Mechanism and Optimization Strategy of Mesenchymal Stem Cell Exosomes against Liver Fibrosis

Xiaolei ZHOU, Yan XU, Yu JIN, Junsong YE

China Biotechnology ›› 2024, Vol. 44 ›› Issue (6) : 41-52.

PDF(862 KB)
PDF(862 KB)
China Biotechnology ›› 2024, Vol. 44 ›› Issue (6) : 41-52. DOI: 10.13523/j.cb.2312051

Mechanism and Optimization Strategy of Mesenchymal Stem Cell Exosomes against Liver Fibrosis

Author information +
History +

Abstract

Liver fibrosis is a major problem in the treatment of liver disease. Mesenchymal stem cells can be used to treat liver fibrosis, but are limited by their potential carcinogenicity, and the large number of cells required for transplantation. In recent years, mesenchymal stem cell exosomes have become a research hotspot due to their smaller size, lower immunogenicity and non-carcinogenicity. However, the clinical application of mesenchymal stem cell exosomes is limited for the following reasons. First, there is a lack of standard and uniform methods for extracting and identifying exosomes from different types of mesenchymal stem cells. Second, the mechanism of mesenchymal stem cell exosomes to treat liver fibrosis is not clear. Finally, mesenchymal stem cell exosome therapy has some problems such as weak ability to target aHSC, low exosome production, low drug loading ability and low delivery efficiency. In view of the above reasons, the factors causing the difference in exosome extraction effect of common mesenchymal stem cells, the mechanism of mesenchymal stem cell exosomes ameliorating liver fibrosis and its optimization strategy were reviewed, providing new understanding and new ideas for the treatment of liver fibrosis by mesenchymal stem cell exosomes.

Key words

Mesenchymal stem cell exosomes / Hepatic fibrosis / Mechanism / Optimization strategy

Cite this article

Download Citations
Xiaolei ZHOU, Yan XU, Yu JIN, et al. Mechanism and Optimization Strategy of Mesenchymal Stem Cell Exosomes against Liver Fibrosis[J]. China Biotechnology, 2024, 44(6): 41-52 https://doi.org/10.13523/j.cb.2312051

