Skip to main content

EDITORIAL article

Front. Bioeng. Biotechnol., 25 July 2023
Sec. Nanobiotechnology
This article is part of the Research Topic Engineered Extracellular Vesicles for Tissue Repairing View all 6 articles

Editorial: Engineered extracellular vesicles for tissue repairing

Shiyi Yu,&#x;Shiyi Yu1,2Guoyong Chen&#x;Guoyong Chen3Siim PauklinSiim Pauklin4Yunjiao ZhangYunjiao Zhang5Yuliang FengYuliang Feng4Hao Chen,
Hao Chen1,2*
  • 1Department of Gastroenterology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
  • 2The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
  • 3Department of Stomatology, Zhuzhou Central Hospital, Xiangya School of Medicine, Central South University, Zhuzhou, China
  • 4Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
  • 5School of Medicine and Institute for Life Sciences, South China University of Technology, Guangzhou, China

Extracellular vesicles (EVs) are a type of nanoscale vesicles encapsulated by bilayer lipid membrane, which are mainly divided into several categories based on varied diameter and biological pathway: exosomes (Exos), microvesicles (MVs), and apoptotic bodies (ApoBDs) (EL Andaloussi et al., 2013). Almost all types of cells can secrete extracellular vesicles, which carry a variety of macromolecules such as nucleic acids (DNA, mRNA, microRNA, lncRNA), lipids, proteins, and metabolites, mediating intercellular communication. Native EVs can play a physiological role in promoting cell proliferation and regulating inflammation. However, due to their small production, low concentration, and high levels of impurities, native EVs have lower potential in therapeutic effects compared with engineered EVs. In this Research Topic, multiple studies have demonstrated the role and mechanism of engineered extracellular vesicles in promoting tissue healing and inflammatory regulation, and introduced the cutting-edge methods for preparing engineered extracellular vesicles.

In terms of the mechanism of tissue repairing by EVs, Lyu et al. demonstrated that M2-EXO enhanced the angiogenic ability of human umbilical vein endothelial cells (HUVECs) in vitro by transferring miR-21-5p, which inhibited PTEN expression and activated the AKT/mTOR pathway. Extensive research has shown that macrophages are closely related to angiogenesis at the wound site (Hesketh et al., 2017). Previous studies have mostly focused on the effect of M2 macrophages on wound healing, with few articles analyzing the role of M2-EXO, an extracellular vesicle secreted by M2 macrophages. In this study, the researchers used a full thickness cutaneous wound mice model to demonstrate that M2-EXO treatment promotes angiogenesis and accelerates skin healing. These findings highlight the promising therapeutic potential of M2-EXO as a cell-free method for enhancing skin healing and angiogenesis, which shows implications to translating into clinical use.

The study conducted by Wang et al. provides valuable insights into the potential of using extracellular vesicles derived from 4D culture (4D-sEVs) to reduce inflammatory response after spinal cord injury (SCI). Researchers obtained 4D-sEVs from human umbilical cord mesenchymal stem cells (hUC-MSCs) using 3D culture technology based on porous scaffolds. Proteomics analysis revealed significant changes in protein mass spectrometry, including the upregulation of Epidermal growth factor receptor (EGFR) and Insulin-like growth factor-binding protein 2 (IGFBP2) in 4D-sEV. In vitro and in SCI rats, 4D-sEV demonstrated the effective induction of macrophages/Microglia from pro-inflammatory M1 to anti-inflammatory M2 phenotype polarization. In addition, delivering 4D-sEV to the injured site leads to reduced neuroinflammation and increased tissue repair. This new method is expected to suppress the inflammatory response and promote recovery in SCI patients.

The structure and characteristics of EVs’ carrier can affect its effectiveness in tissue repairing. Xia et al. developed a photocrosslinked hydrogel capable of loading exosomes, which can promote hepatocyte regeneration after Hepatectomy. At present, the commonly used hemostasis methods of Hepatectomy include suture hemostasis, electrocoagulation, ultrasonic scalpel, etc. As a new postoperative hemostasis treatment, the light crosslinked adhesive hydrogel loaded with extracellular vesicles can promote hemostasis, accelerate cell proliferation and migration, and promote wound repair and tissue regeneration. The hydrogel is composed of modified gelatin matrix (GelMA) and alginate dopamine (Alg-DA). In order to further enhance its application in liver regeneration, adipose derived mesenchymal stem cell exosomes (AD-MSC-Exo) were loaded into GelMA/Alg-DA-1 hydrogel to improve cell proliferation and migration.

