Mesenchymal Stem Cell-Derived Extracellular Vesicles as Idiopathic Pulmonary Fibrosis Microenvironment Targeted Delivery
Abstract
:1. Introduction
2. Characteristics of EVs
3. Microenvironment in IPF
3.1. Inflammatory Microenvironment in IPF
3.2. Immune Dysregulation in IPF
3.2.1. Innate Immune Response
3.2.2. Adaptive Immune Response
3.3. ECM Microenvironment in IPF
4. MSC-EVs Mediated Homeostasis on IPF
4.1. MSC-EVs Plays an Anti-Inflammatory Role in IPF
4.2. MSC-EVs Can Modulate the Immune Response in IPF
4.3. MSC-EVs Regulate ECM Homeostasis in IPF
5. MSC-EVs Treatment Administration for IPF
5.1. Systemic Delivery of MSC-EVs
5.2. Direct Delivery of MSC-EVs to Lung
5.3. MSC-EVs as a Targeted Delivery Vector for IPF Treatment
6. Challenges for Application
7. Conclusions and Prospects
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Shenderov, K.; Collins, S.L.; Powell, J.D.; Horton, M.R. Immune dysregulation as a driver of idiopathic pulmonary fibrosis. J. Clin. Investig. 2021, 131, e143226. [Google Scholar] [CrossRef]
- Richeldi, L.; Collard, H.R.; Jones, M.G. Idiopathic pulmonary fibrosis. Lancet 2017, 389, 1941–1952. [Google Scholar] [CrossRef]
- Betensley, A.; Sharif, R.; Karamichos, D. A Systematic Review of the Role of Dysfunctional Wound Healing in the Pathogenesis and Treatment of Idiopathic Pulmonary Fibrosis. J. Clin. Med. 2016, 6, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raghu, G.; Collard, H.R.; Egan, J.J.; Martinez, F.J.; Behr, J.; Brown, K.K.; Colby, T.V.; Cordier, J.F.; Flaherty, K.R.; Lasky, J.A.; et al. An official ATS/ERS/JRS/ALAT statement: Idiopathic pulmonary fibrosis: Evidence-based guidelines for diagnosis and management. Am. J. Respir. Crit. Care Med. 2011, 183, 788–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seifirad, S. Pirfenidone: A novel hypothetical treatment for COVID-19. Med. Hypotheses 2020, 144, 110005. [Google Scholar] [CrossRef]
- Lin, X.; Yu, M.; Wu, K.; Yuan, H.; Zhong, H. Effects of pirfenidone on proliferation, migration, and collagen contraction of human Tenon’s fibroblasts in vitro. Investig. Ophthalmol. Vis. Sci. 2009, 50, 3763–3770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shihab, F.S.; Bennett, W.M.; Yi, H.; Andoh, T.F. Pirfenidone Treatment Decreases Transforming Growth Factor-β1 and Matrix Proteins and Ameliorates Fibrosis in Chronic Cyclosporine Nephrotoxicity. Am. J. Transplant. 2002, 2, 111–119. [Google Scholar] [CrossRef] [PubMed]
- Richeldi, L.; du Bois, R.M.; Raghu, G.; Azuma, A.; Brown, K.K.; Costabel, U.; Cottin, V.; Flaherty, K.R.; Hansell, D.M.; Inoue, Y.; et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N. Engl. J. Med. 2014, 370, 2071–2082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lancaster, L.H.; de Andrade, J.A.; Zibrak, J.D.; Padilla, M.L.; Albera, C.; Nathan, S.D.; Wijsenbeek, M.S.; Stauffer, J.L.; Kirchgaessler, K.U.; Costabel, U. Pirfenidone safety and adverse event management in idiopathic pulmonary fibrosis. Eur. Respir. Rev. 2017, 26, 170057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wynn, T.A. Integrating mechanisms of pulmonary fibrosis. J. Exp. Med. 2011, 208, 1339–1350. [Google Scholar] [CrossRef] [Green Version]
- Fernandez, I.E.; Eickelberg, O. New cellular and molecular mechanisms of lung injury and fibrosis in idiopathic pulmonary fibrosis. Lancet 2012, 380, 680–688. [Google Scholar] [CrossRef]
- Greenburg, G.; Hay, E.D. Cytodifferentiation and Tissue Phenotype Change during Transformation__of Embryonic Lens Epithelium to Mesenchyme-like Cells in vitro. Dev. Biol. 1986, 115, 363–379. [Google Scholar] [CrossRef]
- Zhao, Y.L.; Zhu, R.T.; Sun, Y.L. Epithelial-mesenchymal transition in liver fibrosis. Biomed. Rep. 2016, 4, 269–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rout-Pitt, N.; Farrow, N.; Parsons, D.; Donnelley, M. Epithelial mesenchymal transition (EMT): A universal process in lung diseases with implications for cystic fibrosis pathophysiology. Respir. Res. 2018, 19, 136. [Google Scholar] [CrossRef] [PubMed]
- Peng, L.; Wen, L.; Shi, Q.F.; Gao, F.; Huang, B.; Meng, J.; Hu, C.P.; Wang, C.M. Scutellarin ameliorates pulmonary fibrosis through inhibiting NF-kappaB/NLRP3-mediated epithelial-mesenchymal transition and inflammation. Cell Death Dis. 2020, 11, 978. [Google Scholar] [CrossRef] [PubMed]
- Fingerlin, T.E.; Murphy, E.; Zhang, W.; Peljto, A.L.; Brown, K.K.; Steele, M.P.; Loyd, J.E.; Cosgrove, G.P.; Lynch, D.; Groshong, S.; et al. Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis. Nat. Genet. 2013, 45, 613–620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mathai, S.K.; Newton, C.A.; Schwartz, D.A.; Garcia, C.K. Pulmonary fibrosis in the era of stratified medicine. Thorax 2016, 71, 1154–1160. [Google Scholar] [CrossRef] [Green Version]
- Mushiroda, T.; Wattanapokayakit, S.; Takahashi, A.; Nukiwa, T.; Kudoh, S.; Ogura, T.; Taniguchi, H.; Kubo, M.; Kamatani, N.; Nakamura, Y.; et al. A genome-wide association study identifies an association of a common variant in TERT with susceptibility to idiopathic pulmonary fibrosis. J. Med. Genet. 2008, 45, 654–656. [Google Scholar] [CrossRef]
- Kadota, T.; Fujita, Y.; Araya, J.; Watanabe, N.; Fujimoto, S.; Kawamoto, H.; Minagawa, S.; Hara, H.; Ohtsuka, T.; Yamamoto, Y.; et al. Human bronchial epithelial cell-derived extracellular vesicle therapy for pulmonary fibrosis via inhibition of TGF-beta-WNT crosstalk. J. Extracell. Vesicles 2021, 10, e12124. [Google Scholar] [CrossRef]
- Xiao, K.; He, W.; Guan, W.; Hou, F.; Yan, P.; Xu, J.; Zhou, T.; Liu, Y.; Xie, L. Mesenchymal stem cells reverse EMT process through blocking the activation of NF-kappaB and Hedgehog pathways in LPS-induced acute lung injury. Cell Death Dis. 2020, 11, 863. [Google Scholar] [CrossRef]
- Rana, T.; Jiang, C.; Liu, G.; Miyata, T.; Antony, V.; Thannickal, V.J.; Liu, R.-M. PAI-1 Regulation of TGF-β1-induced Alveolar Type II Cell Senescence, SASP Secretion, and SASP-mediated Activation of Alveolar Macrophages. Am. J. Respir. Cell Mol. Biol. 2020, 62, 319–330. [Google Scholar] [CrossRef] [PubMed]
- Yao, C.; Guan, X.; Carraro, G.; Parimon, T.; Liu, X.; Huang, G.; Soukiasian, H.J.; David, G.; Weigt, S.S.; Belperio, J.A.; et al. Senescence of Alveolar Type 2 Cells Drives Progressive Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2019, 203, 707–717. [Google Scholar] [CrossRef] [PubMed]