References

[1]
Saeed A, Bartuzi P, Heegsma J, et al. Impaired hepatic vitamin A metabolism in NAFLD mice leading to vitamin A accumulation in hepatocytes. Cellular and Molecular Gastroenterology and Hepatology, 2021, 11(1): 309-325.e3.
Systemic retinol (vitamin A) homeostasis is controlled by the liver, involving close collaboration between hepatocytes and hepatic stellate cells (HSCs). Genetic variants in retinol metabolism (PNPLA3 and HSD17B13) are associated with non-alcoholic fatty liver disease (NAFLD) and disease progression. Still, little mechanistic details are known about hepatic vitamin A metabolism in NAFLD, which may affect carbohydrate and lipid metabolism, inflammation, oxidative stress and the development of fibrosis and cancer, e.g. all risk factors of NAFLD.Here, we analyzed vitamin A metabolism in 2 mouse models of NAFLD; mice fed a high-fat, high-cholesterol (HFC) diet and Leptin mutant (ob/ob) mice.Hepatic retinol and retinol binding protein 4 (RBP4) levels were significantly reduced in both mouse models of NAFLD. In contrast, hepatic retinyl palmitate levels (the vitamin A storage form) were significantly elevated in these mice. Transcriptome analysis revealed a hyperdynamic state of hepatic vitamin A metabolism, with enhanced retinol storage and metabolism (upregulated Lrat, Dgat1, Pnpla3, Raldh's and RAR/RXR-target genes) in fatty livers, in conjunction with induced hepatic inflammation (upregulated Cd68, Tnfα, Nos2, Il1β, Il-6) and fibrosis (upregulated Col1a1, Acta2, Tgfβ, Timp1). Autofluorescence analyses revealed prominent vitamin A accumulation in hepatocytes rather than HSC in HFC-fed mice. Palmitic acid exposure increased Lrat mRNA levels in primary rat hepatocytes and promoted retinyl palmitate accumulation when co-treated with retinol, which was not detected for similarly-treated primary rat HSCs.NAFLD leads to cell type-specific rearrangements in retinol metabolism leading to vitamin A accumulation in hepatocytes. This may promote disease progression and/or affect therapeutic approaches targeting nuclear receptors.Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.
[2]
Lee C B, Kim M, Han J, et al. Mesenchymal stem cells influence activation of hepatic stellate cells, and constitute a promising therapy for liver fibrosis. Biomedicines, 2021, 9(11): 1598.
[3]
Guo Y, Chen B, Chen L J, et al. Current status and future prospects of mesenchymal stem cell therapy for liver fibrosis. Journal of Zhejiang University: Science B, 2016, 17(11): 831-841.
[4]
Lou G H, Chen Z, Zheng M, et al. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Experimental & Molecular Medicine, 2017, 49(6): e346.
[5]
Evers M J W, van de Wakker S I, de Groot E M, et al. Functional siRNA delivery by extracellular vesicle-liposome hybrid nanoparticles. Advanced Healthcare Materials, 2022, 11(5): e2101202.
[6]
Doyle L M, Wang M Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells, 2019, 8(7): 727.
[7]
Cheng L L, Zhang X G, Tang J J, et al. Gene-engineered exosomes-thermosensitive liposomes hybrid nanovesicles by the blockade of CD 47 signal for combined photothermal therapy and cancer immunotherapy. Biomaterials, 2021, 275: 120964.
[8]
Sun L N, Fan M R, Huang D, et al. Clodronate-loaded liposomal and fibroblast-derived exosomal hybrid system for enhanced drug delivery to pulmonary fibrosis. Biomaterials, 2021, 271: 120761.
[9]
Piffoux M, Silva A K A, Wilhelm C, et al. Modification of extracellular vesicles by fusion with liposomes for the design of personalized biogenic drug delivery systems. ACS Nano, 2018, 12(7): 6830-6842.
Extracellular vesicles (EVs) are recognized as nature's own carriers to transport macromolecules throughout the body. Hijacking this endogenous communication system represents an attractive strategy for advanced drug delivery. However, efficient and reproducible loading of EVs with therapeutic or imaging agents still represents a bottleneck for their use as a drug delivery system. Here, we developed a method for modifying cell-derived EVs through their fusion with liposomes containing both membrane and soluble cargoes. The fusion of EVs with functionalized liposomes was triggered by polyethylene glycol (PEG) to create smart biosynthetic hybrid vectors. This versatile method proved to be efficient to enrich EVs with exogenous lipophilic or hydrophilic compounds, while preserving their intrinsic content and biological properties. Hybrid EVs improved cellular delivery efficiency of a chemotherapeutic compound by a factor of 3-4, as compared to the free drug or the drug-loaded liposome precursor. On one side, this method allows the biocamouflage of liposomes by enriching their lipid bilayer and inner compartment with biogenic molecules. On the other side, the proposed fusion strategy enables efficient EV loading, and the pharmaceutical development of EVs with adaptable activity and drug delivery property.
[10]
Yu L L, Zhu J, Liu J X, et al. A comparison of traditional and novel methods for the separation of exosomes from human samples. BioMed Research International, 2018, 2018: 3634563.
[11]
Zhao A G, Shah K, Cromer B, et al. Comparative analysis of extracellular vesicles isolated from human mesenchymal stem cells by different isolation methods and visualisation of their uptake. Experimental Cell Research, 2022, 414(2): 113097.
[12]
Stam J, Bartel S, Bischoff R, et al. Isolation of extracellular vesicles with combined enrichment methods. Journal of Chromatography B, Analytical Technologies in the Biomedical and Life Sciences, 2021, 1169: 122604.
[13]
Kurian T K, Banik S, Gopal D, et al. Elucidating methods for isolation and quantification of exosomes: a review. Molecular Biotechnology, 2021, 63(4): 249-266.
Exosomes are the smallest extracellular vesicles present in most of the biological fluids. They are found to play an important role in cell signaling, immune response, tumor metastasis, etc. Studies have shown that these vesicles also have diagnostic and therapeutic roles for which their accurate detection and quantification is essential. Due to the complexity in size and structure of exosomes, even the gold standard methods face challenges. This comprehensive review discusses the various standard methods such as ultracentrifugation, ultrafiltration, size-exclusion chromatography, precipitation, immunoaffinity, and microfluidic technologies for the isolation of exosomes. The principle of isolation of each method is described, as well as their specific advantages and disadvantages. Quantification of exosomes by nanoparticle tracking analysis, flow cytometry, tunable resistive pulse sensing, electron microscopy, dynamic light scattering, and microfluidic devices are also described, along with the applications of exosomes in various biomedical domains.
[14]
Colao I L, Corteling R, Bracewell D, et al. Manufacturing exosomes: a promising therapeutic platform. Trends in Molecular Medicine, 2018, 24(3): 242-256.
Extracellular vesicles, in particular the subclass exosomes, are rapidly emerging as a novel therapeutic platform. However, currently very few clinical validation studies and no clearly defined manufacturing process exist. As exosomes progress towards the clinic for treatment of a vast array of diseases, it is important to define the engineering basis for their manufacture early in the development cycle to ensure they can be produced cost-effectively at the appropriate scale. We hypothesize that transitioning to defined manufacturing platforms will increase consistency of the exosome product and improve their clinical advancement as a new therapeutic tool. We present manufacturing technologies and strategies that are being implemented and consider their application for the transition from bench-scale to clinical production of exosomes.Copyright © 2018 Elsevier Ltd. All rights reserved.
