Exosomes are small nano-sized vesicles that deliver biologically active RNA molecules and proteins to recipient cells through binding, fusion or endocytosis. There is emerging evidence that endogenous exosomes released by cardiovascular cells and progenitor cells impact cell survival and proliferation, thus regulating angiogenesis, cardiac protection and repair. These cardioprotective and regenerative traits have the potential to translate in to novel therapeutic options for post-ischaemic cardiac regeneration, thus potentially delaying the progression to ischaemic heart failure. Cellular stressors influence exosomes' secretion and the molecular composition of the exosome cargo, thus impacting on the above processes. Evidences are emerging that loading of proteins and RNAs in the exosomes cargos can be manipulated. Similarly, manipulation of exosomes surface proteins' expression to target exosomes to specific cells and tissues is doable. In addition, nature-inspired synthetic exosomes can be assembled to deliver specific clues to the recipient cells, including proliferative and differentiation stimuli, or shed paracrine signals enabling to reconstructing the heart homeostatic micro-environment. This review will describe exosome biogenesis and emerging evidence of exosome-mediated regenerative cell-to-cell communications and will conclude discussing possibilities of using exosomes to treat ischemic heart disease.
Collapse
Affiliation(s)
- Mayooran Shanmuganathan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | - Jeff Vughs
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|