- O’Dwyer, D.N.; Ashley, S.L.; Gurczynski, S.J.; Xia, M.; Wilke, C.; Falkowski, N.R.; Norman, K.C.; Arnold, K.B.; Huffnagle, G.B.; Salisbury, M.L.; et al. Lung Microbiota Contribute to Pulmonary Inflammation and Disease Progression in Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2019, 199, 1127–1138. [Google Scholar] [CrossRef] [PubMed]
- Organ, S.L.; Tsao, M.-S. An overview of the c MET signaling pathway. Ther. Adv. Med. Oncol. 2011, 3, S7–S19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tzouvelekis, A.; Gomatou, G.; Bouros, E.; Trigidou, R.; Tzilas, V.; Bouros, D. Common Pathogenic Mechanisms Between Idiopathic Pulmonary Fibrosis and Lung Cancer. Chest 2019, 156, 383–391. [Google Scholar] [CrossRef] [PubMed]
- Tsoyi, K.; Liang, X.; De Rossi, G.; Ryter, S.W.; Xiong, K.; Chu, S.G.; Liu, X.; Ith, B.; Celada, L.J.; Romero, F.; et al. CD148 Deficiency in Fibroblasts Promotes the Development of Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2021, 204, 312–325. [Google Scholar] [CrossRef] [PubMed]
- Conte, E.; Gili, E.; Fruciano, M.; Korfei, M.; Fagone, E.; Iemmolo, M.; Lo Furno, D.; Giuffrida, R.; Crimi, N.; Guenther, A.; et al. PI3K p110gamma overexpression in idiopathic pulmonary fibrosis lung tissue and fibroblast cells: In vitro effects of its inhibition. Lab. Investig. 2013, 93, 566–576. [Google Scholar] [CrossRef]
- Wei, X.; Han, J.; Chen, Z.Z.; Qi, B.W.; Wang, G.C.; Ma, Y.H.; Zheng, H.; Luo, Y.F.; Wei, Y.Q.; Chen, L.J. A phosphoinositide 3-kinase-gamma inhibitor, AS605240 prevents bleomycin-induced pulmonary fibrosis in rats. Biochem. Biophys. Res. Commun. 2010, 397, 311–317. [Google Scholar] [CrossRef]
- Kim, K.K.; Sheppard, D.; Chapman, H.A. TGF-β1 Signaling and Tissue Fibrosis. Cold Spring Harb. Perspect. Biol. 2018, 10, a022293. [Google Scholar] [CrossRef] [Green Version]
- Averyanov, A.; Koroleva, I.; Konoplyannikov, M.; Revkova, V.; Lesnyak, V.; Kalsin, V.; Danilevskaya, O.; Nikitin, A.; Sotnikova, A.; Kotova, S.; et al. First-in-human high-cumulative-dose stem cell therapy in idiopathic pulmonary fibrosis with rapid lung function decline. Stem Cells Transl. Med. 2020, 9, 6–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Periera-Simon, S.; Xia, X.; Catanuto, P.; Coronado, R.; Kurtzberg, J.; Bellio, M.; Lee, Y.S.; Khan, A.; Smith, R.; Elliot, S.J.; et al. Anti-fibrotic effects of different sources of MSC in bleomycin-induced lung fibrosis in C57BL6 male mice. Respirology 2021, 26, 161–170. [Google Scholar] [CrossRef] [PubMed]
- Gnecchi, M.; Danieli, P.; Malpasso, G.; Ciuffreda, M.C. Paracrine Mechanisms of Mesenchymal Stem Cells in Tissue Repair. Methods Mol. Biol. 2016, 1416, 123–146. [Google Scholar] [PubMed]
- Liu, H.; Liu, S.; Qiu, X.; Yang, X.; Bao, L.; Pu, F.; Liu, X.; Li, C.; Xuan, K.; Zhou, J.; et al. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy 2020, 16, 2140–2155. [Google Scholar] [CrossRef]
- Barczyk, M.; Schmidt, M.; Mattoli, S. Stem Cell-Based Therapy in Idiopathic Pulmonary Fibrosis. Stem Cell Rev. Rep. 2015, 11, 598–620. [Google Scholar] [CrossRef] [PubMed]
- Caplan, A.I.; Dennis, J.E. Mesenchymal stem cells as trophic mediators. J. Cell Biochem. 2006, 98, 1076–1084. [Google Scholar] [CrossRef] [PubMed]
- Weng, Z.; Zhang, B.; Wu, C.; Yu, F.; Han, B.; Li, B.; Li, L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J. Hematol. Oncol. 2021, 14, 136. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Li, R.; Liu, T.; Yang, L.; Yin, G.; Xie, Q. Immunomodulatory Effects of Mesenchymal Stem Cells and Mesenchymal Stem Cell-Derived Extracellular Vesicles in Rheumatoid Arthritis. Front. Immunol. 2020, 11, 1912. [Google Scholar] [CrossRef] [PubMed]
- Vader, P.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol. Med. 2014, 20, 385–393. [Google Scholar] [CrossRef] [Green Version]
- Chen, P.; Wang, L.; Fan, X.; Ning, X.; Yu, B.; Ou, C.; Chen, M. Targeted delivery of extracellular vesicles in heart injury. Theranostics 2021, 11, 2263–2277. [Google Scholar] [CrossRef] [PubMed]
- Simon, T.; Jackson, E.; Giamas, G. Breaking through the glioblastoma micro-environment via extracellular vesicles. Oncogene 2020, 39, 4477–4490. [Google Scholar] [CrossRef]
- 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]
- Xu, R.; Greening, D.W.; Zhu, H.J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crivelli, B.; Chlapanidas, T.; Perteghella, S.; Lucarelli, E.; Pascucci, L.; Brini, A.T.; Ferrero, I.; Marazzi, M.; Pessina, A.; Torre, M.L.; et al. Mesenchymal stem/stromal cell extracellular vesicles: From active principle to next generation drug delivery system. J. Control. Release 2017, 262, 104–117. [Google Scholar] [CrossRef] [PubMed]
- Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 2008, 10, 619–624. [Google Scholar] [CrossRef] [PubMed]
- Luzio, J.P.; Pryor, P.R.; Bright, N.A. Lysosomes: Fusion and function. Nat. Rev. Mol. Cell Biol. 2007, 8, 622–632. [Google Scholar] [CrossRef]
- Henderson, N.C.; Rieder, F.; Wynn, T.A. Fibrosis: From mechanisms to medicines. Nature 2020, 587, 555–566. [Google Scholar] [CrossRef]
- Parra, E.R.; Kairalla, R.A.; Ribeiro de Carvalho, C.R.; Eher, E.; Capelozzi, V.L. Inflammatory Cell Phenotyping of the Pulmonary Interstitium in Idiopathic Interstitial Pneumonia. Respiration 2007, 74, 159–169. [Google Scholar] [CrossRef]
- Kapanci, Y.; Desmoulière, A.; Pache, J.C.; Redard, M.; Gabbiani, G. Cytoskeletal protein modulation in pulmonary alveolar myofibroblasts during idiopathic pulmonary fibrosis. Possible role of transforming growth factor beta and tumor necrosis factor alpha. Am. J. Respir. Crit. Care Med. 1995, 152, 2163–2169. [Google Scholar] [CrossRef]
- Carré, P.C.; Mortenson, R.L.; King, T.E.; Noble, P.W.; Sable, C.L.; Riches, D.W. Increased expression of the interleukin-8 gene by alveolar macrophages in idiopathic pulmonary fibrosis. A potential mechanism for the recruitment and activation of neutrophils in lung fibrosis. J. Clin. Investig. 1991, 88, 1802–1810. [Google Scholar] [CrossRef]
- Shaykhiev, R.; Otaki, F.; Bonsu, P.; Dang, D.T.; Teater, M.; Strulovici-Barel, Y.; Salit, J.; Harvey, B.G.; Crystal, R.G. Cigarette smoking reprograms apical junctional complex molecular architecture in the human airway epithelium in vivo. Cell Mol. Life Sci. 2011, 68, 877–892. [Google Scholar] [CrossRef] [Green Version]
