Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD)
Abstract
:1. Introduction
2. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD)
3. MSC Extracellular Vesicles (MSC-EVs)
3.1. Properties of MSC
3.2. General Features of EVs
3.3. MSC-EVs
4. MSC-EV Therapy in MASH-like Models
4.1. Human Umbilical Cord MSC (hUC MSC) EVs
4.2. Bone Marrow MSC (BM MSC) EVs
4.3. Adipose MSC (AD MSC) EVs
4.4. Embryonic Stem Cell-Derived MSC (ESC MSC) EVs
4.5. Modification of MSC-EVs
5. Unanswered Questions
6. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P.; et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Ann. Hepatol. 2024, 29, 101133. [Google Scholar] [CrossRef]
- Fan, W.; Bradford, T.M.; Torok, N.J. Metabolic dysfunction-associated liver disease and diabetes: Matrix remodeling, fibrosis, and therapeutic implications. Ann. N. Y. Acad. Sci. 2024, 1538, 21–33. [Google Scholar] [CrossRef]
- Pouwels, S.; Sakran, N.; Graham, Y.; Leal, A.; Pintar, T.; Yang, W.; Kassir, R.; Singhal, R.; Mahawar, K.; Ramnarain, D. Non-alcoholic fatty liver disease (NAFLD): A review of pathophysiology, clinical management and effects of weight loss. BMC Endocr. Disord. 2022, 22, 63. [Google Scholar] [CrossRef] [PubMed]
- van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
- Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.J.; Wang, C. A review of the regulatory mechanisms of extracellular vesicles-mediated intercellular communication. Cell Commun. Signal 2023, 21, 77. [Google Scholar] [CrossRef]
- Van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [Google Scholar] [CrossRef] [PubMed]
- Blans, K.; Hansen, M.S.; Sorensen, L.V.; Hvam, M.L.; Howard, K.A.; Moller, A.; Wiking, L.; Larsen, L.B.; Rasmussen, J.T. Pellet-free isolation of human and bovine milk extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2017, 6, 1294340. [Google Scholar] [CrossRef] [PubMed]
- Clayton, A.; Boilard, E.; Buzas, E.I.; Cheng, L.; Falcon-Perez, J.M.; Gardiner, C.; Gustafson, D.; Gualerzi, A.; Hendrix, A.; Hoffman, A.; et al. Considerations towards a roadmap for collection, handling and storage of blood extracellular vesicles. J. Extracell. Vesicles 2019, 8, 1647027. [Google Scholar] [CrossRef] [PubMed]
- Gallo, A.; Tandon, M.; Alevizos, I.; Illei, G.G. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS ONE 2012, 7, e30679. [Google Scholar] [CrossRef]
- Iwai, K.; Minamisawa, T.; Suga, K.; Yajima, Y.; Shiba, K. Isolation of human salivary extracellular vesicles by iodixanol density gradient ultracentrifugation and their characterizations. J. Extracell. Vesicles 2016, 5, 30829. [Google Scholar] [CrossRef]
- Simpson, R.J.; Lim, J.W.; Moritz, R.L.; Mathivanan, S. Exosomes: Proteomic insights and diagnostic potential. Expert. Rev. Proteom. 2009, 6, 267–283. [Google Scholar] [CrossRef] [PubMed]
- Srinivasan, S.; Duval, M.X.; Kaimal, V.; Cuff, C.; Clarke, S.H. Assessment of methods for serum extracellular vesicle small RNA sequencing to support biomarker development. J. Extracell. Vesicles 2019, 8, 1684425. [Google Scholar] [CrossRef]
- Zonneveld, M.I.; Brisson, A.R.; van Herwijnen, M.J.; Tan, S.; van de Lest, C.H.; Redegeld, F.A.; Garssen, J.; Wauben, M.H.; Nolte-‘t Hoen, E.N. Recovery of extracellular vesicles from human breast milk is influenced by sample collection and vesicle isolation procedures. J. Extracell. Vesicles 2014, 3, 24215. [Google Scholar] [CrossRef] [PubMed]
- Rai, A.; Claridge, B.; Lozano, J.; Greening, D.W. The Discovery of Extracellular Vesicles and Their Emergence as a Next-Generation Therapy. Circ. Res. 2024, 135, 198–221. [Google Scholar] [CrossRef] [PubMed]
- Bahadorani, M.; Nasiri, M.; Dellinger, K.; Aravamudhan, S.; Zadegan, R. Engineering Exosomes for Therapeutic Applications: Decoding Biogenesis, Content Modification, and Cargo Loading Strategies. Int. J. Nanomed. 2024, 19, 7137–7164. [Google Scholar] [CrossRef] [PubMed]
- Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016, 428, 688–692. [Google Scholar] [CrossRef] [PubMed]
- Zhao, W.; Li, K.; Li, L.; Wang, R.; Lei, Y.; Yang, H.; Sun, L. Mesenchymal Stem Cell-Derived Exosomes as Drug Delivery Vehicles in Disease Therapy. Int. J. Mol. Sci. 2024, 25, 7715. [Google Scholar] [CrossRef] [PubMed]
- Sanz-Ros, J.; Mas-Bargues, C.; Romero-Garcia, N.; Huete-Acevedo, J.; Dromant, M.; Borras, C. Extracellular Vesicles as Therapeutic Resources in the Clinical Environment. Int. J. Mol. Sci. 2023, 24, 2344. [Google Scholar] [CrossRef]
- Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.; Chan, Y.S.; Wei, L.; Chin, S.M.; Azad, A.; et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 2359. [Google Scholar] [CrossRef]
- Zou, X.; Yuan, M.; Zhang, T.; Zheng, N.; Wu, Z. EVs Containing Host Restriction Factor IFITM3 Inhibited ZIKV Infection of Fetuses in Pregnant Mice through Trans-placenta Delivery. Mol. Ther. 2021, 29, 176–190. [Google Scholar] [CrossRef]
- Zou, X.; Yuan, M.; Zhang, T.; Wei, H.; Xu, S.; Jiang, N.; Zheng, N.; Wu, Z. Extracellular vesicles expressing a single-chain variable fragment of an HIV-1 specific antibody selectively target Env(+) tissues. Theranostics 2019, 9, 5657–5671. [Google Scholar] [CrossRef] [PubMed]
- Devarbhavi, H.; Asrani, S.K.; Arab, J.P.; Nartey, Y.A.; Pose, E.; Kamath, P.S. Global burden of liver disease: 2023 update. J. Hepatol. 2023, 79, 516–537. [Google Scholar] [CrossRef] [PubMed]
- Asrani, S.K.; Devarbhavi, H.; Eaton, J.; Kamath, P.S. Burden of liver diseases in the world. J. Hepatol. 2019, 70, 151–171. [Google Scholar] [CrossRef] [PubMed]