[15]
Sun J W, Chen Z, Tian K W, et al. Magnetic bead-based adsorption strategy for exosome isolation. Frontiers in Bioengineering and Biotechnology, 2022, 10: 942077.
[16]
Patil S M, Sawant S S, Kunda N K. Exosomes as drug delivery systems: a brief overview and progress update. European Journal of Pharmaceutics and Biopharmaceutics, 2020, 154: 259-269.
Exosomes are intracellular membrane-based vesicles with diverse compositions that are involved in biological and pathological processes. Since the discovery of exosomes, they have been used as diagnostic biomarkers and as potential drug delivery vehicles based on their size and competence to transfer biological materials to recipient cells. The properties of exosomes such as biocompatibility, preferred tumor homing, adjustable targeting efficiency, and stability make them striking and excellent drug delivery vehicles for use in various diseases and cancer therapy. In this article, we provide a brief overview of the biogenesis, functions, and contents of exosomes along with the separation and characterization techniques. Our major focus is on the recent progress made in application of exosomes as drug delivery systems involving delivery of small molecules, macromolecules, and nucleotides. Further, we discuss the challenges faced when using exosomes as a drug delivery vehicle.Copyright © 2020 Elsevier B.V. All rights reserved.
[17]
Tiwari S, Kumar V, Randhawa S, et al. Preparation and characterization of extracellular vesicles. American Journal of Reproductive Immunology, 2021, 85(2): e13367.
[18]
Abhange K, Makler A, Wen Y, et al. Small extracellular vesicles in cancer. Bioactive Materials, 2021, 6(11): 3705-3743.
Extracellular vesicles (EV) are lipid-bilayer enclosed vesicles in submicron size that are released from cells. A variety of molecules, including proteins, DNA fragments, RNAs, lipids, and metabolites can be selectively encapsulated into EVs and delivered to nearby and distant recipient cells. In tumors, through such intercellular communication, EVs can regulate initiation, growth, metastasis and invasion of tumors. Recent studies have found that EVs exhibit specific expression patterns which mimic the parental cell, providing a fingerprint for early cancer diagnosis and prognosis as well as monitoring responses to treatment. Accordingly, various EV isolation and detection technologies have been developed for research and diagnostic purposes. Moreover, natural and engineered EVs have also been used as drug delivery nanocarriers, cancer vaccines, cell surface modulators, therapeutic agents and therapeutic targets. Overall, EVs are under intense investigation as they hold promise for pathophysiological and translational discoveries. This comprehensive review examines the latest EV research trends over the last five years, encompassing their roles in cancer pathophysiology, diagnostics and therapeutics. This review aims to examine the full spectrum of tumor-EV studies and provide a comprehensive foundation to enhance the field. The topics which are discussed and scrutinized in this review encompass isolation techniques and how these issues need to be overcome for EV-based diagnostics, EVs and their roles in cancer biology, biomarkers for diagnosis and monitoring, EVs as vaccines, therapeutic targets, and EVs as drug delivery systems. We will also examine the challenges involved in EV research and promote a framework for catalyzing scientific discovery and innovation for tumor-EV-focused research.© 2021 The Authors.
[19]
韩杰, 葛安, 马晓霞, 等. 外泌体提取及保存技术研究进展. 中国细胞生物学学报, 2021, 43(2): 451-459.
Han J, Ge A, Ma X X, et al. Research progress on the isolation and preservation techniques of exosomes. Chinese Journal of Cell Biology, 2021, 43(2): 451-459.
[20]
Shirejini S Z, Inci F. The Yin and Yang of exosome isolation methods: conventional practice, microfluidics, and commercial kits. Biotechnology Advances, 2022, 54: 107814.
[21]
Sidhom K, Obi P O, Saleem A. A review of exosomal isolation methods: is size exclusion chromatography the best option? International Journal of Molecular Sciences, 2020, 21(18): 6466.
[22]
Haraszti R A, Miller R, Stoppato M, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Molecular Therapy, 2018, 26(12): 2838-2847.
Exosomes can deliver therapeutic RNAs to neurons. The composition and the safety profile of exosomes depend on the type of the exosome-producing cell. Mesenchymal stem cells are considered to be an attractive cell type for therapeutic exosome production. However, scalable methods to isolate and manufacture exosomes from mesenchymal stem cells are lacking, a limitation to the clinical translation of exosome technology. We evaluate mesenchymal stem cells from different sources and find that umbilical cord-derived mesenchymal stem cells produce the highest exosome yield. To optimize exosome production, we cultivate umbilical cord-derived mesenchymal stem cells in scalable microcarrier-based three-dimensional (3D) cultures. In combination with the conventional differential ultracentrifugation, 3D culture yields 20-fold more exosomes (3D-UC-exosomes) than two-dimensional cultures (2D-UC-exosomes). Tangential flow filtration (TFF) in combination with 3D mesenchymal stem cell cultures further improves the yield of exosomes (3D-TFF-exosomes) 7-fold over 3D-UC-exosomes. 3D-TFF-exosomes are seven times more potent in small interfering RNA (siRNA) transfer to neurons compared with 2D-UC-exosomes. Microcarrier-based 3D culture and TFF allow scalable production of biologically active exosomes from mesenchymal stem cells. These findings lift a major roadblock for the clinical utility of mesenchymal stem cell exosomes.Copyright © 2018. Published by Elsevier Inc.
[23]
张亚萍. 负载三维培养脐带间充质干细胞外泌体的卵巢基质凝胶治疗化疗所致卵巢损伤的研究. 济南: 山东大学, 2023.
Zhang Y P. Study of ovarian ECM gel loaded with 3D-UC-MSC-exos for ovarian reserve against chemotherapy-induced injury. Jinan: Shandong University, 2023.
[24]
吴杰. 低氧培养人脐带间充质干细胞后分泌的外泌体对小鼠过敏性鼻炎的治疗作用及其机制. 南昌: 南昌大学, 2022.
Wu J. Therapeutic effect and mechanism of the exosomes secreted from human umbilical cord mesenchymal stem cells cultured with hypoxia on allergic rhinitis in mice. Nanchang: Nanchang University, 2022.
[25]
王琪. 三维培养的人脐带间充质干细胞来源外泌体在白癜风治疗中的作用和机制研究. 西安: 中国人民解放军空军军医大学, 2022.
Wang Q. Role and mechanism of exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells in the treatment of vitiligo. Xi’an: Chinese People’s Liberation Army Air Force Medical University, 2022.
[26]
Jafari D, Malih S, Eini M, et al. Improvement, scaling-up, and downstream analysis of exosome production. Critical Reviews in Biotechnology, 2020, 40(8): 1098-1112.
[27]
王宪峰, 欧昕, 邓必勇. 不同来源间充质干细胞外泌体治疗骨关节炎疗效的比较. 中国组织工程研究, 2022, 26(25): 3980-3985.
Wang X F, Ou X, Deng B Y. Comparison of effects of exosomes secreted by different mesenchymal stem cells for the treatment of osteoarthritis. Chinese Journal of Tissue Engineering Research, 2022, 26(25): 3980-3985.
[28]
Cai J X, Wu J Y, Wang J M, et al. Extracellular vesicles derived from different sources of mesenchymal stem cells: therapeutic effects and translational potential. Cell & Bioscience, 2020, 10: 69.
[29]