- Barkauskas, C.E.; Cronce, M.J.; Rackley, C.R.; Bowie, E.J.; Keene, D.R.; Stripp, B.R.; Randell, S.H.; Noble, P.W.; Hogan, B.L. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Investig. 2013, 123, 3025–3036. [Google Scholar] [CrossRef] [PubMed]
- Murray, L.A.; Chen, Q.; Kramer, M.S.; Hesson, D.P.; Argentieri, R.L.; Peng, X.; Gulati, M.; Homer, R.J.; Russell, T.; van Rooijen, N.; et al. TGF-beta driven lung fibrosis is macrophage dependent and blocked by Serum amyloid P. Int. J. Biochem. Cell Biol. 2011, 43, 154–162. [Google Scholar] [CrossRef] [PubMed]
- Khalil, N.; Parekh, T.V.; O’Connor, R.N.; Gold, L.I. Differential expression of transforming growth factor-beta type I and II receptors by pulmonary cells in bleomycin-induced lung injury: Correlation with repair and fibrosis. Exp. Lung Res. 2002, 28, 233–250. [Google Scholar] [CrossRef] [PubMed]
- Demayo, F.; Minoo, P.; Plopper, C.G.; Schuger, L.; Shannon, J.; Torday, J.S. Mesenchymal-epithelial interactions in lung development and repair: Are modeling and remodeling the same process? Am. J. Physiol. Lung Cell Mol. Physiol. 2002, 283, L510–L517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maniatis, N.A.; Orfanos, S.E. The endothelium in acute lung injury/acute respiratory distress syndrome. Curr. Opin. Crit. Care 2008, 14, 22–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noth, I.; Zhang, Y.; Ma, S.-F.; Flores, C.; Barber, M.; Huang, Y.; Broderick, S.M.; Wade, M.S.; Hysi, P.; Scuirba, J.; et al. Genetic variants associated with idiopathic pulmonary fibrosis susceptibility and mortality: A genome-wide association study. Lancet Respir. Med. 2013, 1, 309–317. [Google Scholar] [CrossRef] [Green Version]
- Kondoh, Y.; Taniguchi, H.; Yokoi, T.; Nishiyama, O.; Ohishi, T.; Kato, T.; Suzuki, K.; Suzuki, R. Cyclophosphamide and low-dose prednisolone in idiopathic pulmonary fibrosis and fibrosing nonspecific interstitial pneumonia. Eur. Respir. J. 2005, 25, 528–533. [Google Scholar] [CrossRef]
- Lee, S.H.; Park, M.S.; Kim, S.Y.; Kim, D.S.; Kim, Y.W.; Chung, M.P.; Uh, S.T.; Park, C.S.; Park, S.W.; Jeong, S.H.; et al. Factors affecting treatment outcome in patients with idiopathic nonspecific interstitial pneumonia: A nationwide cohort study. Respir. Res. 2017, 18, 204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raghu, G. Idiopathic pulmonary fibrosis: Lessons from clinical trials over the past 25 years. Eur. Respir. J. 2017, 50, 1701209. [Google Scholar] [CrossRef] [Green Version]
- Raghu, G.; Brown, K.K.; Costabel, U.; Cottin, V.; du Bois, R.M.; Lasky, J.A.; Thomeer, M.; Utz, J.P.; Khandker, R.K.; McDermott, L.; et al. Treatment of idiopathic pulmonary fibrosis with etanercept: An exploratory, placebo-controlled trial. Am. J. Respir. Crit. Care Med. 2008, 178, 948–955. [Google Scholar] [CrossRef] [PubMed]
- Martinez, F.J.; de Andrade, J.A.; Anstrom, K.J.; King, T.E., Jr.; Raghu, G. Randomized trial of acetylcysteine in idiopathic pulmonary fibrosis. N. Engl. J. Med. 2014, 370, 2093–2101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parker, J.M.; Glaspole, I.N.; Lancaster, L.H.; Haddad, T.J.; She, D.; Roseti, S.L.; Fiening, J.P.; Grant, E.P.; Kell, C.M.; Flaherty, K.R. A Phase 2 Randomized Controlled Study of Tralokinumab in Subjects with Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2018, 197, 94–103. [Google Scholar] [CrossRef]
- Swigris, J.O.T.; Scholand, M.; Glaspole, I.; Maher, T.; Kardatzke, D.; Kaminski, J.; Castro, M.; Owens, R.; Neighbors, M.; Belloni, P. The RIFF study (cohort a): A phase II, randomized, double-blind, placebo-controlled trial of lebrikizumab as monotherapy in patients with idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2018, 197, A166. [Google Scholar]
- Wijsenbeek, M.S.; Kool, M.; Cottin, V. Targeting interleukin-13 in idiopathic pulmonary fibrosis: From promising path to dead end. Eur. Respir. J. 2018, 52, 1802111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Kuai, Q.; Gao, F.; Wang, Y.; He, M.; Zhou, H.; Han, G.; Jiang, X.; Ren, S.; Yu, Q. Overexpression of TIM-3 in Macrophages Aggravates Pathogenesis of Pulmonary Fibrosis in Mice. Am. J. Respir. Cell Mol. Biol. 2019, 61, 727–736. [Google Scholar] [CrossRef] [PubMed]
- Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef] [PubMed]
- Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef]
- Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mills, C.D. M1 and M2 Macrophages: Oracles of Health and Disease. Crit. Rev. Immunol. 2012, 32, 463–488. [Google Scholar] [CrossRef] [Green Version]
- Duru, N.; Wolfson, B.; Zhou, Q. Mechanisms of the alternative activation of macrophages and non-coding RNAs in the development of radiation-induced lung fibrosis. World J. Biol. Chem. 2016, 7, 231–239. [Google Scholar] [CrossRef]
- Murray, P.J. Macrophage Polarization. Annu. Rev. Physiol. 2017, 79, 541–566. [Google Scholar] [CrossRef] [PubMed]
- Chenivesse, C.; Tsicopoulos, A. CCL18—Beyond chemotaxis. Cytokine 2018, 109, 52–56. [Google Scholar] [CrossRef] [PubMed]
- Lech, M.; Anders, H.J. Macrophages and fibrosis: How resident and infiltrating mononuclear phagocytes orchestrate all phases of tissue injury and repair. Biochim. Biophys. Acta 2013, 1832, 989–997. [Google Scholar] [CrossRef] [Green Version]
- Marchal-Sommé, J.; Uzunhan, Y.; Marchand-Adam, S.; Valeyre, D.; Soumelis, V.; Crestani, B.; Soler, P. Cutting edge: Nonproliferating mature immune cells form a novel type of organized lymphoid structure in idiopathic pulmonary fibrosis. J. Immunol. 2006, 176, 5735–5739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nuovo, G.J.; Hagood, J.S.; Magro, C.M.; Chin, N.; Kapil, R.; Davis, L.; Marsh, C.B.; Folcik, V.A. The distribution of immunomodulatory cells in the lungs of patients with idiopathic pulmonary fibrosis. Mod. Pathol. 2011, 25, 416–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Todd, N.W.; Scheraga, R.G.; Galvin, J.R.; Iacono, A.T.; Britt, E.J.; Luzina, I.G.; Burke, A.P.; Atamas, S.P. Lymphocyte aggregates persist and accumulate in the lungs of patients with idiopathic pulmonary fibrosis. J. Inflamm. Res. 2013, 6, 63–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papiris, S.A.; Kollintza, A.; Kitsanta, P.; Kapotsis, G.; Karatza, M.; Milic-Emili, J.; Roussos, C.; Daniil, Z. Relationship of BAL and lung tissue CD4+ and CD8+ T lymphocytes, and their ratio in idiopathic pulmonary fibrosis. Chest 2005, 128, 2971–2977. [Google Scholar] [CrossRef]