- Ayada, I.; van Kleef, L.A.; Alferink, L.J.M.; Li, P.; de Knegt, R.J.; Pan, Q. Systematically comparing epidemiological and clinical features of MAFLD and NAFLD by meta-analysis: Focusing on the non-overlap groups. Liver Int. 2022, 42, 277–287. [Google Scholar] [CrossRef] [PubMed]
- Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef]
- Riazi, K.; Azhari, H.; Charette, J.H.; Underwood, F.E.; King, J.A.; Afshar, E.E.; Swain, M.G.; Congly, S.E.; Kaplan, G.G.; Shaheen, A.A. The prevalence and incidence of NAFLD worldwide: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2022, 7, 851–861. [Google Scholar] [CrossRef]
- Estes, C.; Razavi, H.; Loomba, R.; Younossi, Z.; Sanyal, A.J. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 2018, 67, 123–133. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Henry, L. Epidemiology of non-alcoholic fatty liver disease and hepatocellular carcinoma. JHEP Rep. 2021, 3, 100305. [Google Scholar] [CrossRef]
- Stepanova, M.; Kabbara, K.; Mohess, D.; Verma, M.; Roche-Green, A.; AlQahtani, S.; Ong, J.; Burra, P.; Younossi, Z.M. Nonalcoholic steatohepatitis is the most common indication for liver transplantation among the elderly: Data from the United States Scientific Registry of Transplant Recipients. Hepatol. Commun. 2022, 6, 1506–1515. [Google Scholar] [CrossRef] [PubMed]
- Woestemeier, A.; Scognamiglio, P.; Zhao, Y.; Wagner, J.; Muscate, F.; Casar, C.; Siracusa, F.; Cortesi, F.; Agalioti, T.; Muller, S.; et al. Multicytokine-producing CD4+ T cells characterize the livers of patients with NASH. JCI Insight 2023, 8, e153831. [Google Scholar] [CrossRef]
- Xu, J.; Wang, Y.; Khoshdeli, M.; Peach, M.; Chuang, J.C.; Lin, J.; Tsai, W.W.; Mahadevan, S.; Minto, W.; Diehl, L.; et al. IL-31 levels correlate with pruritus in patients with cholestatic and metabolic liver diseases and is farnesoid X receptor responsive in NASH. Hepatology 2023, 77, 20–32. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Zhang, B.; Lai, R.C.; Sim, W.K.; Lam, K.P.; Lim, S.K. MSC-sEV Treatment Polarizes Pro-Fibrotic M2 Macrophages without Exacerbating Liver Fibrosis in NASH. Int. J. Mol. Sci. 2023, 24, 8092. [Google Scholar] [CrossRef] [PubMed]
- Hendrikx, T.; Porsch, F.; Kiss, M.G.; Rajcic, D.; Papac-Milicevic, N.; Hoebinger, C.; Goederle, L.; Hladik, A.; Shaw, L.E.; Horstmann, H.; et al. Soluble TREM2 levels reflect the recruitment and expansion of TREM2(+) macrophages that localize to fibrotic areas and limit NASH. J. Hepatol. 2022, 77, 1373–1385. [Google Scholar] [CrossRef] [PubMed]
- Moreno-Fernandez, M.E.; Miraldi, E.R.; Divanovic, S. Not Chopped Liver-A Careful, Fate-Mapping Study of Macrophages in NASH. Cell Metab. 2020, 32, 328–330. [Google Scholar] [CrossRef]
- Loomba, R.; Friedman, S.L.; Shulman, G.I. Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell 2021, 184, 2537–2564. [Google Scholar] [CrossRef] [PubMed]
- Mendez-Sanchez, N.; Valencia-Rodriguez, A.; Coronel-Castillo, C.; Vera-Barajas, A.; Contreras-Carmona, J.; Ponciano-Rodriguez, G.; Zamora-Valdes, D. The cellular pathways of liver fibrosis in non-alcoholic steatohepatitis. Ann. Transl. Med. 2020, 8, 400. [Google Scholar] [CrossRef]
- Greenhill, C. NASH: Understanding how steatosis progresses to NASH. Nat. Rev. Endocrinol. 2017, 13, 5. [Google Scholar] [CrossRef]
- St Rose, K.; Yan, J.; Xu, F.; Williams, J.; Dweck, V.; Saxena, D.; Schwabe, R.F.; Caviglia, J.M. Mouse model of NASH that replicates key features of the human disease and progresses to fibrosis stage 3. Hepatol. Commun. 2022, 6, 2676–2688. [Google Scholar] [CrossRef]
- Sookoian, S.; Pirola, C.J. Nonalcoholic Fatty Liver Disease Progresses into Severe NASH when Physiological Mechanisms of Tissue Homeostasis Collapse. Ann. Hepatol. 2018, 17, 182–186. [Google Scholar] [CrossRef]
- Angulo, P.; Kleiner, D.E.; Dam-Larsen, S.; Adams, L.A.; Bjornsson, E.S.; Charatcharoenwitthaya, P.; Mills, P.R.; Keach, J.C.; Lafferty, H.D.; Stahler, A.; et al. Liver Fibrosis, but No Other Histologic Features, Is Associated with Long-term Outcomes of Patients with Nonalcoholic Fatty Liver Disease. Gastroenterology 2015, 149, 389–397 e310. [Google Scholar] [CrossRef] [PubMed]
- Ekstedt, M.; Hagstrom, H.; Nasr, P.; Fredrikson, M.; Stal, P.; Kechagias, S.; Hultcrantz, R. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015, 61, 1547–1554. [Google Scholar] [CrossRef]
- Vilar-Gomez, E.; Calzadilla-Bertot, L.; Wai-Sun Wong, V.; Castellanos, M.; Aller-de la Fuente, R.; Metwally, M.; Eslam, M.; Gonzalez-Fabian, L.; Alvarez-Quinones Sanz, M.; Conde-Martin, A.F.; et al. Fibrosis Severity as a Determinant of Cause-Specific Mortality in Patients With Advanced Nonalcoholic Fatty Liver Disease: A Multi-National Cohort Study. Gastroenterology 2018, 155, 443–457 e417. [Google Scholar] [CrossRef]
- Vilar-Gomez, E.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Friedman, S.L.; Diago, M.; Romero-Gomez, M. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology 2015, 149, 367–378 e365; quiz e314–365. [Google Scholar] [CrossRef]
- Dixon, J.B.; Bhathal, P.S.; Hughes, N.R.; O’Brien, P.E. Nonalcoholic fatty liver disease: Improvement in liver histological analysis with weight loss. Hepatology 2004, 39, 1647–1654. [Google Scholar] [CrossRef]
- Naoumov, N.V.; Brees, D.; Loeffler, J.; Chng, E.; Ren, Y.; Lopez, P.; Tai, D.; Lamle, S.; Sanyal, A.J. Digital pathology with artificial intelligence analyses provides greater insights into treatment-induced fibrosis regression in NASH. J. Hepatol. 2022, 77, 1399–1409. [Google Scholar] [CrossRef]