Hao et al. summarized the application and therapeutic mechanism of engineered stem cell extracellular vesicles in oral and maxillofacial wound healing. Through different methods such as genetic engineering, co incubation, parental cell surface modification, and artificial synthesis, stem cell extracellular vesicles with higher purity, targeting, and drug delivery efficiency can be engineered and prepared, achieving more significant therapeutic effects. Engineering stem cell exosomes can promote the healing of oral and maxillofacial wounds by regulating inflammatory reaction, promoting fibroblast proliferation, angiogenesis, and reducing the formation of scars, thus have a promising prospect for their application in Regenerative medicine.

Wei et al. reviewed the application of engineered extracellular vesicles in the treatment of degenerative orthopedic diseases. EV carries a variety of biologically active molecules, which can play an anti-inflammatory role, reduce chondrocyte apoptosis, and promote tissue repair (Silverman, 1986; Joseph et al., 2016; Lee et al., 2021; Mustonen and Nieminen, 2021). It has made certain research progress in the treatment of various DOD diseases such as OA, OP, and IDD. The engineering EVs typically have better performance and thermal potential than native EV. Currently, the main engineering methods include active loading and passive loading.

As a cell-free therapy, engineered extracellular vesicles have higher safety, lower immunogenicity and higher targeting, compared with stem cell therapy. In addition to the clinical applications mentioned above, EVs have also found application value in cardiac repair, fatty liver treatment and other fields (Wang et al., 2021; Pezzana et al., 2021). How to obtain therapeutic EVs with a well targeting property and good efficacy is the biggest challenge to promote its clinical application. EVs’ excellent ability in cell-growth promoting, anti-inflammatory and immune regulatory effects will widen its application in an increasing number of clinical fields.

Author contributions

SY and GC wrote the manuscript together. SP, YZ, YF, and HC were involved in conceptualization and supervision. HC carefully reviewed the manuscript. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

El Andaloussi, S., Mäger, I., Breakefield, X., and Wood, M. J. A. (2013). Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347–357. doi:10.1038/nrd3978

PubMed Abstract | CrossRef Full Text | Google Scholar

Hesketh, M., Sahin, K. B., West, Z. E., and Murray, R. Z. (2017). Macrophage phenotypes regulate scar formation and chronic wound healing. Int. J. Mol. Sci. 18 (7), E1545. doi:10.3390/ijms18071545

CrossRef Full Text | Google Scholar

Joseph, W., Cameron, M., Yutao, L., Hunter, M., Fulzele, S., and Hamrick, M. W. (2016). Extracellular vesicles in the pathogenesis of rheumatoid arthritis and osteoarthritis. Arthritis Res. Ther. 18, 286. doi:10.1186/s13075-016-1178-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, K. S., Lee, J., Kim, H. K., Yeom, S. H., Woo, C. H., Jung, Y. J., et al. (2021). Extracellular vesicles from adipose tissue-derived stem cells alleviate osteoporosis through osteoprotegerin and miR-21-5p. J. Extracell. Vesicles 10 (12), e12152. doi:10.1002/jev2.12152

PubMed Abstract | CrossRef Full Text | Google Scholar

Mustonen, A. M., and Nieminen, P. (2021). Extracellular vesicles and their potential significance in the pathogenesis and treatment of osteoarthritis. Pharm. (Basel) 14 (4), 315. doi:10.3390/ph14040315

CrossRef Full Text | Google Scholar

Pezzana, C., Agnely, F., Bochot, A., Siepmann, J., and Menasché, P. (2021). Extracellular vesicles and biomaterial design: New therapies for cardiac repair. Trends Mol. Med. 27 (3), 231–247. doi:10.1016/j.molmed.2020.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Silverman, S. (1986). Infectious and sexually transmitted diseases: Implications for dental public health. J. Public Health Dent. 46 (1), 7–12. doi:10.1111/j.1752-7325.1986.tb03086.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., Mehal, W., Nagy, L. E., and Rotman, Y. (2021). Immunological mechanisms and therapeutic targets of fatty liver diseases. Cell Mol. Immunol. 18, 73–91. doi:10.1038/s41423-020-00579-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: extracellular vesicle, bioengineering, biotechnology, tissue repairing, diagnosis, targeted therapy

Citation: Yu S, Chen G, Pauklin S, Zhang Y, Feng Y and Chen H (2023) Editorial: Engineered extracellular vesicles for tissue repairing. Front. Bioeng. Biotechnol. 11:1257165. doi: 10.3389/fbioe.2023.1257165

Received: 12 July 2023; Accepted: 14 July 2023;
Published: 25 July 2023.

Edited and reviewed by:

Gianni Ciofani, Italian Institute of Technology (IIT), Italy

Copyright © 2023 Yu, Chen, Pauklin, Zhang, Feng and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Hao Chen, chenhao@gdph.org.cn

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.