- Schrier, D.J.; Phan, S.H.; McGarry, B.M. The effects of the nude (nu/nu) mutation on bleomycin-induced pulmonary fibrosis. A biochemical evaluation. Am. Rev. Respir. Dis. 1983, 127, 614–617. [Google Scholar] [CrossRef] [PubMed]
- Sandler, N.G.; Mentink-Kane, M.M.; Cheever, A.W.; Wynn, T.A. Global gene expression profiles during acute pathogen-induced pulmonary inflammation reveal divergent roles for Th1 and Th2 responses in tissue repair. J. Immunol. 2003, 171, 3655–3667. [Google Scholar] [CrossRef] [Green Version]
- Shao, D.D.; Suresh, R.; Vakil, V.; Gomer, R.H.; Pilling, D. Pivotal Advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J. Leukoc. Biol. 2008, 83, 1323–1333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wynn, T.A. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat. Rev. Immunol. 2004, 4, 583–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker, J.A.; McKenzie, A.N.J. TH2 cell development and.d function. Nat. Rev. Immunol. 2018, 18, 121–133. [Google Scholar] [CrossRef] [PubMed]
- King, T.E.; Albera, C.; Bradford, W.Z.; Costabel, U.; Hormel, P.; Lancaster, L.; Noble, P.W.; Sahn, S.A.; Szwarcberg, J.; Thomeer, M.; et al. Effect of interferon gamma-1b on survival in patients with idiopathic pulmonary fibrosis (INSPIRE): A multicentre, randomised, placebo-controlled trial. Lancet 2009, 374, 222–228. [Google Scholar] [CrossRef]
- Hashimoto-Kataoka, T.; Hosen, N.; Sonobe, T.; Arita, Y.; Yasui, T.; Masaki, T.; Minami, M.; Inagaki, T.; Miyagawa, S.; Sawa, Y.; et al. Interleukin-6/interleukin-21 signaling axis is critical in the pathogenesis of pulmonary arterial hypertension. Proc. Natl. Acad. Sci. USA 2015, 112, E2677–E2686. [Google Scholar] [CrossRef] [Green Version]
- Niu, W.; Xu, Y.; Zha, X.; Zeng, J.; Qiao, S.; Yang, S.; Zhang, H.; Tan, L.; Sun, L.; Pang, G.; et al. IL-21/IL-21R Signaling Aggravated Respiratory Inflammation Induced by Intracellular Bacteria through Regulation of CD4(+) T Cell Subset Responses. J. Immunol. 2021, 206, 1586–1596. [Google Scholar] [CrossRef]
- Chen, J.; Zhang, X.; Xie, J.; Xue, M.; Liu, L.; Yang, Y.; Qiu, H. Overexpression of TGFβ1 in murine mesenchymal stem cells improves lung inflammation by impacting the Th17/Treg balance in LPS-induced ARDS mice. Stem Cell Res. Ther. 2020, 11, 311. [Google Scholar] [CrossRef]
- Dong, Z.; Lu, X.; Yang, Y.; Zhang, T.; Li, Y.; Chai, Y.; Lei, W.; Li, C.; Ai, L.; Tai, W. IL-27 alleviates the bleomycin-induced pulmonary fibrosis by regulating the Th17 cell differentiation. BMC Pulm. Med. 2015, 15, 13. [Google Scholar] [CrossRef] [Green Version]
- Galati, D.; De Martino, M.; Trotta, A.; Rea, G.; Bruzzese, D.; Cicchitto, G.; Stanziola, A.A.; Napolitano, M.; Sanduzzi, A.; Bocchino, M. Peripheral depletion of NK cells and imbalance of the Treg/Th17 axis in idiopathic pulmonary fibrosis patients. Cytokine 2014, 66, 119–126. [Google Scholar] [CrossRef] [Green Version]
- Boveda-Ruiz, D.; D’Alessandro-Gabazza, C.N.; Toda, M.; Takagi, T.; Naito, M.; Matsushima, Y.; Matsumoto, T.; Kobayashi, T.; Gil-Bernabe, P.; Chelakkot-Govindalayathil, A.L.; et al. Differential role of regulatory T cells in early and late stages of pulmonary fibrosis. Immunobiology 2013, 218, 245–254. [Google Scholar] [CrossRef]
- Kotsianidis, I.; Nakou, E.; Bouchliou, I.; Tzouvelekis, A.; Spanoudakis, E.; Steiropoulos, P.; Sotiriou, I.; Aidinis, V.; Margaritis, D.; Tsatalas, C.; et al. Global impairment of CD4+CD25+FOXP3+ regulatory T cells in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2009, 179, 1121–1130. [Google Scholar] [CrossRef]
- Chakraborty, K.; Chatterjee, S.; Bhattacharyya, A. Impact of Treg on other T cell subsets in progression of fibrosis in experimental lung fibrosis. Tissue Cell 2018, 53, 87–92. [Google Scholar] [CrossRef] [PubMed]
- Garibaldi, B.T.; D’Alessio, F.R.; Mock, J.R.; Files, D.C.; Chau, E.; Eto, Y.; Drummond, M.B.; Aggarwal, N.R.; Sidhaye, V.; King, L.S. Regulatory T cells reduce acute lung injury fibroproliferation by decreasing fibrocyte recruitment. Am. J. Respir. Cell Mol. Biol. 2013, 48, 35–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiong, S.; Pan, X.; Xu, L.; Yang, Z.; Guo, R.; Gu, Y.; Li, R.; Wang, Q.; Xiao, F.; Du, L.; et al. Regulatory T Cells Promote beta-Catenin--Mediated Epithelium-to-Mesenchyme Transition During Radiation-Induced Pulmonary Fibrosis. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 425–435. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.H.; Deng, J.H.; Yao, X.L.; Wang, J.L.; Xiao, J.H. CD4(+)CD25(+) Tregs as dependent factor in the course of bleomycin-induced pulmonary fibrosis in mice. Exp. Cell Res. 2020, 386, 111700. [Google Scholar] [CrossRef]
- Schiller, H.B.; Mayr, C.H.; Leuschner, G.; Strunz, M.; Staab-Weijnitz, C.; Preisendörfer, S.; Eckes, B.; Moinzadeh, P.; Krieg, T.; Schwartz, D.A.; et al. Deep Proteome Profiling Reveals Common Prevalence of MZB1-Positive Plasma B Cells in Human Lung and Skin Fibrosis. Am. J. Respir. Crit. Care Med. 2017, 196, 1298–1310. [Google Scholar] [CrossRef] [PubMed]
- Vuga, L.J.; Tedrow, J.R.; Pandit, K.V.; Tan, J.; Kass, D.J.; Xue, J.; Chandra, D.; Leader, J.K.; Gibson, K.F.; Kaminski, N.; et al. C-X-C motif chemokine 13 (CXCL13) is a prognostic biomarker of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2014, 189, 966–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menon, M.; Hussell, T.; Ali Shuwa, H. Regulatory B cells in respiratory health and diseases. Immunol. Rev. 2021, 299, 61–73. [Google Scholar] [CrossRef]
- Lu, Y.; Liu, F.; Li, C.; Chen, Y.; Weng, D.; Chen, J. IL-10-Producing B Cells Suppress Effector T Cells Activation and Promote Regulatory T Cells in Crystalline Silica-Induced Inflammatory Response in vitro. Mediat. Inflamm. 2017, 2017, 8415094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Magro, C.M.; Waldman, W.J.; Knight, D.A.; Allen, J.N.; Nadasdy, T.; Frambach, G.E.; Ross, P.; Marsh, C.B. Idiopathic pulmonary fibrosis related to endothelial injury and antiendothelial cell antibodies. Hum. Immunol. 2006, 67, 284–297. [Google Scholar] [CrossRef] [PubMed]