- Lassailly, G.; Caiazzo, R.; Ntandja-Wandji, L.C.; Gnemmi, V.; Baud, G.; Verkindt, H.; Ningarhari, M.; Louvet, A.; Leteurtre, E.; Raverdy, V.; et al. Bariatric Surgery Provides Long-term Resolution of Nonalcoholic Steatohepatitis and Regression of Fibrosis. Gastroenterology 2020, 159, 1290–1301 e1295. [Google Scholar] [CrossRef]
- Romero-Gomez, M.; Zelber-Sagi, S.; Trenell, M. Treatment of NAFLD with diet, physical activity and exercise. J. Hepatol. 2017, 67, 829–846. [Google Scholar] [CrossRef]
- Wang, S.; Friedman, S.L. Found in translation-Fibrosis in metabolic dysfunction-associated steatohepatitis (MASH). Sci. Transl. Med. 2023, 15, eadi0759. [Google Scholar] [CrossRef]
- Harrison, S.A.; Bedossa, P.; Guy, C.D.; Schattenberg, J.M.; Loomba, R.; Taub, R.; Labriola, D.; Moussa, S.E.; Neff, G.W.; Rinella, M.E.; et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N. Engl. J. Med. 2024, 390, 497–509. [Google Scholar] [CrossRef]
- Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed]
- Kwon, S.G.; Kwon, Y.W.; Lee, T.W.; Park, G.T.; Kim, J.H. Recent advances in stem cell therapeutics and tissue engineering strategies. Biomater. Res. 2018, 22, 36. [Google Scholar] [CrossRef]
- Huang, G.T.; Gronthos, S.; Shi, S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: Their biology and role in regenerative medicine. J. Dent. Res. 2009, 88, 792–806. [Google Scholar] [CrossRef]
- Calle, A.; Gutierrez-Reinoso, M.A.; Re, M.; Blanco, J.; De la Fuente, J.; Monguio-Tortajada, M.; Borras, F.E.; Yanez-Mo, M.; Ramirez, M.A. Bovine peripheral blood MSCs chemotax towards inflammation and embryo implantation stimuli. J. Cell Physiol. 2021, 236, 1054–1067. [Google Scholar] [CrossRef]
- Beeravolu, N.; McKee, C.; Alamri, A.; Mikhael, S.; Brown, C.; Perez-Cruet, M.; Chaudhry, G.R. Isolation and Characterization of Mesenchymal Stromal Cells from Human Umbilical Cord and Fetal Placenta. J. Vis. Exp. 2017, 122, 55224. [Google Scholar] [CrossRef]
- Altrock, E.; Sens-Albert, C.; Hofmann, F.; Riabov, V.; Schmitt, N.; Xu, Q.; Jann, J.C.; Rapp, F.; Steiner, L.; Streuer, A.; et al. Significant improvement of bone marrow-derived MSC expansion from MDS patients by defined xeno-free medium. Stem Cell Res. Ther. 2023, 14, 156. [Google Scholar] [CrossRef]
- Barbash, I.M.; Chouraqui, P.; Baron, J.; Feinberg, M.S.; Etzion, S.; Tessone, A.; Miller, L.; Guetta, E.; Zipori, D.; Kedes, L.H.; et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: Feasibility, cell migration, and body distribution. Circulation 2003, 108, 863–868. [Google Scholar] [CrossRef]
- Jager, M.; Zilkens, C.; Bittersohl, B.; Krauspe, R. Cord blood—An alternative source for bone regeneration. Stem Cell Rev. Rep. 2009, 5, 266–277. [Google Scholar] [CrossRef]
- Owston, H.E.; Moisley, K.M.; Tronci, G.; Russell, S.J.; Giannoudis, P.V.; Jones, E. Induced Periosteum-Mimicking Membrane with Cell Barrier and Multipotential Stromal Cell (MSC) Homing Functionalities. Int. J. Mol. Sci. 2020, 21, 5233. [Google Scholar] [CrossRef]
- Xie, Q.; Liu, R.; Jiang, J.; Peng, J.; Yang, C.; Zhang, W.; Wang, S.; Song, J. What is the impact of human umbilical cord mesenchymal stem cell transplantation on clinical treatment? Stem Cell Res. Ther. 2020, 11, 519. [Google Scholar] [CrossRef] [PubMed]
- Uder, C.; Bruckner, S.; Winkler, S.; Tautenhahn, H.M.; Christ, B. Mammalian MSC from selected species: Features and applications. Cytometry A 2018, 93, 32–49. [Google Scholar] [CrossRef] [PubMed]
- Deniz, I.A.; Karbanova, J.; Wobus, M.; Bornhauser, M.; Wimberger, P.; Kuhlmann, J.D.; Corbeil, D. Mesenchymal stromal cell-associated migrasomes: A new source of chemoattractant for cells of hematopoietic origin. Cell Commun. Signal 2023, 21, 36. [Google Scholar] [CrossRef] [PubMed]
- Liu, F.; Qiu, H.; Xue, M.; Zhang, S.; Zhang, X.; Xu, J.; Chen, J.; Yang, Y.; Xie, J. MSC-secreted TGF-beta regulates lipopolysaccharide-stimulated macrophage M2-like polarization via the Akt/FoxO1 pathway. Stem Cell Res. Ther. 2019, 10, 345. [Google Scholar] [CrossRef] [PubMed]
- Tan, T.T.; Toh, W.S.; Lai, R.C.; Lim, S.K. Practical considerations in transforming MSC therapy for neurological diseases from cell to EV. Exp. Neurol. 2022, 349, 113953. [Google Scholar] [CrossRef]
- Li, T.; Su, X.; Lu, P.; Kang, X.; Hu, M.; Li, C.; Wang, S.; Lu, D.; Shen, S.; Huang, H.; et al. Bone Marrow Mesenchymal Stem Cell-Derived Dermcidin-Containing Migrasomes enhance LC3-Associated Phagocytosis of Pulmonary Macrophages and Protect against Post-Stroke Pneumonia. Adv. Sci. 2023, 10, e2206432. [Google Scholar] [CrossRef]
- Najar, M.; Fahmi, H.; Merimi, M. The Medicinal Potential of Mesenchymal Stem/Stromal Cells in Immuno- and Cancer Therapy. Biomolecules 2023, 13, 1171. [Google Scholar] [CrossRef]
- Sarsenova, M.; Issabekova, A.; Abisheva, S.; Rutskaya-Moroshan, K.; Ogay, V.; Saparov, A. Mesenchymal Stem Cell-Based Therapy for Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 11592. [Google Scholar] [CrossRef]
- Lou, S.; Duan, Y.; Nie, H.; Cui, X.; Du, J.; Yao, Y. Mesenchymal stem cells: Biological characteristics and application in disease therapy. Biochimie 2021, 185, 9–21. [Google Scholar] [CrossRef] [PubMed]
- Shadmanfar, S.; Labibzadeh, N.; Emadedin, M.; Jaroughi, N.; Azimian, V.; Mardpour, S.; Kakroodi, F.A.; Bolurieh, T.; Hosseini, S.E.; Chehrazi, M.; et al. Intra-articular knee implantation of autologous bone marrow-derived mesenchymal stromal cells in rheumatoid arthritis patients with knee involvement: Results of a randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy 2018, 20, 499–506. [Google Scholar] [CrossRef] [PubMed]