Deng H M, Zhu L N, Zhang Y G, et al. Differential lung protective capacity of exosomes derived from human adipose tissue, bone marrow, and umbilical cord mesenchymal stem cells in sepsis-induced acute lung injury. Oxidative Medicine and Cellular Longevity, 2022, 2022: 7837837.
[30]
Soni N, Gupta S, Rawat S, et al. MicroRNA-enriched exosomes from different sources of mesenchymal stem cells can differentially modulate functions of immune cells and neurogenesis. Biomedicines, 2021, 10(1): 69.
[31]
Pomatto M, Gai C, Negro F, et al. Differential therapeutic effect of extracellular vesicles derived by bone marrow and adipose mesenchymal stem cells on wound healing of diabetic ulcers and correlation to their cargoes. International Journal of Molecular Sciences, 2021, 22(8): 3851.
[32]
Chen S W, Wang Y B, Liao L M, et al. Similar repair effects of human placenta, bone marrow mesenchymal stem cells, and their exosomes for damaged SVOG ovarian granulosa cells. Stem Cells International, 2020, 2020: 8861557.
[33]
Semerci Sevimli T, Sevimli M, Qomi Ekenel E, et al. Comparison of exosomes secreted by synovial fluid-derived mesenchymal stem cells and adipose tissue-derived mesenchymal stem cells in culture for microRNA-127-5p expression during chondrogenesis. Gene, 2023, 865: 147337.
[34]
Théry C, Witwer K W, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles, 2018, 7(1): 1535750.
[35]
单政铭, 陶述春, 胡春梅, 等. 人脐带间充质干细胞来源外泌体的提取、鉴定和蛋白组学分析. 中国组织工程研究, 2022, 26(19): 3036-3042.
Shan Z M, Tao S C, Hu C M, et al. Extraction, identification and proteomic analysis of exosomes derived from human umbilical cord mesenchymal stem cells. Chinese Journal of Tissue Engineering Research, 2022, 26(19): 3036-3042.
[36]
Gupta S, Pinky, Vishal, et al. Comparative evaluation of anti-fibrotic effect of tissue specific mesenchymal stem cells derived extracellular vesicles for the amelioration of CCl4 induced chronic liver injury. Stem Cell Reviews and Reports, 2022, 18(3): 1097-1112.
[37]
Wu B T, Feng J X, Guo J Y, et al. ADSCs-derived exosomes ameliorate hepatic fibrosis by suppressing stellate cell activation and remodeling hepatocellular glutamine synthetase-mediated glutamine and ammonia homeostasis. Stem Cell Research & Therapy, 2022, 13(1): 494.
[38]
Rong X L, Liu J Z, Yao X, et al. Human bone marrow mesenchymal stem cells-derived exosomes alleviate liver fibrosis through the Wnt/β-catenin pathway. Stem Cell Research & Therapy, 2019, 10(1): 98.
[39]
Jiang W Q, Tan Y W, Cai M J, et al. Human umbilical cord MSC-derived exosomes suppress the development of CCl(4)-induced liver injury through antioxidant effect. Stem Cells International, 2018, 2018: 6079642.
[40]
游茂春, 刘广益, 程俊, 等. 脂肪干细胞及其外泌体减轻肝细胞凋亡改善大鼠肝纤维化. 中国比较医学杂志, 2020, 30(7): 30-37.
You M C, Liu G Y, Cheng J, et al. Adipose stem cells and their derived exosomes alleviate liver fibrosis in rats by reducing apoptosis. Chinese Journal of Comparative Medicine, 2020, 30(7): 30-37.
[41]
郑嵘炅, 邓泽润, 韩丹, 等. 骨髓间充质干细胞来源外泌体调节大鼠肝细胞凋亡的机制. 中国组织工程研究, 2024, 28(1): 44-49.
Zheng R J, Deng Z R, Han D, et al. Mechanism underlying rat hepatocyte apoptosis regulated by exosomes derived from bone marrow mesenchymal stem cells. Chinese Journal of Tissue Engineering Research, 2024, 28(1): 44-49.
[42]
Tan C Y, Lai R C, Wong W, et al. Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models. Stem Cell Research & Therapy, 2014, 5(3): 76.
[43]
Tian S Y, Zhou X, Zhang M, et al. Mesenchymal stem cell-derived exosomes protect against liver fibrosis via delivering miR-148a to target KLF6/STAT3 pathway in macrophages. Stem Cell Research & Therapy, 2022, 13(1): 330.
[44]
Hu Z Q, Zhao Y P, Jiang J, et al. Exosome-derived miR-142-5p from liver stem cells improves the progression of liver fibrosis by regulating macrophage polarization through CTSB. Environmental Toxicology, 2023, 38(8): 1860-1873.
[45]
Gan L H, Zheng L, Yao L, et al. Exosomes from adipose-derived mesenchymal stem cells improve liver fibrosis by regulating the miR-20a-5p/TGFBR2 axis to affect the p 38 MAPK/NF-κB pathway. Cytokine, 2023, 172: 156386.
[46]
Ma J, Li Y, Chen M X, et al. hMSCs-derived exosome circCDK 13 inhibits liver fibrosis by regulating the expression of MFGE8 through miR-17-5p/KAT2B. Cell Biology and Toxicology, 2023, 39(2): 1-22.
[47]
蔡梦洁. HucMSC来源的外泌体转运miR-373和Let-7b缓解肝纤维化. 镇江: 江苏大学, 2018.
Cai M J. Role and mechanism of hucMSC-ex derived miR-373 and let-7b in liver fibrosis. Zhenjiang: Jiangsu University, 2018.
[48]
Tan Y W, Huang Y, Mei R, et al. HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death & Disease, 2022, 13: 319.
[49]
王晨. 人脐带间质干细胞外泌体转运USP9X抑制肝窦内皮细胞毛细血管化缓解肝纤维化. 镇江: 江苏大学, 2022.
Wang C. Human umbilical cord mscs derived exosomes transport USP9X to inhibit liver sinusoidal endothelial cells capillarization and alleviate liver fibrosis. Zhenjiang: Jiangsu University, 2022.
[50]
Shah R A, Kowdley K V. Obeticholic acid for the treatment of nonalcoholic steatohepatitis. Expert Review of Gastroenterology & Hepatology, 2020, 14(5): 311-321.
[51]
Hindson J. Obeticholic acid for the treatment of NASH. Nature Reviews Gastroenterology & Hepatology, 2020, 17: 66.
[52]
Younossi Z M, Stepanova M, Nader F, et al. Obeticholic acid impact on quality of life in patients with nonalcoholic steatohepatitis: regenerate 18-month interim analysis. Clinical Gastroenterology and Hepatology, 2022, 20(9): 2050-2058.e12.
[53]
Jiang L Y, Zhang H J, Xiao D S, et al. Farnesoid X receptor (FXR): structures and ligands. Computational and Structural Biotechnology Journal, 2021, 19: 2148-2159.
Farnesoid X receptor (FXR) is a bile acid activated nuclear receptor (BAR) and is mainly expressed in the liver and intestine. Upon ligand binding, FXR regulates key genes involved in the metabolic process of bile acid synthesis, transport and reabsorption and is also involved in the metabolism of carbohydrates and lipids. Because of its important functions, FXR is considered as a promising drug target for the therapy of bile acid-related liver diseases. With the approval of obeticholic acid (OCA) as the first small molecule to target FXR, many other small molecules are being evaluated in clinical trials. This review summarizes the structures of FXR, especially its ligand binding domain, and the development of small molecules (including agonists and antagonists) targeting FXR.© 2021 The Author(s).
[54]
Zivko C, Witt F, Koeberle A, et al. Formulating elafibranor and obeticholic acid with phospholipids decreases drug-induced association of SPARC to extracellular vesicles from LX-2 human hepatic stellate cells. European Journal of Pharmaceutics and Biopharmaceutics, 2023, 182: 32-40.
[55]
Azizsoltani A, Hatami B, Zali M R, et al. Obeticholic acid-loaded exosomes attenuate liver fibrosis through dual targeting of the FXR signaling pathway and ECM remodeling. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, 2023, 168: 115777.
[56]
Wan Z, Zhao L B, Lu F, et al. Mononuclear phagocyte system blockade improves therapeutic exosome delivery to the myocardium. Theranostics, 2020, 10(1): 218-230.