- Cargnoni, A.; Romele, P.; Bonassi Signoroni, P.; Farigu, S.; Magatti, M.; Vertua, E.; Toschi, I.; Cesari, V.; Silini, A.R.; Stefani, F.R.; et al. Amniotic MSCs reduce pulmonary fibrosis by hampering lung B-cell recruitment, retention, and maturation. Stem Cells Transl. Med. 2020, 9, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
- Ueha, S.; Shand, F.H.; Matsushima, K. Cellular and molecular mechanisms of chronic inflammation-associated organ fibrosis. Front. Immunol. 2012, 3, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- King, T.E.; Pardo, A.; Selman, M. Idiopathic pulmonary fibrosis. Lancet 2011, 378, 1949–1961. [Google Scholar] [CrossRef]
- Philp, C.J.; Siebeke, I.; Clements, D.; Miller, S.; Habgood, A.; John, A.E.; Navaratnam, V.; Hubbard, R.B.; Jenkins, G.; Johnson, S.R. Extracellular Matrix Cross-Linking Enhances Fibroblast Growth and Protects against Matrix Proteolysis in Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2018, 58, 594–603. [Google Scholar] [CrossRef] [PubMed]
- Barkauskas, C.E.; Noble, P.W. Cellular mechanisms of tissue fibrosis. 7. New insights into the cellular mechanisms of pulmonary fibrosis. Am. J. Physiol. Cell Physiol. 2014, 306, C987–C996. [Google Scholar] [CrossRef] [Green Version]
- Andersson-Sjöland, A.; de Alba, C.G.; Nihlberg, K.; Becerril, C.; Ramírez, R.; Pardo, A.; Westergren-Thorsson, G.; Selman, M. Fibrocytes are a potential source of lung fibroblasts in idiopathic pulmonary fibrosis. Int. J. Biochem. Cell Biol. 2008, 40, 2129–2140. [Google Scholar] [CrossRef]
- Maharaj, S.S.; Baroke, E.; Gauldie, J.; Kolb, M.R. Fibrocytes in chronic lung disease--facts and controversies. Pulm. Pharmacol. Ther. 2012, 25, 263–267. [Google Scholar] [CrossRef] [PubMed]
- Mansouri, N.; Willis, G.R.; Fernandez-Gonzalez, A.; Reis, M.; Nassiri, S.; Mitsialis, S.A.; Kourembanas, S. Mesenchymal stromal cell exosomes prevent and revert experimental pulmonary fibrosis through modulation of monocyte phenotypes. JCI Insight 2019, 4, e128060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, T.; Yan, Y.; Wang, B.; Qian, H.; Zhang, X.; Shen, L.; Wang, M.; Zhou, Y.; Zhu, W.; Li, W.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013, 22, 845–854. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Wu, Y.; Wang, P.; Shi, M.; Wang, J.; Ma, H.; Sun, D. PSC-MSC-Derived Exosomes Protect against Kidney Fibrosis In Vivo and In Vitro through the SIRT6/beta-Catenin Signaling Pathway. Int. J. Stem Cells 2021, 14, 310–319. [Google Scholar] [CrossRef]
- Tikhomirov, R.; Donnell, B.R.; Catapano, F.; Faggian, G.; Gorelik, J.; Martelli, F.; Emanueli, C. Exosomes: From Potential Culprits to New Therapeutic Promise in the Setting of Cardiac Fibrosis. Cells 2020, 9, 592. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Bai, L.; Liu, X.; Shen, W.; Tian, H.; Liu, W.; Yu, B. Cardiac microvascular functions improved by MSC-derived exosomes attenuate cardiac fibrosis after ischemia-reperfusion via PDGFR-beta modulation. Int. J. Cardiol. 2021, 344, 13–24. [Google Scholar] [CrossRef]
- Li, M.; Zhang, H.P.; Wang, X.Y.; Chen, Z.G.; Lin, X.F.; Zhu, W. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Dermal Fibrosis in a Murine Model of Bleomycin-Induced Scleroderma. Stem Cells Dev. 2021, 30, 981–990. [Google Scholar] [CrossRef] [PubMed]
- Immune Modulation by Exosomes in COVID-19 (IMECOV19). 2022. Available online: https://www.clinicaltrials.gov/ct2/show/NCT05191381?term=exosome&cond=Lung+Fibrosis&draw=2&rank=1 (accessed on 21 July 2022).
- Farhat, W.; Hasan, A.; Lucia, L.; Becquart, F.; Ayoub, A.; Kobeissy, F. Hydrogels for Advanced Stem Cell Therapies: A Biomimetic Materials Approach for Enhancing Natural Tissue Function. IEEE Rev. Biomed. Eng. 2019, 12, 333–351. [Google Scholar] [CrossRef] [PubMed]
- Willis, G.R.; Fernandez-Gonzalez, A.; Anastas, J.; Vitali, S.H.; Liu, X.; Ericsson, M.; Kwong, A.; Mitsialis, S.A.; Kourembanas, S. Mesenchymal Stromal Cell Exosomes Ameliorate Experimental Bronchopulmonary Dysplasia and Restore Lung Function through Macrophage Immunomodulation. Am. J. Respir. Crit. Care Med. 2018, 197, 104–116. [Google Scholar] [CrossRef] [PubMed]
- Tan, J.L.; Lau, S.N.; Leaw, B.; Nguyen, H.P.T.; Salamonsen, L.A.; Saad, M.I.; Chan, S.T.; Zhu, D.; Krause, M.; Kim, C.; et al. Amnion Epithelial Cell-Derived Exosomes Restrict Lung Injury and Enhance Endogenous Lung Repair. Stem Cells Transl. Med. 2018, 7, 180–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Dwyer, D.N.; Armstrong, M.E.; Trujillo, G.; Cooke, G.; Keane, M.P.; Fallon, P.G.; Simpson, A.J.; Millar, A.B.; McGrath, E.E.; Whyte, M.K.; et al. The Toll-like receptor 3 L412F polymorphism and disease progression in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2013, 188, 1442–1450. [Google Scholar] [CrossRef] [PubMed]
- Whyte, M.; Hubbard, R.; Meliconi, R.; Whidborne, M.; Eaton, V.; Bingle, C.; Timms, J.; Duff, G.; Facchini, A.; Pacilli, A.; et al. Increased risk of fibrosing alveolitis associated with interleukin-1 receptor antagonist and tumor necrosis factor-alpha gene polymorphisms. Am. J. Respir. Crit. Care Med. 2000, 162 Pt 1, 755–758. [Google Scholar] [CrossRef] [PubMed]
- Korthagen, N.M.; van Moorsel, C.H.; Kazemier, K.M.; Ruven, H.J.; Grutters, J.C. IL1RN genetic variations and risk of IPF: A meta-analysis and mRNA expression study. Immunogenetics 2012, 64, 371–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shentu, T.P.; Huang, T.S.; Cernelc-Kohan, M.; Chan, J.; Wong, S.S.; Espinoza, C.R.; Tan, C.; Gramaglia, I.; van der Heyde, H.; Chien, S.; et al. Thy-1 dependent uptake of mesenchymal stem cell-derived extracellular vesicles blocks myofibroblastic differentiation. Sci. Rep. 2017, 7, 18052. [Google Scholar] [CrossRef] [Green Version]
- Wolters, P.J.; Collard, H.R.; Jones, K.D. Pathogenesis of idiopathic pulmonary fibrosis. Annu. Rev. Pathol. 2014, 9, 157–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Collard, H.R.; Moore, B.B.; Flaherty, K.R.; Brown, K.K.; Kaner, R.J.; King, T.E.; Lasky, J.A.; Loyd, J.E.; Noth, I.; Olman, M.A.; et al. Acute exacerbations of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2007, 176, 636–643. [Google Scholar] [CrossRef] [Green Version]
- Dinh, P.C.; Paudel, D.; Brochu, H.; Popowski, K.D.; Gracieux, M.C.; Cores, J.; Huang, K.; Hensley, M.T.; Harrell, E.; Vandergriff, A.C.; et al. Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis. Nat. Commun. 2020, 11, 1064. [Google Scholar] [CrossRef] [PubMed]