- Ghoryani, M.; Shariati-Sarabi, Z.; Tavakkol-Afshari, J.; Ghasemi, A.; Poursamimi, J.; Mohammadi, M. Amelioration of clinical symptoms of patients with refractory rheumatoid arthritis following treatment with autologous bone marrow-derived mesenchymal stem cells: A successful clinical trial in Iran. Biomed. Pharmacother. 2019, 109, 1834–1840. [Google Scholar] [CrossRef]
- Lan, T.; Luo, M.; Wei, X. Mesenchymal stem/stromal cells in cancer therapy. J. Hematol. Oncol. 2021, 14, 195. [Google Scholar] [CrossRef] [PubMed]
- Wu, R.; Fan, X.; Wang, Y.; Shen, M.; Zheng, Y.; Zhao, S.; Yang, L. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Liver Immunity and Therapy. Front. Immunol. 2022, 13, 833878. [Google Scholar] [CrossRef]
- Vulliet, P.R.; Greeley, M.; Halloran, S.M.; MacDonald, K.A.; Kittleson, M.D. Intra-coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. Lancet 2004, 363, 783–784. [Google Scholar] [CrossRef] [PubMed]
- Jeong, J.O.; Han, J.W.; Kim, J.M.; Cho, H.J.; Park, C.; Lee, N.; Kim, D.W.; Yoon, Y.S. Malignant tumor formation after transplantation of short-term cultured bone marrow mesenchymal stem cells in experimental myocardial infarction and diabetic neuropathy. Circ. Res. 2011, 108, 1340–1347. [Google Scholar] [CrossRef] [PubMed]
- Braid, L.R.; Wood, C.A.; Wiese, D.M.; Ford, B.N. Intramuscular administration potentiates extended dwell time of mesenchymal stromal cells compared to other routes. Cytotherapy 2018, 20, 232–244. [Google Scholar] [CrossRef] [PubMed]
- Jung, J.W.; Kwon, M.; Choi, J.C.; Shin, J.W.; Park, I.W.; Choi, B.W.; Kim, J.Y. Familial occurrence of pulmonary embolism after intravenous, adipose tissue-derived stem cell therapy. Yonsei Med. J. 2013, 54, 1293–1296. [Google Scholar] [CrossRef] [PubMed]
- Yong, K.W.; Wan Safwani, W.K.; Xu, F.; Wan Abas, W.A.; Choi, J.R.; Pingguan-Murphy, B. Cryopreservation of Human Mesenchymal Stem Cells for Clinical Applications: Current Methods and Challenges. Biopreserv. Biobank 2015, 13, 231–239. [Google Scholar] [CrossRef] [PubMed]
- Dehnavi, S.; Sadeghi, M.; Tavakol Afshari, J.; Mohammadi, M. Interactions of mesenchymal stromal/stem cells and immune cells following MSC-based therapeutic approaches in rheumatoid arthritis. Cell Immunol. 2023, 393–394, 104771. [Google Scholar] [CrossRef]
- Wolf, P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef]
- El Andaloussi, S.; Mager, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
- Ju, Y.; Bai, H.; Ren, L.; Zhang, L. The Role of Exosome and the ESCRT Pathway on Enveloped Virus Infection. Int. J. Mol. Sci. 2021, 22, 9060. [Google Scholar] [CrossRef]
- Yu, J.; Sane, S.; Kim, J.E.; Yun, S.; Kim, H.J.; Jo, K.B.; Wright, J.P.; Khoshdoozmasouleh, N.; Lee, K.; Oh, H.T.; et al. Biogenesis and delivery of extracellular vesicles: Harnessing the power of EVs for diagnostics and therapeutics. Front. Mol. Biosci. 2023, 10, 1330400. [Google Scholar] [CrossRef] [PubMed]
- Perissinotto, F.; Rondelli, V.; Senigagliesi, B.; Brocca, P.; Almasy, L.; Bottyan, L.; Merkel, D.G.; Amenitsch, H.; Sartori, B.; Pachler, K.; et al. Structural insights into fusion mechanisms of small extracellular vesicles with model plasma membranes. Nanoscale 2021, 13, 5224–5233. [Google Scholar] [CrossRef] [PubMed]
- Lamanuzzi, A.; Saltarella, I.; Reale, A.; Melaccio, A.; Solimando, A.G.; Altamura, C.; Tamma, G.; Storlazzi, C.T.; Tolomeo, D.; Desantis, V.; et al. Uptake-Dependent and -Independent Effects of Fibroblasts-Derived Extracellular Vesicles on Bone Marrow Endothelial Cells from Patients with Multiple Myeloma: Therapeutic and Clinical Implications. Biomedicines 2023, 11, 1400. [Google Scholar] [CrossRef]
- Xu, Y.P.; Jiang, T.; Yang, X.F.; Chen, Z.B. Methods, Mechanisms, and Application Prospects for Enhancing Extracellular Vesicle Uptake. Curr. Med. Sci. 2024, 44, 247–260. [Google Scholar] [CrossRef]
- Svensson, K.J.; Christianson, H.C.; Wittrup, A.; Bourseau-Guilmain, E.; Lindqvist, E.; Svensson, L.M.; Morgelin, M.; Belting, M. Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J. Biol. Chem. 2013, 288, 17713–17724. [Google Scholar] [CrossRef] [PubMed]
- Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef]
- Vora, A.; Zhou, W.; Londono-Renteria, B.; Woodson, M.; Sherman, M.B.; Colpitts, T.M.; Neelakanta, G.; Sultana, H. Arthropod EVs mediate dengue virus transmission through interaction with a tetraspanin domain containing glycoprotein Tsp29Fb. Proc. Natl. Acad. Sci. USA 2018, 115, E6604–E6613. [Google Scholar] [CrossRef]
- Chahar, H.S.; Bao, X.; Casola, A. Exosomes and Their Role in the Life Cycle and Pathogenesis of RNA Viruses. Viruses 2015, 7, 3204–3225. [Google Scholar] [CrossRef] [PubMed]
- Bukong, T.N.; Momen-Heravi, F.; Kodys, K.; Bala, S.; Szabo, G. Exosomes from hepatitis C infected patients transmit HCV infection and contain replication competent viral RNA in complex with Ago2-miR122-HSP90. PLoS Pathog. 2014, 10, e1004424. [Google Scholar] [CrossRef]
- Meckes, D.G., Jr. Exosomal communication goes viral. J. Virol. 2015, 89, 5200–5203. [Google Scholar] [CrossRef] [PubMed]
- Puzar Dominkus, P.; Ferdin, J.; Plemenitas, A.; Peterlin, B.M.; Lenassi, M. Nef is secreted in exosomes from Nef.GFP-expressing and HIV-1-infected human astrocytes. J. Neurovirol. 2017, 23, 713–724. [Google Scholar] [CrossRef] [PubMed]