Exosomes are emerging as a promising drug delivery carrier. However, rapid uptake of exosomes by the mononuclear phagocyte system (MPS) remains an obstacle for drug delivery into other targeted organs, including the heart. We hypothesized that prior blocking of uptake of exosomes by the MPS would improve their delivery to the targeted organs. Exosomes were isolated from the cell culture medium. Fluorescence-labeled exosomes were tracked and by fluorescence imaging. The expression of clathrin heavy chain (Cltc), cavolin1, Pak1 and Rhoa, known genes for endocytosis, were profiled in various cell lines and organs by qPCR. The knockdown efficiency of siRNA against Cltc was analyzed by Western blotting. Exosome and exosome were constructed by encapsulating isolated exosomes with siControl or siClathrin via electroporation, while exosome was constructed by encapsulating isolated exosomes with miR-21a. Doxorubicin-induced cardiotoxicity model was used to verify the therapeutic efficiency of the exosome-based miR-21a delivery by echocardiography. Exosomes were preferentially accumulated in the liver and spleen, mainly due to the presence of abundant macrophages. Besides the well-known phagocytic effect, efficient endocytosis also contributes to the uptake of exosomes by macrophages. Cltc was found to be highly expressed in the macrophages compared with other endocytosis-associated genes. Accordingly, knockdown of Cltc significantly decreased the uptake of exosomes by macrophages and. Moreover, prior injection of exosome strikingly improved the delivery efficiency of exosomes to organs other than spleen and liver. Consistently, compared with the direct injection of exosome, prior injection of exosome produced a much better therapeutic effect on cardiac function in the doxorubicin-induced cardiotoxicity mouse model. Prior blocking of endocytosis of exosomes by macrophages with exosome successfully and efficiently improves the distribution of following exosome in targeted organs, like the heart. The established two-step exosome delivery strategy (blocking the uptake of exosomes first followed by delivery of therapeutic exosomes) would be a promising method for gene therapy.© The author(s).
[57]
Parada N, Romero-Trujillo A, Georges N, et al. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. Journal of Advanced Research, 2021, 31: 61-74.
Even though exosome-based therapy has been shown to be able to control the progression of different pathologies, the data revealed by pharmacokinetic studies warn of the low residence time of exogenous exosomes in circulation that can hinder the clinical translation of therapeutic exosomes. The macrophages related to the organs of the mononuclear phagocytic system are responsible primarily for the rapid clearance and retention of exosomes, which strongly limits the amount of exosomal particles available to reach the target tissue, accumulate in it and release with high efficiency its therapeutic cargo in acceptor target cells to exert the desired biological effect.Endowing exosomes with surface modifications to evade the immune system is a plausible strategy to contribute to the suppression of exosomal clearance and increase the efficiency of their targeted content delivery. Here, we summarize the current evidence about the mechanisms underlying the recognition and sequestration of therapeutic exosomes by phagocytic cells. Also, we propose different strategies to generate 'invisible' exosomes for the immune system, through the incorporation of different anti-phagocytic molecules on the exosomes' surface that allow increasing the circulating half-life of therapeutic exosomes with the purpose to increase their bioavailability to reach the target tissue, transfer their therapeutic molecular cargo and improve their efficacy profile.Macrophage-mediated phagocytosis are the main responsible behind the short half-life in circulation of systemically injected exosomes, hindering their therapeutic effect. Exosomes 'Camouflage Cloak' strategy using antiphagocytic molecules can contribute to the inhibition of exosomal clearance, hence, increasing the on-target effect. Some candidate molecules that could exert an antiphagocytic role are CD47, CD24, CD44, CD31, β2M, PD-L1, App1, and DHMEQ. Pre- and post-isolation methods for exosome engineering are compatible with the loading of therapeutic cargo and the expression of antiphagocytic surface molecules.© 2021 The Authors. Published by Elsevier B.V. on behalf of Cairo University.
[58]
Lin Y, Yan M C, Bai Z T, et al. Huc-MSC-derived exosomes modified with the targeting peptide of aHSCs for liver fibrosis therapy. Journal of Nanobiotechnology, 2022, 20(1): 432.
Effective therapeutics to stop or reverse liver fibrosis have not emerged, because these potential agents cannot specifically target activated hepatic stellate cells (aHSCs) or are frequently toxic to parenchymal cells. Human umbilical cord mesenchymal stem cell (Huc-MSC)-derived exosomes show promise in nanomedicine for the treatment of liver fibrosis. However, systemic injection showed that unmodified exosomes were mainly taken up by the mononuclear phagocyte system. The discovery of ligands that selectively bind to a specific target plays a crucial role in clinically relevant diagnostics and therapeutics. Herein, we aimed to identify the targeting peptide of aHSCs by screening a phage-displayed peptide library, and modify Huc-MSC-derived exosomes with the targeting peptide.In this study, we screened a phage-displayed peptide library by biopanning for peptides preferentially bound to HSC-T6 cells. The identified peptide, HSTP1, also exhibited better targeting ability to aHSCs in pathological sections of fibrotic liver tissues. Then, HSTP1 was fused with exosomal enriched membrane protein (Lamp2b) and was displayed on the surface of exosomes through genetic engineering technology. The engineered exosomes (HSTP1-Exos) could be more efficiently internalized by HSC-T6 cells and outperformed both unmodified exosomes (Blank-Exos) and Lamp2b protein overexpressed exosomes (Lamp2b + Exos) in enhancing the ability of exosomes to promote HSC-T6 reversion to a quiescent phenotype. In vivo results showed HSTP1-Exos could specifically target to the aHSC region after intravenous administration, as demonstrated by coimmunofluorescence with the typical aHSCs marker α-SMA, and enhance the therapeutic effect on liver fibrosis.These results suggest that HSTP1 is a reliable targeting peptide that can specifically bind to aHSCs and that HSTP1-modified exosomes realize the precise treatment for aHSCs in complex liver tissue. We provide a novel strategy for clinical liver fibrosis therapy.© 2022. The Author(s).
[59]
Hao Y M, Song K C, Tan X C, et al. Reactive oxygen species-responsive polypeptide drug delivery system targeted activated hepatic stellate cells to ameliorate liver fibrosis. ACS Nano, 2022, 16(12): 20739-20757.
Hepatic fibrosis is a chronic liver disease that lacks effective pharmacotherapeutic treatments. As part of the disease's mechanism, hepatic stellate cells (HSCs) are activated by damage-related stimuli to secrete excessive extracellular matrix, leading to collagen deposition. Currently, the drug delivery system that targets HSCs in the treatment of liver fibrosis remains an urgent challenge due to the poor controllability of drug release. Since the level of reactive oxygen species (ROS) increases sharply in activated HSCs (aHSCs), we designed ROS-responsive micelles for the HSC-specific delivery of a traditional Chinese medicine, resveratrol (RES), for treatment of liver fibrosis. The micelles were prepared by the ROS-responsive amphiphilic block copolymer poly(l-methionine--N-trifluoro-acetyl-l-lysine) (PMK) and a PEG shell modified with a CRGD peptide insertion. The CRGD-targeted and ROS-responsive micelles (CRGD-PMK-MCs) could target aHSCs and control the release of RES under conditions of high intracellular ROS in aHSCs. The CRGD-PMK-MCs treatment specifically enhanced the targeted delivery of RES to aHSCs both in vitro and in vivo. In vitro experiments show that CRGD-PMK-MCs could significantly promote ROS consumption, reduce collagen accumulation, and avert activation of aHSCs. In vivo results demonstrate that CRGD-PMK-MCs could alleviate inflammatory infiltration, prevent fibrosis, and protect hepatocytes from damage in fibrotic mice. In conclusion, CRGD-PMK-MCs show great potential for targeted and ROS-responsive controlled drug release in the aHSCs of liver fibrosis.