- Wan, X.; Chen, S.; Fang, Y.; Zuo, W.; Cui, J.; Xie, S. Mesenchymal stem cell-derived extracellular vesicles suppress the fibroblast proliferation by downregulating FZD6 expression in fibroblasts via micrRNA-29b-3p in idiopathic pulmonary fibrosis. J. Cell Physiol. 2020, 235, 8613–8625. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Zhu, M.; Feng, W.; Lin, Y.; Yin, J.; Jin, J.; Wang, Y. Exosomal miRNA Let-7 from Menstrual Blood-Derived Endometrial Stem Cells Alleviates Pulmonary Fibrosis through Regulating Mitochondrial DNA Damage. Oxidative Med. Cell. Longev. 2019, 2019, 4506303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kusuma, G.D.; Li, A.; Zhu, D.; McDonald, H.; Inocencio, I.M.; Chambers, D.C.; Sinclair, K.; Fang, H.; Greening, D.W.; Frith, J.E.; et al. Effect of 2D and 3D Culture Microenvironments on Mesenchymal Stem Cell-Derived Extracellular Vesicles Potencies. Front. Cell Dev. Biol. 2022, 10, 819726. [Google Scholar] [CrossRef]
- Choi, M.; Ban, T.; Rhim, T. Therapeutic use of stem cell transplantation for cell replacement or cytoprotective effect of microvesicle released from mesenchymal stem cell. Mol. Cells 2014, 37, 133–139. [Google Scholar] [CrossRef] [Green Version]
- Bandeira, E.; Oliveira, H.; Silva, J.D.; Menna-Barreto, R.F.S.; Takyia, C.M.; Suk, J.S.; Witwer, K.W.; Paulaitis, M.E.; Hanes, J.; Rocco, P.R.M.; et al. Therapeutic effects of adipose-tissue-derived mesenchymal stromal cells and their extracellular vesicles in experimental silicosis. Respir. Res. 2018, 19, 104. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Hu, H.; Zhang, S.; Jiang, L.; Cheng, Y.; Xie, H.; Wang, X.; Jiang, J.; Wang, H.; Zhang, Q. Human umbilical cord mesenchymal stem cell-derived exosomes alleviate pulmonary fibrosis in mice by inhibiting epithelial-mesenchymal transition. Nan Fang Yi Ke Da Xue Xue Bao 2020, 40, 988–994. [Google Scholar] [PubMed]
- Xu, C.; Zhao, J.; Li, Q.; Hou, L.; Wang, Y.; Li, S.; Jiang, F.; Zhu, Z.; Tian, L. Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model. Stem Cell Res. Ther. 2020, 11, 503. [Google Scholar] [CrossRef] [PubMed]
- Rozier, P.; Maumus, M.; Maria, A.T.J.; Toupet, K.; Jorgensen, C.; Guilpain, P.; Noel, D. Lung Fibrosis Is Improved by Extracellular Vesicles from IFNgamma-Primed Mesenchymal Stromal Cells in Murine Systemic Sclerosis. Cells 2021, 10, 2727. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Lin, Y.; Kang, X.; Liu, Z.; Zhang, W.; Xu, F. microRNA-186 in extracellular vesicles from bone marrow mesenchymal stem cells alleviates idiopathic pulmonary fibrosis via interaction with SOX4 and DKK1. Stem Cell Res. Ther. 2021, 12, 96. [Google Scholar] [CrossRef] [PubMed]
- Zhang, E.; Geng, X.; Shan, S.; Li, P.; Li, S.; Li, W.; Yu, M.; Peng, C.; Wang, S.; Shao, H.; et al. Exosomes derived from bone marrow mesenchymal stem cells reverse epithelial-mesenchymal transition potentially via attenuating Wnt/β-catenin signaling to alleviate silica-induced pulmonary fibrosis. Toxicol. Mech. Methods 2021, 31, 655–666. [Google Scholar] [CrossRef] [PubMed]
- Shi, L.; Ren, J.; Li, J.; Wang, D.; Wang, Y.; Qin, T.; Li, X.; Zhang, G.; Li, C.; Wang, Y. Extracellular vesicles derived from umbilical cord mesenchymal stromal cells alleviate pulmonary fibrosis by means of transforming growth factor-β signaling inhibition. Stem Cell Res. Ther. 2021, 12, 230. [Google Scholar] [CrossRef] [PubMed]
- Lei, X.; He, N.; Zhu, L.; Zhou, M.; Zhang, K.; Wang, C.; Huang, H.; Chen, S.; Li, Y.; Liu, Q.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Radiation-Induced Lung Injury via miRNA-214-3p. Antioxid. Redox Signal. 2021, 35, 849–862. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Shen, Z.; Jiang, X.; Wang, Y.; Yang, Z.; Mao, Y.; Wu, Z.; Li, G.; Chen, H. Mouse mesenchymal stem cell-derived exosomal miR-466f-3p reverses EMT process through inhibiting AKT/GSK3beta pathway via c-MET in radiation-induced lung injury. J. Exp. Clin. Cancer Res. 2022, 41, 128. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Hou, L.; Zhao, J.; Wang, Y.; Jiang, F.; Jiang, Q.; Zhu, Z.; Tian, L. Exosomal let-7i-5p from three-dimensional cultured human umbilical cord mesenchymal stem cells inhibits fibroblast activation in silicosis through targeting TGFBR1. Ecotoxicol. Environ. Saf. 2022, 233, 113302. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.; Liu, P.; Gao, T.; Song, D.; Zhao, X.; Li, Y.; Wu, J.; Wang, L.; Wang, Z.; Hao, J.; et al. Every road leads to Rome: Therapeutic effect and mechanism of the extracellular vesicles of human embryonic stem cell-derived immune and matrix regulatory cells administered to mouse models of pulmonary fibrosis through different routes. Stem Cell Res. Ther. 2022, 13, 163. [Google Scholar] [CrossRef] [PubMed]
- Morishita, M.; Takahashi, Y.; Nishikawa, M.; Takakura, Y. Pharmacokinetics of Exosomes-An Important Factor for Elucidating the Biological Roles of Exosomes and for the Development of Exosome-Based Therapeutics. J. Pharm. Sci. 2017, 106, 2265–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, L.; Hao, Q.; Sugita, S.; Naito, Y.; He, H.; Yeh, C.C.; Lee, J.W. Role of CD44 in increasing the potency of mesenchymal stem cell extracellular vesicles by hyaluronic acid in severe pneumonia. Stem Cell Res. Ther. 2021, 12, 293. [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] [Green Version]
- Lai, R.C.; Tan, S.S.; Yeo, R.W.; Choo, A.B.; Reiner, A.T.; Su, Y.; Shen, Y.; Fu, Z.; Alexander, L.; Sze, S.K.; et al. MSC secretes at least 3 EV types each with a unique permutation of membrane lipid, protein and RNA. J. Extracell. Vesicles 2016, 5, 29828. [Google Scholar] [CrossRef]
- Baglio, S.R.; Rooijers, K.; Koppers-Lalic, D.; Verweij, F.J.; Perez Lanzon, M.; Zini, N.; Naaijkens, B.; Perut, F.; Niessen, H.W.; Baldini, N.; et al. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res. Ther. 2015, 6, 127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bari, E.; Perteghella, S.; Di Silvestre, D.; Sorlini, M.; Catenacci, L.; Sorrenti, M.; Marrubini, G.; Rossi, R.; Tripodo, G.; Mauri, P.; et al. Pilot Production of Mesenchymal Stem/Stromal Freeze-Dried Secretome for Cell-Free Regenerative Nanomedicine: A Validated GMP-Compliant Process. Cells 2018, 7, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W. Lyophilization and development of solid protein pharmaceuticals. Int. J. Pharm. 2000, 203, 1–60. [Google Scholar] [CrossRef]