- Booth, A.M.; Fang, Y.; Fallon, J.K.; Yang, J.M.; Hildreth, J.E.; Gould, S.J. Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. J. Cell Biol. 2006, 172, 923–935. [Google Scholar] [CrossRef]
- Chatterjee, S.; Kordbacheh, R.; Sin, J. Extracellular Vesicles: A Novel Mode of Viral Propagation Exploited by Enveloped and Non-Enveloped Viruses. Microorganisms 2024, 12, 274. [Google Scholar] [CrossRef]
- Vulpis, E.; Loconte, L.; Peri, A.; Molfetta, R.; Caracciolo, G.; Masuelli, L.; Tomaipitinca, L.; Peruzzi, G.; Petillo, S.; Petrucci, M.T.; et al. Impact on NK cell functions of acute versus chronic exposure to extracellular vesicle-associated MICA: Dual role in cancer immunosurveillance. J. Extracell. Vesicles 2022, 11, e12176. [Google Scholar] [CrossRef] [PubMed]
- Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer—Implications for future improvements in cancer care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef]
- Xue, V.W.; Wong, S.C.C.; Song, G.; Cho, W.C.S. Promising RNA-based cancer gene therapy using extracellular vesicles for drug delivery. Expert. Opin. Biol. Ther. 2020, 20, 767–777. [Google Scholar] [CrossRef]
- Hao, S.; Bai, O.; Li, F.; Yuan, J.; Laferte, S.; Xiang, J. Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology 2007, 120, 90–102. [Google Scholar] [CrossRef]
- Zech, D.; Rana, S.; Buchler, M.W.; Zoller, M. Tumor-exosomes and leukocyte activation: An ambivalent crosstalk. Cell Commun. Signal 2012, 10, 37. [Google Scholar] [CrossRef] [PubMed]
- Jalilian, E.; Massoumi, H.; Bigit, B.; Amin, S.; Katz, E.A.; Guaiquil, V.H.; Anwar, K.N.; Hematti, P.; Rosenblatt, M.I.; Djalilian, A.R. Bone marrow mesenchymal stromal cells in a 3D system produce higher concentration of extracellular vesicles (EVs) with increased complexity and enhanced neuronal growth properties. Stem Cell Res. Ther. 2022, 13, 425. [Google Scholar] [CrossRef]
- Gorgun, C.; Ceresa, D.; Lesage, R.; Villa, F.; Reverberi, D.; Balbi, C.; Santamaria, S.; Cortese, K.; Malatesta, P.; Geris, L.; et al. Dissecting the effects of preconditioning with inflammatory cytokines and hypoxia on the angiogenic potential of mesenchymal stromal cell (MSC)-derived soluble proteins and extracellular vesicles (EVs). Biomaterials 2021, 269, 120633. [Google Scholar] [CrossRef] [PubMed]
- Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, A.G.; Cheng, K.; Marban, E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef] [PubMed]
- Lai, R.C.; Yeo, R.W.; Lim, S.K. Mesenchymal stem cell exosomes. Semin. Cell Dev. Biol. 2015, 40, 82–88. [Google Scholar] [CrossRef] [PubMed]
- Han, C.; Sun, X.; Liu, L.; Jiang, H.; Shen, Y.; Xu, X.; Li, J.; Zhang, G.; Huang, J.; Lin, Z.; et al. Exosomes and Their Therapeutic Potentials of Stem Cells. Stem Cells Int. 2016, 2016, 7653489. [Google Scholar] [CrossRef]
- Lai, P.; Weng, J.; Guo, L.; Chen, X.; Du, X. Novel insights into MSC-EVs therapy for immune diseases. Biomark. Res. 2019, 7, 6. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M.; Liu, S.; Wang, C.; Wang, Y.; Wan, M.; Liu, F.; Gong, M.; Yuan, Y.; Chen, Y.; Cheng, J.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA. ACS Nano 2021, 15, 1519–1538. [Google Scholar] [CrossRef] [PubMed]
- Lu, T.; Zhang, J.; Cai, J.; Xiao, J.; Sui, X.; Yuan, X.; Li, R.; Li, Y.; Yao, J.; Lv, G.; et al. Extracellular vesicles derived from mesenchymal stromal cells as nanotherapeutics for liver ischaemia-reperfusion injury by transferring mitochondria to modulate the formation of neutrophil extracellular traps. Biomaterials 2022, 284, 121486. [Google Scholar] [CrossRef]
- Kim, H.; Lee, M.J.; Bae, E.H.; Ryu, J.S.; Kaur, G.; Kim, H.J.; Kim, J.Y.; Barreda, H.; Jung, S.Y.; Choi, J.M.; et al. Comprehensive Molecular Profiles of Functionally Effective MSC-Derived Extracellular Vesicles in Immunomodulation. Mol. Ther. 2020, 28, 1628–1644. [Google Scholar] [CrossRef] [PubMed]
- Tan, Y.; Cai, J.; Wang, Z. Epsilon-caprolactone-modified polyethylenimine as a genetic vehicle for stem cell-based bispecific antibody and exosome synergistic therapy. Regen. Biomater. 2023, 10, rbac090. [Google Scholar] [CrossRef]
- Zhang, L.; Xiang, J.; Zhang, F.; Liu, L.; Hu, C. MSCs can be a double-edged sword in tumorigenesis. Front. Oncol. 2022, 12, 1047907. [Google Scholar] [CrossRef] [PubMed]
- Shamshiripour, P.; Rahnama, M.; Nikoobakht, M.; Hajiahmadi, F.; Moradi, A.R.; Ahmadvand, D. A dynamic study of VEGF-A siDOX-EVs trafficking through the in-vitro insert co-culture blood-brain barrier model by digital holographic microscopy. Front. Oncol. 2024, 14, 1292083. [Google Scholar] [CrossRef] [PubMed]
- Rufino-Ramos, D.; Albuquerque, P.R.; Carmona, V.; Perfeito, R.; Nobre, R.J.; Pereira de Almeida, L. Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. J. Control Release 2017, 262, 247–258. [Google Scholar] [CrossRef] [PubMed]
- Padinharayil, H.; Varghese, J.; Wilson, C.; George, A. Mesenchymal stem cell-derived exosomes: Characteristics and applications in disease pathology and management. Life Sci. 2024, 342, 122542. [Google Scholar] [CrossRef] [PubMed]
- Firouzabadi, S.R.; Mohammadi, I.; Ghafourian, K.; Kiani, A.; Hashemi, S.M. Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Asthma in Murine Models: A Systematic Review and Meta-analysis. Stem Cell Rev. Rep. 2024, 20, 1162–1183. [Google Scholar] [CrossRef]
- Zou, J.; Yang, W.; Cui, W.; Li, C.; Ma, C.; Ji, X.; Hong, J.; Qu, Z.; Chen, J.; Liu, A.; et al. Therapeutic potential and mechanisms of mesenchymal stem cell-derived exosomes as bioactive materials in tendon-bone healing. J. Nanobiotechnol. 2023, 21, 14. [Google Scholar] [CrossRef] [PubMed]