[60]
Ji D, Wang Q H, Zhao Q, et al. Co-delivery of miR-29b and germacrone based on cyclic RGD-modified nanoparticles for liver fibrosis therapy. Journal of Nanobiotechnology, 2020, 18(1): 86.
Hepatic stellate cells (HSCs) were activated and secreted excessive amounts of extracellular matrix (ECM) proteins during pathogenetic progress of liver fibrosis. Germacrone (GMO) and miR-29b can play an important role in inhibiting growth of HSCs and production of type I collagen. GMO and miR-29b were co-encapsulated into nanoparticles (NPs) based on poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PEG-PLGA). Then, NPs were modified with cyclic RGD peptides (cRGDfK). cRGDfK is an effective ligand to bind integrin αβ and increase the targeting ability for fibrotic liver. GMO- and miR-29b-loaded NPs exhibited great cytotoxicity to activated HSCs and significantly inhibited production of type I collagen. Liver fibrosis model of mice was induced by administration of carbon tetrachloride. Great targeting ability was achieved in liver fibrotic mice treated with cRGD-modified NPs. Significant ant-fibrotic effects have been presented based on hematoxylin and eosin (H&E), Masson and Sirius Red staining results of liver tissues collected from mice treated with drug-loaded NPs. All these results indicate GMO- and miR-29b-loaded cRGD-modified NPs have the potential for clinical use to treat liver fibrosis.
[61]
Li W H, Zhou C C, Fu Y, et al. Targeted delivery of hyaluronic acid nanomicelles to hepatic stellate cells in hepatic fibrosis rats. Acta Pharmaceutica Sinica B, 2020, 10(4): 693-710.
Hepatic fibrosis is one kind of liver diseases with a high mortality rate and incidence. The activation and proliferation of hepatic stellate cells (HSCs) is the most fundamental reason of hepatic fibrosis. There are no specific and effective drug delivery carriers for the treatment of hepatic fibrosis at present. We found that when hepatic fibrosis occurs, the expression of CD44 receptors on the surface of HSCs is significantly increased. Based on this finding, we designed silibinin-loaded hyaluronic acid (SLB-HA) micelles to achieve the treatment of hepatic fibrosis. Meanwhile, we constructed liver fibrosis rat model using Sprague-Dawley rats. We demonstrated that HA micelles had specific uptake to HSCs while avoiding the distribution in normal liver cells and the phagocytosis of macrophages. Importantly, HA micelles showed a significant liver targeting effect, especially in fibrotic liver which highly expressed CD44 receptors. In addition, SLB-HA micelles could selectively kill activated HSCs, having an excellent anti-hepatic fibrosis effect and a significant sustained release effect, and also had a good biological safety and biocompatibility. Overall, HA micelles represented a novel nanomicelle system which showed great potentiality in anti-hepatic fibrosis drugs delivery.© 2020 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences. Production and hosting by Elsevier B.V.
[62]
Dutta R, Kumar V, Peng Y, et al. Pharmacokinetics and biodistribution of GDC-0449 loaded micelles in normal and liver fibrotic mice. Pharmaceutical Research, 2017, 34(3): 564-578.
To determine the pharmacokinetic parameters and biodistribution of GDC-0449 loaded polymeric micelles after systemic administration into common bile duct ligation (CBDL) induced liver fibrotic mice.We used GDC-0449 encapsulated methoxy poly (ethylene glycol)-block-poly (2-methyl-2-carboxyl-propylene carbonate)-graft-dodecanol (PEG-PCD) non-targeted polymeric micelles for GDC-0449 delivery to normal and liver fibrotic mice. To maximize GDC-0449 delivery to hepatic stellate cells (HSCs), mixed micelles formulations with 10, 20 and 30% w/w mannose-6-phosphate (M6P)-conjugated micelles were administered to normal and liver fibrotic mice for targeting M6P/IGF-IIR overexpressed on activated HSCs and biodistribution of GDC-0449 was determined at 30 and 120 min post systemic administration.GDC-0449 distributed to all major organs after systemic administration of drug loaded micelles, with higher accumulation in the liver of both normal and fibrotic mice. The plasma concentration versus time profiles suggest rapid clearance of GDC-0449 after systemic administration of drug loaded micelles in both normal and fibrotic mice, with similar plasma clearance (CL), area under the curve (AUC) and volume of distribution at steady state (V). However, there is significant increase in GDC-0449 accumulation in the liver when M6P-conjugated mixed micelles were injected, with the highest GDC-0449 concentration in the liver with mixed micelles carrying 30% M6P-conjugated polymer. HSCs accounted for 14.19% of GDC-0449 accumulation for M6P-targeted micelles in fibrotic mice compared to 5.62% of non-targeted micelles in the liver uptake study.M6P-conjugated GDC-0449 loaded mixed micelles may be used as a potential drug delivery vehicle for treating liver fibrosis.
[63]
You D G, Oh B H, Nguyen V Q, et al. Vitamin A-coupled stem cell-derived extracellular vesicles regulate the fibrotic cascade by targeting activated hepatic stellate cells in vivo. Journal of Controlled Release, 2021, 336: 285-295.
[64]
Zhang Y W, Hou L S, Xing J H, et al. Two-membrane hybrid nanobiomimetic delivery system for targeted autophagy inhibition of activated hepatic stellate cells to synergistically treat liver fibrosis. ACS Applied Materials & Interfaces, 2023. DOI: 10.1021/acsami.3c11046.
[65]
Lee J, Lee H, Goh U, et al. Cellular engineering with membrane fusogenic liposomes to produce functionalized extracellular vesicles. ACS Applied Materials & Interfaces, 2016, 8(11): 6790-6795.
[66]
Hood J L, Scott M J, Wickline S A. Maximizing exosome colloidal stability following electroporation. Analytical Biochemistry, 2014, 448: 41-49.
Development of exosome-based semisynthetic nanovesicles for diagnostic and therapeutic purposes requires novel approaches to load exosomes with cargo. Electroporation has previously been used to load exosomes with RNA. However, investigations into exosome colloidal stability following electroporation have not been considered. Herein, we report the development of a unique trehalose pulse media (TPM) that minimizes exosome aggregation following electroporation. Dynamic light scattering (DLS) and RNA absorbance were employed to determine the extent of exosome aggregation and electroextraction post electroporation in TPM compared to common PBS pulse media or sucrose pulse media (SPM). Use of TPM to disaggregate melanoma exosomes post electroporation was dependent on both exosome concentration and electric field strength. TPM maximized exosome dispersal post electroporation for both homogenous B16 melanoma and heterogeneous human serum-derived populations of exosomes. Moreover, TPM enabled heavy cargo loading of melanoma exosomes with 5nm superparamagnetic iron oxide nanoparticles (SPION5) while maintaining original exosome size and minimizing exosome aggregation as evidenced by transmission electron microscopy. Loading exosomes with SPION5 increased exosome density on sucrose gradients. This provides a simple, label-free means of enriching exogenously modified exosomes and introduces the potential for MRI-driven theranostic exosome investigations in vivo. Copyright © 2013 Elsevier Inc. All rights reserved.