- Ren, J.; Liu, Y.; Yao, Y.; Feng, L.; Zhao, X.; Li, Z.; Yang, L. Intranasal delivery of MSC-derived exosomes attenuates allergic asthma via expanding IL-10 producing lung interstitial macrophages in mice. Int. Immunopharmacol. 2021, 91, 107288. [Google Scholar] [CrossRef]
- Zhao, R.; Wang, L.; Wang, T.; Xian, P.; Wang, H.; Long, Q. Inhalation of MSC-EVs is a noninvasive strategy for ameliorating acute lung injury. J. Control. Release 2022, 345, 214–230. [Google Scholar] [CrossRef] [PubMed]
- Zoulikha, M.; Xiao, Q.; Boafo, G.F.; Sallam, M.A.; Chen, Z.; He, W. Pulmonary delivery of siRNA against acute lung injury/acute respiratory distress syndrome. Acta Pharm. Sin. B 2022, 12, 600–620. [Google Scholar] [CrossRef]
- Shamili, F.H.; Bayegi, H.R.; Salmasi, Z.; Sadri, K.; Mahmoudi, M.; Kalantari, M.; Ramezani, M.; Abnous, K. Exosomes derived from TRAIL-engineered mesenchymal stem cells with effective anti-tumor activity in a mouse melanoma model. Int. J. Pharm. 2018, 549, 218–229. [Google Scholar] [CrossRef] [PubMed]
- Pascucci, L.; Cocce, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Vigano, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. [Google Scholar] [CrossRef] [PubMed]
- Kore, R.A.; Wang, X.; Ding, Z.; Griffin, R.J.; Tackett, A.J.; Mehta, J.L. MSC exosome-mediated cardioprotection in ischemic mouse heart comparative proteomics of infarct and peri-infarct areas. Mol. Cell. Biochem. 2021, 476, 1691–1704. [Google Scholar] [CrossRef] [PubMed]
- Xian, P.; Hei, Y.; Wang, R.; Wang, T.; Yang, J.; Li, J.; Di, Z.; Liu, Z.; Baskys, A.; Liu, W.; et al. Mesenchymal stem cell-derived exosomes as a nanotherapeutic agent for amelioration of inflammation-induced astrocyte alterations in mice. Theranostics 2019, 9, 5956–5975. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Fan, M.; Huang, D.; Li, B.; Xu, R.; Gao, F.; Chen, Y. Clodronate-loaded liposomal and fibroblast-derived exosomal hybrid system for enhanced drug delivery to pulmonary fibrosis. Biomaterials 2021, 271, 120761. [Google Scholar] [CrossRef] [PubMed]
- Lu, M.; Xing, H.; Xun, Z.; Yang, T.; Zhao, X.; Cai, C.; Wang, D.; Ding, P. Functionalized extracellular vesicles as advanced therapeutic nanodelivery systems. Eur. J. Pharm. Sci. 2018, 121, 34–46. [Google Scholar] [CrossRef]
- Armstrong, J.P.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, N.; Song, Y.; Huang, Z.; Chen, J.; Tan, H.; Yang, H.; Fan, M.; Li, Q.; Wang, Q.; Gao, J.; et al. Monocyte mimics improve mesenchymal stem cell-derived extracellular vesicle homing in a mouse MI/RI model. Biomaterials 2020, 255, 120168. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Xu, Y.; Lv, K.; Wang, Y.; Zhong, Z.; Xiao, C.; Zhu, K.; Ni, C.; Wang, K.; Kong, M.; et al. Small extracellular vesicles containing miR-486-5p promote angiogenesis after myocardial infarction in mice and nonhuman primates. Sci. Transl. Med. 2021, 13, eabb0202. [Google Scholar] [CrossRef]
- Hung, M.E.; Leonard, J.N. Stabilization of exosome-targeting peptides via engineered glycosylation. J. Biol. Chem. 2015, 290, 8166–8172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kooijmans, S.A.A.; Aleza, C.G.; Roffler, S.R.; van Solinge, W.W.; Vader, P.; Schiffelers, R.M. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J. Extracell. Vesicles 2016, 5, 31053. [Google Scholar] [CrossRef] [PubMed]
- Wan, Z.; Zhao, L.; Lu, F.; Gao, X.; Dong, Y.; Zhao, Y.; Wei, M.; Yang, G.; Xing, C.; Liu, L. Mononuclear phagocyte system blockade improves therapeutic exosome delivery to the myocardium. Theranostics 2020, 10, 218–230. [Google Scholar] [CrossRef]
- Belhadj, Z.; He, B.; Deng, H.; Song, S.; Zhang, H.; Wang, X.; Dai, W.; Zhang, Q. A combined “eat me/don’t eat me” strategy based on extracellular vesicles for anticancer nanomedicine. J. Extracell. Vesicles 2020, 9, 1806444. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Li, Y.; Liu, P.; Gong, D.; Zhou, H.; Li, W.; Zhang, H.; Zheng, W.; Xu, J.; Cheng, H.; et al. Phosphatase Shp2 regulates biogenesis of small extracellular vesicles by dephosphorylating Syntenin. J. Extracell. Vesicles 2021, 10, e12078. [Google Scholar] [CrossRef] [PubMed]
- Park, K.S.; Svennerholm, K.; Shelke, G.V.; Bandeira, E.; Lasser, C.; Jang, S.C.; Chandode, R.; Gribonika, I.; Lotvall, J. Mesenchymal stromal cell-derived nanovesicles ameliorate bacterial outer membrane vesicle-induced sepsis via IL-10. Stem Cell Res. Ther. 2019, 10, 231. [Google Scholar] [CrossRef] [Green Version]
- Champion, J.A.; Walker, A.; Mitragotri, S. Role of particle size in phagocytosis of polymeric microspheres. Pharm. Res. 2008, 25, 1815–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Porzionato, A.; Zaramella, P.; Dedja, A.; Guidolin, D.; van Wemmel, K.; Macchi, V.; Jurga, M.; Perilongo, G.; de Caro, R.; Baraldi, E.; et al. Intratracheal administration of clinical-grade mesenchymal stem cell-derived extracellular vesicles reduces lung injury in a rat model of bronchopulmonary dysplasia. Am. J. Physiol. Cell. Mol. Physiol. 2019, 316, L6–L19. [Google Scholar] [CrossRef] [PubMed]
- Aliotta, J.M.; Pereira, M.; Wen, S.; Dooner, M.S.; del Tatto, M.; Papa, E.; Goldberg, L.R.; Baird, G.L.; Ventetuolo, C.E.; Quesenberry, P.J.; et al. Exosomes induce and reverse monocrotaline-induced pulmonary hypertension in mice. Cardiovasc. Res. 2016, 110, 319–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Monsel, A.; Zhu, Y.; Gennai, S.; Hao, Q.; Hu, S.; Rouby, J.; Rosenzwajg, M.; Matthay, M.A.; Lee, J.W. Therapeutic Effects of Human Mesenchymal Stem Cell-derived Microvesicles in Severe Pneumonia in Mice. Am. J. Respir. Crit. Care Med. 2015, 192, 324–336. [Google Scholar] [CrossRef] [Green Version]
- Qiu, X.; Li, Z.; Han, X.; Zhen, L.; Luo, C.; Liu, M.; Yu, K.; Ren, Y. Tumor-derived nanovesicles promote lung distribution of the therapeutic nanovector through repression of Kupffer cell-mediated phagocytosis. Theranostics 2019, 9, 2618–2636. [Google Scholar] [CrossRef] [PubMed]
- Gyorgy, B.; Hung, M.E.; Breakefield, X.O.; Leonard, J.N. Therapeutic applications of extracellular vesicles: Clinical promise and open questions. Annu. Rev. Pharmacol. Toxicol. 2015, 55, 439–464. [Google Scholar] [CrossRef] [Green Version]