- Zhao, S.; Qi, W.; Zheng, J.; Tian, Y.; Qi, X.; Kong, D.; Zhang, J.; Huang, X. Exosomes Derived from Adipose Mesenchymal Stem Cells Restore Functional Endometrium in a Rat Model of Intrauterine Adhesions. Reprod. Sci. 2020, 27, 1266–1275. [Google Scholar] [CrossRef]
- Cheng, J.; Sun, Y.; Ma, Y.; Ao, Y.; Hu, X.; Meng, Q. Engineering of MSC-Derived Exosomes: A Promising Cell-Free Therapy for Osteoarthritis. Membranes 2022, 12, 739. [Google Scholar] [CrossRef] [PubMed]
- Gao, S.; Ogawa, M.; Takami, A.; Takeshita, K.; Kato, H. Practical and Safe Method of Long-Term Cryopreservation for Clinical Application of Human Adipose-Derived Mesenchymal Stem Cells Without a Programmable Freezer Or Serum. Cryo Lett. 2020, 41, 337–343. [Google Scholar]
- Cheng, Y.; Zeng, Q.; Han, Q.; Xia, W. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell 2019, 10, 295–299. [Google Scholar] [CrossRef] [PubMed]
- Sotiropoulou, P.A.; Perez, S.A.; Salagianni, M.; Baxevanis, C.N.; Papamichail, M. Characterization of the optimal culture conditions for clinical scale production of human mesenchymal stem cells. Stem Cells 2006, 24, 462–471. [Google Scholar] [CrossRef]
- Toh, W.S.; Lai, R.C.; Zhang, B.; Lim, S.K. MSC exosome works through a protein-based mechanism of action. Biochem. Soc. Trans. 2018, 46, 843–853. [Google Scholar] [CrossRef] [PubMed]
- Fong, C.Y.; Chak, L.L.; Biswas, A.; Tan, J.H.; Gauthaman, K.; Chan, W.K.; Bongso, A. Human Wharton’s jelly stem cells have unique transcriptome profiles compared to human embryonic stem cells and other mesenchymal stem cells. Stem Cell Rev. Rep. 2011, 7, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Karahuseyinoglu, S.; Cinar, O.; Kilic, E.; Kara, F.; Akay, G.G.; Demiralp, D.O.; Tukun, A.; Uckan, D.; Can, A. Biology of stem cells in human umbilical cord stroma: In situ and in vitro surveys. Stem Cells 2007, 25, 319–331. [Google Scholar] [CrossRef]
- Bai, L.; Li, D.; Li, J.; Luo, Z.; Yu, S.; Cao, S.; Shen, L.; Zuo, Z.; Ma, X. Bioactive molecules derived from umbilical cord mesenchymal stem cells. Acta Histochem. 2016, 118, 761–769. [Google Scholar] [CrossRef] [PubMed]
- Yin, K.; Wang, S.; Zhao, R.C. Exosomes from mesenchymal stem/stromal cells: A new therapeutic paradigm. Biomark. Res. 2019, 7, 8. [Google Scholar] [CrossRef] [PubMed]
- Maqsood, M.; Kang, M.; Wu, X.; Chen, J.; Teng, L.; Qiu, L. Adult mesenchymal stem cells and their exosomes: Sources, characteristics, and application in regenerative medicine. Life Sci. 2020, 256, 118002. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Yang, X.; Wang, S.; Wu, Y.; Zheng, L.; Tang, Y.; Gao, Y.; Niu, J. Human umbilical cord mesenchymal stromal cell-derived exosomes protect against MCD-induced NASH in a mouse model. Stem Cell Res. Ther. 2022, 13, 517. [Google Scholar] [CrossRef]
- Kang, Y.; Song, Y.; Luo, Y.; Song, J.; Li, C.; Yang, S.; Guo, J.; Yu, J.; Zhang, X. Exosomes derived from human umbilical cord mesenchymal stem cells ameliorate experimental non-alcoholic steatohepatitis via Nrf2/NQO-1 pathway. Free Radic. Biol. Med. 2022, 192, 25–36. [Google Scholar] [CrossRef]
- Du, X.; Li, H.; Han, X.; Ma, W. Mesenchymal stem cells-derived exosomal miR-24-3p ameliorates non-alcohol fatty liver disease by targeting Keap-1. Biochem. Biophys. Res. Commun. 2022, 637, 331–340. [Google Scholar] [CrossRef] [PubMed]
- Yang, F.; Wu, Y.; Chen, Y.; Xi, J.; Chu, Y.; Jin, J.; Yan, Y. Human umbilical cord mesenchymal stem cell-derived exosomes ameliorate liver steatosis by promoting fatty acid oxidation and reducing fatty acid synthesis. JHEP Rep. 2023, 5, 100746. [Google Scholar] [CrossRef] [PubMed]
- Cheng, L.; Yu, P.; Li, F.; Jiang, X.; Jiao, X.; Shen, Y.; Lai, X. Human umbilical cord-derived mesenchymal stem cell-exosomal miR-627-5p ameliorates non-alcoholic fatty liver disease by repressing FTO expression. Hum. Cell 2021, 34, 1697–1708. [Google Scholar] [CrossRef] [PubMed]
- Prockop, D.J. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997, 276, 71–74. [Google Scholar] [CrossRef] [PubMed]
- Lin, H.; Sohn, J.; Shen, H.; Langhans, M.T.; Tuan, R.S. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials 2019, 203, 96–110. [Google Scholar] [CrossRef] [PubMed]
- Baberg, F.; Geyh, S.; Waldera-Lupa, D.; Stefanski, A.; Zilkens, C.; Haas, R.; Schroeder, T.; Stuhler, K. Secretome analysis of human bone marrow derived mesenchymal stromal cells. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 434–441. [Google Scholar] [CrossRef]
- Jarosz-Popek, J.; Wolska, M.; Gasecka, A.; Czajka, P.; Jakubik, D.; Sharif, L.; Adem, T.; Liu, W.L.; Mirowska-Guzel, D.; Postula, M.; et al. The Importance of Non-Coding RNAs in Neurodegenerative Processes of Diabetes-Related Molecular Pathways. J. Clin. Med. 2020, 10, 9. [Google Scholar] [CrossRef] [PubMed]
- Oskowitz, A.; McFerrin, H.; Gutschow, M.; Carter, M.L.; Pochampally, R. Serum-deprived human multipotent mesenchymal stromal cells (MSCs) are highly angiogenic. Stem Cell Res. 2011, 6, 215–225. [Google Scholar] [CrossRef]
- Katagiri, W.; Kawai, T.; Osugi, M.; Sugimura-Wakayama, Y.; Sakaguchi, K.; Kojima, T.; Kobayashi, T. Angiogenesis in newly regenerated bone by secretomes of human mesenchymal stem cells. Maxillofac. Plast. Reconstr. Surg. 2017, 39, 8. [Google Scholar] [CrossRef] [PubMed]