[67]
Kim M S, Haney M J, Zhao Y L, et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine, 2016, 12(3): 655-664.
Exosomes have recently come into focus as "natural nanoparticles" for use as drug delivery vehicles. Our objective was to assess the feasibility of an exosome-based drug delivery platform for a potent chemotherapeutic agent, paclitaxel (PTX), to treat MDR cancer. Herein, we developed different methods of loading exosomes released by macrophages with PTX (exoPTX), and characterized their size, stability, drug release, and in vitro antitumor efficacy. Reformation of the exosomal membrane upon sonication resulted in high loading efficiency and sustained drug release. Importantly, incorporation of PTX into exosomes increased cytotoxicity more than 50 times in drug resistant MDCKMDR1 (Pgp+) cells. Next, our studies demonstrated a nearly complete co-localization of airway-delivered exosomes with cancer cells in a model of murine Lewis lung carcinoma pulmonary metastases, and a potent anticancer effect in this mouse model. We conclude that exoPTX holds significant potential for the delivery of various chemotherapeutics to treat drug resistant cancers.Exosomes are membrane-derived natural vesicles of ~40 - 200 nm size. They have been under extensive research as novel drug delivery vehicles. In this article, the authors developed exosome-based system to carry formulation of PTX and showed efficacy in the treatment of multi-drug resistant cancer cells. This novel system may be further developed to carry other chemotherapeutic agents in the future.Published by Elsevier Inc.
[68]
Ashour A A, El-Kamel A H, Mehanna R A, et al. Luteolin-loaded exosomes derived from bone marrow mesenchymal stem cells: a promising therapy for liver fibrosis. Drug Delivery, 2022, 29(1): 3270-3280.
Liver fibrosis is a global life-threatening disorder with no approved treatment. It leads to serious hepatic complications when progressive, such as cirrhosis and carcinoma. Luteolin (LUT) is a plant flavonoid possessing a promising therapeutic potential in various liver diseases particularly, liver fibrosis. It was reported to have potent anti-inflammatory and antioxidant properties. It also suppresses the proliferation of activated hepatic stellate cells (HSC) and induces their apoptosis. However, its poor aqueous solubility and exposure to metabolism hinder its clinical use. Mesenchymal stem cells (MSCs)-derived exosomes, nano-sized extracellular vesicles, have recently emerged as natural biocompatible drug delivery vehicles permitting efficient drug delivery. Accordingly, the present study aimed for the first time to investigate the potential of bone marrow MSCs-derived exosomes to improve LUTs antifibrotic effectiveness. LUT-loaded exosomes (LUT-Ex) were successfully developed, optimized and subjected to both and characterization. The elaborated LUT-Ex presented good colloidal properties (size; 150 nm, PDI; 0.3 and ζ-potential; -28 mV), typical vesicular shape, reasonable drug entrapment efficiency (40%) with sustained drug release over 72 h. Additionally, the cellular uptake study of coumarin-6-loaded exosomes in HEP-G2 revealed a significant enhancement in their uptake by 78.4% versus free coumarin-6 solution ( ≤ 0.001). Following a single intraperitoneal injection, LUT-Ex revealed a superior antifibrotic activity compared with either LUT-suspension or blank exosomes as evidenced by the results of biochemical and histopathological evaluation. In conclusion, the elaborated LUT-Ex could be addressed as a promising nanocarrier for effective treatment of liver fibrosis.
[69]
Didamoony M A, Atwa A M, Ahmed L A. Modulatory effect of rupatadine on mesenchymal stem cell-derived exosomes in hepatic fibrosis in rats: a potential role for miR-200a. Life Sciences, 2023, 324: 121710.
[70]
Ellakany A R, El Baz H, Shoheib Z S, et al. Stem cell-derived exosomes as a potential therapy for schistosomal hepatic fibrosis in experimental animals. Pathogens and Global Health, 2023: 1-21.
[71]
Willms E, Johansson H J, Mäger I, et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Scientific Reports, 2016, 6: 22519.
Cells release nano-sized membrane vesicles that are involved in intercellular communication by transferring biological information between cells. It is generally accepted that cells release at least three types of extracellular vesicles (EVs): apoptotic bodies, microvesicles and exosomes. While a wide range of putative biological functions have been attributed to exosomes, they are assumed to represent a homogenous population of EVs. We hypothesized the existence of subpopulations of exosomes with defined molecular compositions and biological properties. Density gradient centrifugation of isolated exosomes revealed the presence of two distinct subpopulations, differing in biophysical properties and their proteomic and RNA repertoires. Interestingly, the subpopulations mediated differential effects on the gene expression programmes in recipient cells. In conclusion, we demonstrate that cells release distinct exosome subpopulations with unique compositions that elicit differential effects on recipient cells. Further dissection of exosome heterogeneity will advance our understanding of exosomal biology in health and disease and accelerate the development of exosome-based diagnostics and therapeutics.
[72]
Du Z Y, Wu T C, Liu L S, et al. Extracellular vesicles-derived miR-150-5p secreted by adipose-derived mesenchymal stem cells inhibits CXCL 1 expression to attenuate hepatic fibrosis. Journal of Cellular and Molecular Medicine, 2021, 25(2): 701-715.
[73]
Kim J, Lee C B, Shin Y, et al. sEVs from tonsil-derived mesenchymal stromal cells alleviate activation of hepatic stellate cells and liver fibrosis through miR-486-5p. Molecular Therapy, 2021, 29(4): 1471-1486.
Mesenchymal stromal cells (MSCs) are considered as a promising therapeutic tool for liver fibrosis, a main feature of chronic liver disease. Because small extracellular vesicles (sEVs) harboring a variety of proteins and RNAs are known to have similar functions with their derived cells, MSC-derived sEVs carry out the regenerative capacities of MSCs. Human tonsil-derived MSCs (T-MSCs) are reported as a novel source of MSCs, but their effects on liver fibrosis remain unclear. In the present study, we investigated the effects of T-MSC-derived sEVs on liver fibrosis. The expression of profibrotic genes decreased in human primary hepatic stellate cells (pHSCs) co-cultured with T-MSCs. Treatment of T-MSC-sEVs inactivated human and mouse pHSCs. Administration of T-MSC-sEVs ameliorated hepatic injuries and fibrosis in chronically damaged liver induced by carbon tetrachloride (CCl). miR-486-5p highly enriched in T-MSC-sEVs targeting the hedgehog receptor, smoothened (Smo), was upregulated, whereas Smo and Gli2, the hedgehog target gene, were downregulated in pHSCs and liver tissues treated with T-MSC-sEVs or miR-486-5p mimic, indicating that sEV-miR-486 inactivates HSCs by suppressing hedgehog signaling. Our results showed that T-MSCs attenuate HSC activation and liver fibrosis by delivering sEVs, and miR-486 in the sEVs inactivates hedgehog signaling, suggesting that T-MSCs and their sEVs are novel anti-fibrotic therapeutics for treating chronic liver disease.Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.