- Gimona, M.; Pachler, K.; Laner-Plamberger, S.; Schallmoser, K.; Rohde, E. Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use. Int. J. Mol. Sci. 2017, 18, 1190. [Google Scholar] [CrossRef]
- Rodrigues, S.C.; Cardoso, R.M.S.; Gomes, C.F.; Duarte, F.V.; Freire, P.C.; Neves, R.; Simoes-Correia, J. Toxicological Profile of Umbilical Cord Blood-Derived Small Extracellular Vesicles. Membranes 2021, 11, 647. [Google Scholar] [CrossRef] [PubMed]
- Rudokas, M.; Najlah, M.; Alhnan, M.A.; Elhissi, A. Liposome Delivery Systems for Inhalation: A Critical Review Highlighting Formulation Issues and Anticancer Applications. Med. Princ. Pract. 2016, 25 (Suppl. S2), 60–72. [Google Scholar] [CrossRef] [PubMed]
- Elhissi, A.; Faizi, M.; Naji, W.; Gill, H.; Taylor, K. Physical stability and aerosol properties of liposomes delivered using an air-jet nebulizer and a novel micropump device with large mesh apertures. Int. J. Pharm. 2007, 334, 62–70. [Google Scholar] [CrossRef] [PubMed]
- Harrell, C.R.; Miloradovic, D.; Sadikot, R.; Fellabaum, C.; Markovic, B.S.; Miloradovic, D.; Acovic, A.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Molecular and Cellular Mechanisms Responsible for Beneficial Effects of Mesenchymal Stem Cell-Derived Product “Exo-d-MAPPS” in Attenuation of Chronic Airway Inflammation. Anal. Cell. Pathol. 2020, 2020, 3153891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawson, T.; Kehoe, D.E.; Schnitzler, A.C.; Rapiejko, P.J.; Der, K.A.; Philbrick, K.; Punreddy, S.; Rigby, S.; Smith, R.; Feng, Q.; et al. Process development for expansion of human mesenchymal stromal cells in a 50L single-use stirred tank bioreactor. Biochem. Eng. J. 2017, 120, 49–62. [Google Scholar] [CrossRef]
- Mizukami, A.; Pereira Chilima, T.D.; Orellana, M.D.; Neto, M.A.; Covas, D.T.; Farid, S.S.; Swiech, K. Technologies for large-scale umbilical cord-derived MSC expansion: Experimental performance and cost of goods analysis. Biochem. Eng. J. 2018, 135, 36–48. [Google Scholar] [CrossRef]
- Haraszti, R.A.; Miller, R.; Stoppato, M.; Sere, Y.Y.; Coles, A.; Didiot, M.C.; Wollacott, R.; Sapp, E.; Dubuke, M.L.; Li, X.; et al. Exosomes Produced from 3D Cultures of MSCs by Tangential Flow Filtration Show Higher Yield and Improved Activity. Mol. Ther. 2018, 26, 2838–2847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Characteristic | Exosomes | Microvesicles | Apoptotic Bodies |
---|---|---|---|
Size (nm) | 20–150 | 50–1500 | 50–2000 |
Origin | Invagination of cell membrane | Cell membrane budding and fission | Released by cells undergoing apoptosis, plasma membrane, endoplasmic reticulum |
Morphology | Cup/round shaped | Various shapes | Heterogeneous |
Sucrose gradient | 1.13–1.19 g/mL | 1.04–1.07 g/mL | 1.16–1.28 g/mL |
Surface markers | Annexins, tetraspanins, heat-shock proteins | CD40, cholesterol, sphingomyelin, and ceramide | Annexin V positivity, TSP, C3b |
Contents | Proteins, enzymes, signal transduction factors, biogenesis factors, chaperones, nucleic acids | Proteins, nucleic acids, lipids | Nuclear fractions, DNA, cell organelles |
Isolation technique | Sediment at approximately 100,000 g | Sediment at approximately 10 to 14,000 g | Ultracentrifugation |
Number | Reference | Origin | Target Tissue/Model | Year | Administration | Dosage |
---|---|---|---|---|---|---|
1 | [128] | Human BMSC microvesicles | Lung fibrosis/silica/mouse | 2014 | Tail vein injection | 10 μg |
2 | [117] | hAEC exosomes | Lung fibrosis/bleomycin/mouse | 2018 | Intranasal administration | 10 μg |
3 | [129] | AD-MSCs exosomes | Lung fibrosis/silica/mouse | 2018 | Intratracheal injection | EVs obtained from 100,000 AD-MSCs for 24 h |
4 | [108] | Human BMSC-EVs/exosomes | Lung fibrosis/bleomycin/mouse | 2019 | Tail vein injection | 200 μL, 8.6–108 particles |
5 | [126] | MenSC exosomes | Lung fibrosis/bleomycin/mouse | 2019 | Tail vein injection | 0.5 mg/kg/day |
6 | [124] | Human BMSC exosomes | Lung fibrosis/bleomycin sulfate or silica/mouse | 2020 | Nebulization | 10 × 109 particles/kg |
7 | [20] | Human BMSC exosomes | Lung fibrosis/LPS/mouse | 2020 | Tail vein injection | 70 μg |
8 | [130] | hucMSC exosomes | Lung fibrosis/bleomycin/mouse | 2020 | Tail vein injection | 100 μg/250 μL |
9 | [131] | hucMSC exosomes | Lung fibrosis/silica/mouse | 2020 | Tail vein injection | - |
10 | [125] | Human BMSC-EVs | Lung fibrosis/bleomycin/mouse | 2020 | Tail vein injection | 100 μg |
11 | [132] | Mouse BMSC-EVs | Lung fibrosis/systemic sclerosis/mouse | 2021 | Intravenous injection | 250 ng or 1500 ng |
12 | [133] | Human BMSC-EVs | Lung fibrosis/bleomycin/mouse | 2021 | Tail vein injection | 100 μg |
13 | [134] | Rat-BMSC exosomes | Lung fibrosis/silica/rat | 2021 | Tail vein injection | 200 μg/mL/rat |
14 | [135] | huMSC EVs | Lung fibrosis/bleomycin/mouse | 2021 | Tail vein injection | 20 μg |
15 | [136] | Human placenta-derived MSC-EVs | Lung fibrosis/radiation/mouse | 2021 | Tail vein injection | 100 μg |
16 | [127] | Human BMSC-EVs | Lung fibrosis/bleomycin/mouse | 2022 | Intranasal administration | 10 μg |
17 | [137] | mMSC exosomes | Lung fibrosis/radiation/mouse | 2022 | Tail vein injection | 200 μg |
18 | [138] | hucMSC exosomes | Lung fibrosis/silica/mouse | 2022 | - | - |
19 | [139] | IMRCs EVs | Lung fibrosis/bleomycin /mouse | 2022 | Intratracheal or tail vein injection | 200 μg or 1000 μg |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Sang, L.; Guo, X.; Fan, H.; Shi, J.; Hou, S.; Lv, Q. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Idiopathic Pulmonary Fibrosis Microenvironment Targeted Delivery. Cells 2022, 11, 2322. https://doi.org/10.3390/cells11152322
Sang L, Guo X, Fan H, Shi J, Hou S, Lv Q. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Idiopathic Pulmonary Fibrosis Microenvironment Targeted Delivery. Cells. 2022; 11(15):2322. https://doi.org/10.3390/cells11152322
Chicago/Turabian StyleSang, Lu, Xiaoqin Guo, Haojun Fan, Jie Shi, Shike Hou, and Qi Lv. 2022. "Mesenchymal Stem Cell-Derived Extracellular Vesicles as Idiopathic Pulmonary Fibrosis Microenvironment Targeted Delivery" Cells 11, no. 15: 2322. https://doi.org/10.3390/cells11152322
APA StyleSang, L., Guo, X., Fan, H., Shi, J., Hou, S., & Lv, Q. (2022). Mesenchymal Stem Cell-Derived Extracellular Vesicles as Idiopathic Pulmonary Fibrosis Microenvironment Targeted Delivery. Cells, 11(15), 2322. https://doi.org/10.3390/cells11152322