- El-Derany, M.O.; AbdelHamid, S.G. Upregulation of miR-96-5p by bone marrow mesenchymal stem cells and their exosomes alleviate non-alcoholic steatohepatitis: Emphasis on caspase-2 signaling inhibition. Biochem. Pharmacol. 2021, 190, 114624. [Google Scholar] [CrossRef] [PubMed]
- Satilmis, B.; Cicek, G.S.; Cicek, E.; Akbulut, S.; Sahin, T.T.; Yilmaz, S. Adipose-derived stem cells in the treatment of hepatobiliary diseases and sepsis. World J. Clin. Cases 2022, 10, 4348–4356. [Google Scholar] [CrossRef] [PubMed]
- Bond, S.T.; Calkin, A.C.; Drew, B.G. Adipose-Derived Extracellular Vesicles: Systemic Messengers and Metabolic Regulators in Health and Disease. Front. Physiol. 2022, 13, 837001. [Google Scholar] [CrossRef]
- Ni, J.; Li, H.; Zhou, Y.; Gu, B.; Xu, Y.; Fu, Q.; Peng, X.; Cao, N.; Fu, Q.; Jin, M.; et al. Therapeutic Potential of Human Adipose-Derived Stem Cell Exosomes in Stress Urinary Incontinence—An in Vitro and in Vivo Study. Cell Physiol. Biochem. 2018, 48, 1710–1722. [Google Scholar] [CrossRef] [PubMed]
- Niu, Q.; Wang, T.; Wang, Z.; Wang, F.; Huang, D.; Sun, H.; Liu, H. Adipose-derived mesenchymal stem cell-secreted extracellular vesicles alleviate non-alcoholic fatty liver disease via delivering miR-223-3p. Adipocyte 2022, 11, 572–587. [Google Scholar] [CrossRef]
- Zhang, Y.; Xu, N.; Xu, J.; Kong, B.; Copple, B.; Guo, G.L.; Wang, L. E2F1 is a novel fibrogenic gene that regulates cholestatic liver fibrosis through the Egr-1/SHP/EID1 network. Hepatology 2014, 60, 919–930. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Li, R.; Xu, F.; Zhu, F.; Xu, X. Dehydrovomifoliol Alleviates Nonalcoholic Fatty Liver Disease via the E2F1/AKT/mTOR Axis: Pharmacophore Modeling and Molecular Docking Study. Evid. Based Complement. Alternat. Med. 2023, 2023, 9107598. [Google Scholar] [CrossRef] [PubMed]
- Watanabe, T.; Tsuchiya, A.; Takeuchi, S.; Nojiri, S.; Yoshida, T.; Ogawa, M.; Itoh, M.; Takamura, M.; Suganami, T.; Ogawa, Y.; et al. Development of a non-alcoholic steatohepatitis model with rapid accumulation of fibrosis, and its treatment using mesenchymal stem cells and their small extracellular vesicles. Regen. Ther. 2020, 14, 252–261. [Google Scholar] [CrossRef] [PubMed]
- Jiang, X.; Yang, J.; Liu, F.; Tao, J.; Xu, J.; Zhang, M. Embryonic stem cell-derived mesenchymal stem cells alleviate skeletal muscle injury induced by acute compartment syndrome. Stem Cell Res. Ther. 2022, 13, 313. [Google Scholar] [CrossRef] [PubMed]
- Hawkins, K.E.; Corcelli, M.; Dowding, K.; Ranzoni, A.M.; Vlahova, F.; Hau, K.L.; Hunjan, A.; Peebles, D.; Gressens, P.; Hagberg, H.; et al. Embryonic Stem Cell-Derived Mesenchymal Stem Cells (MSCs) Have a Superior Neuroprotective Capacity Over Fetal MSCs in the Hypoxic-Ischemic Mouse Brain. Stem Cells Transl. Med. 2018, 7, 439–449. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Kimbrel, E.A.; Ijichi, K.; Paul, D.; Lazorchak, A.S.; Chu, J.; Kouris, N.A.; Yavanian, G.J.; Lu, S.J.; Pachter, J.S.; et al. Human ESC-Derived MSCs Outperform Bone Marrow MSCs in the Treatment of an EAE Model of Multiple Sclerosis. Stem Cell Rep. 2021, 16, 370–371. [Google Scholar] [CrossRef]
- Gonzalez-King, H.; Rodrigues, P.G.; Albery, T.; Tangruksa, B.; Gurrapu, R.; Silva, A.M.; Musa, G.; Kardasz, D.; Liu, K.; Kull, B.; et al. Head-to-head comparison of relevant cell sources of small extracellular vesicles for cardiac repair: Superiority of embryonic stem cells. J. Extracell. Vesicles 2024, 13, e12445. [Google Scholar] [CrossRef]
- Yeo, R.W.; Lai, R.C.; Zhang, B.; Tan, S.S.; Yin, Y.; Teh, B.J.; Lim, S.K. Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Adv. Drug Deliv. Rev. 2013, 65, 336–341. [Google Scholar] [CrossRef]
- Tawfeek, G.A.; Kasem, H.A. Curcumin preconditioned mesenchymal stem cells derived exosomes transplantation ameliorate and protect against non- alcoholic steatohepatitis by regulation the expression of key genes of inflammation and oxidative stress. Transpl. Immunol. 2023, 78, 101837. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Lee, S.K.; Jeong, S.Y.; Cho, H.J.; Park, J.; Kim, T.M.; Kim, S. Cargo proteins in extracellular vesicles: Potential for novel therapeutics in non-alcoholic steatohepatitis. J. Nanobiotechnol. 2021, 19, 372. [Google Scholar] [CrossRef] [PubMed]
- Ma, L.; Wei, J.; Zeng, Y.; Liu, J.; Xiao, E.; Kang, Y.; Kang, Y. Mesenchymal stem cell-originated exosomal circDIDO1 suppresses hepatic stellate cell activation by miR-141-3p/PTEN/AKT pathway in human liver fibrosis. Drug Deliv. 2022, 29, 440–453. [Google Scholar] [CrossRef]
- Kim, J.; Lee, C.; Shin, Y.; Wang, S.; Han, J.; Kim, M.; Kim, J.M.; Shin, S.C.; Lee, B.J.; Kim, T.J.; et al. sEVs from tonsil-derived mesenchymal stromal cells alleviate activation of hepatic stellate cells and liver fibrosis through miR-486-5p. Mol. Ther. 2021, 29, 1471–1486. [Google Scholar] [CrossRef] [PubMed]
- Lou, G.; Yang, Y.; Liu, F.; Ye, B.; Chen, Z.; Zheng, M.; Liu, Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J. Cell Mol. Med. 2017, 21, 2963–2973. [Google Scholar] [CrossRef] [PubMed]
- Cheng, F.; Yang, F.; Wang, Y.; Zhou, J.; Qian, H.; Yan, Y. Mesenchymal stem cell-derived exosomal miR-27b-3p alleviates liver fibrosis via downregulating YAP/LOXL2 pathway. J. Nanobiotechnol. 2023, 21, 195. [Google Scholar] [CrossRef]
- Tan, Y.; Huang, Y.; Mei, R.; Mao, F.; Yang, D.; Liu, J.; Xu, W.; Qian, H.; Yan, Y. HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death Dis. 2022, 13, 319. [Google Scholar] [CrossRef]