[74]
Qian G E, Morral N. Role of non-coding RNAs on liver metabolism and NAFLD pathogenesis. Human Molecular Genetics, 2022, 31(R1): R4-R21.
[75]
Lou G H, Yang Y, Liu F F, et al. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. Journal of Cellular and Molecular Medicine, 2017, 21(11): 2963-2973.
Mesenchymal stem cell (MSC) transplantation alone may be insufficient for treatment of liver fibrosis because of complicated histopathological changes in the liver. Given that miR-122 plays an essential role in liver fibrosis by negatively regulating the proliferation and transactivation of hepatic stellate cells (HSCs), this study investigated whether miR-122 modification can improve the therapeutic efficacy of adipose tissue-derived MSCs in treating liver fibrosis. MiR-122-modified AMSCs (AMSC-122) were constructed through lentivirus-mediated transfer of pre-miR-122. MiR-122-modified AMSCs expressed high level of miR-122, while they retained their phenotype and differentiation potential as naïve AMSCs. AMSC-122 more effectively suppressed the proliferation of and collagen maturation in HSCs than scramble miRNA-modified AMSCs. In addition, AMSC-derived exosomes mediated the miR-122 communication between AMSCs and HSCs, further affecting the expression levels of miR-122 target genes, such as insulin-like growth factor receptor 1 (IGF1R), Cyclin G(1) (CCNG1) and prolyl-4-hydroxylase α1 (P4HA1), which are involved in proliferation of and collagen maturation in HSCs. Moreover, miR-122 modification enhanced the therapeutic efficacy of AMSCs in the treatment of carbon tetrachloride (CCl )-induced liver fibrosis by suppressing the activation of HSCs and alleviating collagen deposition. Results demonstrate that miR-122 modification improves the therapeutic efficacy of AMSCs through exosome-mediated miR-122 communication; thus, miR-122 modification is a new potential strategy for treatment of liver fibrosis.© 2017 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.
[76]
Qu Y, Zhang Q D, Cai X B, et al. Exosomes derived from miR-181-5p-modified adipose-derived mesenchymal stem cells prevent liver fibrosis via autophagy activation. Journal of Cellular and Molecular Medicine, 2017, 21(10): 2491-2502.
Proliferating hepatic stellate cells (HSCs) respond to liver damage by secreting collagens that form fibrous scar tissue, which can lead to cirrhosis if in appropriately regulated. Advancement of microRNA (miRNA) hepatic therapies has been hampered by difficulties in delivering miRNA to damaged tissue. However, exosomes secreted by adipose-derived mesenchymal stem cells (ADSCs) can be exploited to deliver miRNAs to HSCs. ADSCs were engineered to overexpress miRNA-181-5p (miR-181-5p-ADSCs) to selectively home exosomes to mouse hepatic stellate (HST-T6) cells or a CCl4-induced liver fibrosis murine model and compared with non-targeting control Caenorhabditis elegans miR-67 (cel-miR-67)-ADSCs. In vitro analysis confirmed that the transfer of miR-181-5p from miR-181-5p-ADSCs occurred via secreted exosomal uptake. Exosomes were visualized in HST-T6 cells using cyc3-labelled pre-miRNA-transfected ADSCs with/without the exosomal inhibitor, GW4869. The effects of miRNA-181-5p overexpression on the fibrosis associated STAT3/Bcl-2/Beclin 1 pathway and components of the extracellular matrix were assessed. Exosomes from miR181-5p-ADSCs down-regulated Stat3 and Bcl-2 and activated autophagy in the HST-T6 cells. Furthermore, the up-regulated expression of fibrotic genes in HST-T6 cells induced by TGF-β1 was repressed following the addition of isolated miR181-5p-ADSC exosomes compared with miR-67-ADSCexosomes. Exosome therapy attenuated liver injury and significantly down-regulated collagen I, vimentin, α-SMA and fibronectin in liver, compared with controls. Taken together, the effective anti-fibrotic function of engineered ADSCs is able to selectively transfer miR-181-5p to damaged liver cells and will pave the way for the use of exosome-ADSCs for therapeutic delivery of miRNA targeting liver disease.© 2017 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.
[77]
Vo J N, Cieslik M, Zhang Y J, et al. The landscape of circular RNA in cancer. Cell, 2019, 176(4): 869-881.e13.
Circular RNAs (circRNAs) are an intriguing class of RNA due to their covalently closed structure, high stability, and implicated roles in gene regulation. Here, we used an exome capture RNA sequencing protocol to detect and characterize circRNAs across >2,000 cancer samples. When compared against Ribo-Zero and RNase R, capture sequencing significantly enhanced the enrichment of circRNAs and preserved accurate circular-to-linear ratios. Using capture sequencing, we built the most comprehensive catalog of circRNA species to date: MiOncoCirc, the first database to be composed primarily of circRNAs directly detected in tumor tissues. Using MiOncoCirc, we identified candidate circRNAs to serve as biomarkers for prostate cancer and were able to detect circRNAs in urine. We further detected a novel class of circular transcripts, termed read-through circRNAs, that involved exons originating from different genes. MiOncoCirc will serve as a valuable resource for the development of circRNAs as diagnostic or therapeutic targets across cancer types.Copyright © 2018 Elsevier Inc. All rights reserved.
[78]
Zhu M, Liu X, Li W, et al. Exosomes derived from mmu_circ_0000623-modified ADSCs prevent liver fibrosis via activating autophagy. Human & Experimental Toxicology, 2020, 39(12): 1619-1627.
[79]
Yu L, Xue J C, Wu Y Y, et al. Therapeutic effect of exosomes derived from hepatocyte-growth-factor-overexpressing adipose mesenchymal stem cells on liver injury. Folia Histochemica et Cytobiologica, 2023, 61(3): 160-171.
[80]
Takeuchi S, Tsuchiya A, Iwasawa T, et al. Small extracellular vesicles derived from interferon-γ pre-conditioned mesenchymal stromal cells effectively treat liver fibrosis. NPJ Regenerative Medicine, 2021, 6: 19.
Mesenchymal stromal cells (MSCs) are used for ameliorating liver fibrosis and aiding liver regeneration after cirrhosis; Here, we analyzed the therapeutic potential of small extracellular vesicles (sEVs) derived from interferon-γ (IFN-γ) pre-conditioned MSCs (γ-sEVs). γ-sEVs effectively induced anti-inflammatory macrophages with high motility and phagocytic abilities in vitro, while not preventing hepatic stellate cell (HSC; the major source of collagen fiber) activation in vitro. The proteome analysis of MSC-derived sEVs revealed anti-inflammatory macrophage inducible proteins (e.g., annexin-A1, lactotransferrin, and aminopeptidase N) upon IFN-γ stimulation. Furthermore, by enabling CXCR1+ macrophage accumulation in the damaged area, γ-sEVs ameliorated inflammation and fibrosis in the cirrhosis mouse model more effectively than sEVs. Single cell RNA-Seq analysis revealed diverse effects, such as induction of anti-inflammatory macrophages and regulatory T cells, in the cirrhotic liver after γ-sEV administration. Overall, IFN-γ pre-conditioning altered sEVs resulted in efficient tissue repair indicating a new therapeutic strategy.
[81]
曾韬. uMSC外泌体治疗肝纤维化的糖基化修饰相关机制及其转化应用的关键技术研究. 上海: 中国人民解放军海军军医大学, 2019.
Zeng T. Study on the glycosylation-related mechanisms of uMSC exosome in the treatment of hepatic fibrosis and the key technologies of its translational applications. Shanghai: Naval Medical University of the People’s Liberation Army of China, 2019.
PDF(862 KB)

Accesses

Citation

Detail

Sections
Recommended

/