- Zhao, P.; Dang, Z.; Liu, M.; Guo, D.; Luo, R.; Zhang, M.; Xie, F.; Zhang, X.; Wang, Y.; Pan, S.; et al. Molecular hydrogen promotes wound healing by inducing early epidermal stem cell proliferation and extracellular matrix deposition. Inflamm. Regen. 2023, 43, 22. [Google Scholar] [CrossRef] [PubMed]
- Gronemeyer, P.; Ditz, R.; Strube, J. Trends in Upstream and Downstream Process Development for Antibody Manufacturing. Bioengineering 2014, 1, 188–212. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Wang, H.X.; Zhu, X.J.; Wu, P.H.; Chen, W.Q.; Zou, P.; Li, Q.B.; Chen, Z.C. Serum deprivation elevates the levels of microvesicles with different size distributions and selectively enriched proteins in human myeloma cells in vitro. Acta Pharmacol. Sin. 2014, 35, 381–393. [Google Scholar] [CrossRef] [PubMed]
- King, H.W.; Michael, M.Z.; Gleadle, J.M. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 2012, 12, 421. [Google Scholar] [CrossRef] [PubMed]
- Piffoux, M.; Nicolas-Boluda, A.; Mulens-Arias, V.; Richard, S.; Rahmi, G.; Gazeau, F.; Wilhelm, C.; Silva, A.K.A. Extracellular vesicles for personalized medicine: The input of physically triggered production, loading and theranostic properties. Adv. Drug Deliv. Rev. 2019, 138, 247–258. [Google Scholar] [CrossRef] [PubMed]
- Momen-Heravi, F.; Bala, S.; Kodys, K.; Szabo, G. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Sci. Rep. 2015, 5, 9991. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Lee, Y.; Johansson, H.J.; Mager, I.; Vader, P.; Nordin, J.Z.; Wiklander, O.P.; Lehtio, J.; Wood, M.J.; Andaloussi, S.E. Serum-free culture alters the quantity and protein composition of neuroblastoma-derived extracellular vesicles. J. Extracell. Vesicles 2015, 4, 26883. [Google Scholar] [CrossRef] [PubMed]
Type of MSCs | Species and Strain of Recipient | Liver Injury Model | Effect After MSC-EVs Treatment | EV Dose, Timing Duration, Route | Ref |
---|---|---|---|---|---|
Human umbilical cord-MSC | Mouse/C57BL/6 J | Methionine-choline-deficient (MCD) diet, for 6 weeks | Decreased ALT and AST; reduced TNF-a, IL-6, and IL-1b in plasma Decreased level of NF-kB protein and p-NF-kB; increased F4/80 positive cells; increased expression of CD206, arginase-1 and IL-10 in liver tissue | 20 mg/kg i.v. 1st week of MCD diet. EV injection once a week | [129] |
Human umbilical cord-MSC | Mouse/C57BL/6 J | 20-week high fat high cholesterol (HFHC) MCD diet for 4 weeks | Serum AST and ALT levels, reduced steatosis, reversal of lipid-related gene expression (SREBP-1c, PPARα, Fabp5, CPT1α, ACOX, FAS), reduced frequency of F4/80 macrophages de-polarized macrophages in the liver | HFHC diet: 100 µg i.v. every 3 days for the last 6 weeks MCD diet: 100 µg i.v. twice a week for the last 2 weeks | [130] |
Human umbilical cord-MSC | Mouse/C57BL/6 J | 16-week HFD | Corrected body weights, liver weights, blood glucose and insulin, and serum AST or ALT, reduced HFD-mediated steatosis, hepatocyte ballooning, macrophage inflammation, reduced production or expression of ROS, TNFα, IL-6, ACCα, FASN, SCD1, PPARγ, NOX2, NOX4, TNF-α, IL-6, IL-1β and CCL2, and restored levels of antioxidant SOD and CAT | 120 µg i.v. weekly | [131] |
Human umbilical cord-MSs | Mouse/C57BL/6 J | 14-week HFD | Reduced levels of steatosis, AST, ALT, liver triglycerides, total cholesterol, hepatic expression of TNF-α, IL-1, and IL-6, and restored insulin sensitivity | 10 µg/g i.v. per week over the last 4 weeks | [132] |
Human umbilical cord-MSC | Sprague Dawley (SD) rat | High fat high fructose for 8 weeks | Reduction in diet-induced body and liver weights, steatosis, insulin tolerance, liver injury | 100 µg i.v. per rat | [133] |
Rats Bone marrow MSCs | Sprague Dawley (SD) rat | HFD for 12 weeks | Suppressed steatosis, reduced hepatocyte ballooning, AST, ALT, and expression of SREBP-1, SREBP-2, ACC, CD36, decreased NAS, increased expression of PPARα and CPT1. Suppression of caspase 2, apoptosis, mitophagy | 15, 30, 120 μg/kg i.v. Twice a week starting on week 7 for 6 weeks | [140] |
Human Adipose-derived MSCs | Mouse/C57BL/6 J | HFD for 8 weeks | Reduced HFD-induced ALT, AST, hepatic lipid, hydroxyproline, fibrosis | 100 μg i.v. twice a week starting the 2nd week of HFD diet | [144] |
Human adipose tissue-MSCs | Mouse/C57BL/6 J (Mc4r-KO) | Western diet for 20 weeks with 0.3 mg/kg LPS twice a week for 4 weeks | Decreased serum ALT, crown-like structures, and fibrosis. Increased frequency of anti-inflammatory macrophages, increased expression of Mmp12, Mmp13 | 1, 2.5, 5 μg i.p. once 4 weeks prior | [147] |
Embryonic stem cell-derived MSCs | Mouse/C57BL/6 J | 200 μg Streptozotocin (STZ) 2 days after birth, HFD from 4 weeks old for 5 weeks | Reduced steatosis, hepatocyte ballooning, inflammation, fibrosis. Increased hepatic frequency of CD163+ M2 macrophages, reduced plasma IL-6 | 1 μg, 10 μg i.p. every other day | [34] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zou, X.; Brigstock, D. Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Biomedicines 2024, 12, 2848. https://doi.org/10.3390/biomedicines12122848
Zou X, Brigstock D. Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Biomedicines. 2024; 12(12):2848. https://doi.org/10.3390/biomedicines12122848
Chicago/Turabian StyleZou, Xue, and David Brigstock. 2024. "Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD)" Biomedicines 12, no. 12: 2848. https://doi.org/10.3390/biomedicines12122848
APA StyleZou, X., & Brigstock, D. (2024). Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Biomedicines, 12(12), 2848. https://doi.org/10.3390/biomedicines12122848