Therapeutic potential of HUC-MSC-exos primed with IFN-γ against LPS-induced acute lung injury

Document Type : Original Article

Authors

1 Kunming Medical University, Kunming, China

2 Department of Emergency Intensive Care Unit, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China

3 Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China

4 Department of Emergency, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China

Abstract

Objective(s): Human umbilical cord mesenchymal stem cells (HUC-MSCs) are pluripotent stem cells with anti-inflammatory and immunomodulatory properties used in the treatment of acute lung injury (ALI). However, the treatment of ALI using exosomes derived from HUC-MSCs (HUC-MSC-exos) primed with interferon-gamma (IFN-γ-exos) has not been described. This study investigated the effects of IFN-γ-exos on ALI.
Materials and Methods: IFN-γ primed and unprimed HUC-MSC-exos (IFN-γ-exos and CON-exos, respectively) were extracted, identified, and traced. A549 cells and mice subjected to lipopolysaccharide (LPS)-induced inflammation were treated with IFN-γ-exos or CON-exos. Viability; interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and reactive oxygen species (ROS) levels; NF-κB p65, and NLRP3 expression and histology and lung injury scores were measured in cell, supernatant or lung tissue. 
Results: Indoleamine 2,3-dioxygenase (IDO) mRNA expression was elevated in HUC-MSCs primed with 5 ng/mL IFN-γ (P<0.001), and IFN-γ-exos and CON-exos were successfully extracted. LPS-induced inflammation resulted in decreased cell viability in A549 cells, and increased IL-1β, IL-6, TNF-α and ROS levels and NF-κB p65 and NLRP3 expression in A549 cells and mice(P<0.05 to P<0.001). Treatment with IFN-γ-exos and CON-exos increased cell viability and decreased the concentrations of IL-1β, and ROS, expression of NF-κB p65 and NLRP3, and the lung injury score, and these effects were more obvious for IFN-γ-exos(P<0.05 to P<0.001). 
Conclusion: IFN-γ-exos reduced oxidative stress and inflammatory responses in LPS-induced A549 cells and mice. The result demonstrated the therapeutic potential of IFN-γ-exos in LPS-induced ALI.

Keywords

Main Subjects


1. Ashbaugh DG, Bigelow DB, Petty TL, Levine BE. Acute respiratory distress in adults. Lancet 1967; 2:319-323.
2. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 2000; 342:1334-1349.
3. Meyer NJ, Gattinoni L, Calfee CS. Acute respiratory distress syndrome. Lancet 2021; 398:622-637.
4. Sinha P, Matthay MA, Calfee CS. Is a “cytokine storm” relevant to COVID-19? JAMA Intern Med 2020; 180:1152-1154.
5. Lu K, Geng ST, Tang S, Yang H, Xiong W, Xu F, et al. Clinical efficacy and mechanism of mesenchymal stromal cells in treatment of COVID-19. Stem Cell Res Ther 2022; 13:61-75.
6. Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries.  2016; 315:788-800.
7. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020; 367:eaau6977.
8. Han QF, Li WJ, Hu KS, Gao J, Zhai WL, Yang JH, et al. Exosome biogenesis: Machinery, regulation, and therapeutic implications in cancer. Mol Cancer 2022; 21:207-232.
9. Chen CC, Liu L, Ma F, Wong CW, Guo XE, Chacko JV, et al. Elucidation of exosome migration across the blood-brain barrier model in vitro. Cell Mol Bioeng 2016; 9:509-529.
10. Wang X, Liu D, Zhang X, Yang L, Xia Z, Zhang Q. Exosomes from adipose-derived mesenchymal stem cells alleviate sepsis-induced lung injury in mice by inhibiting the secretion of IL-27 in macrophages. Cell Death Discov 2022; 8:18-28.
11. Liew LC, Katsuda T, Gailhouste L, Nakagama H, Ochiya T. Mesenchymal stem cell-derived extracellular vesicles: A glimmer of hope in treating Alzheimer’s disease. Int Immunol 2017; 29:11-19.
12. Phinney DG, Pittenger MF. Concise review: MSC-Derived exosomes for cell-free therapy. Stem Cells 2017; 35:851-858.
13. Zhang B, Huang J, Liu J, Lin F, Ding Z, Xu J. Injectable composite hydrogel promotes osteogenesis and angiogenesis in spinal fusion by optimizing the bone marrow mesenchymal stem cell microenvironment and exosomes secretion. Mater Sci Eng C Mater Biol Appl 2021; 123:111782.
14. Wobma H, Tamargo M, Goeta S, Brown L, Duran-Struuck R, Vunjak-Novakovic GJB. The influence of hypoxia and IFN-γ on the proteome and metabolome of therapeutic mesenchymal stem cells. Biomaterials 2018; 167:226-234.
15. Krampera M, Galipeau J, Shi Y, Tarte K, Sensebe L, Therapy MSCCotISfC. Immunological characterization of multipotent mesenchymal stromal cells--The International Society for Cellular Therapy (ISCT) working proposal. Cytotherapy 2013; 15:1054-1061.
16. Grote K, Petri M, Liu C, Jehn P, Spalthoff S, Kokemüller H, et al. Toll-like receptor 2/6-dependent stimulation of mesenchymal stem cells promotes angiogenesis by paracrine factors. Eur Cell Mater 2013; 26:66-79.
17. Liang B, Chen J, Li T, Wu H, Yang W, Li Y, et al. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells: A case report. Medicine (Baltimore) 2020; 99:e21429.
18. Rodríguez-Eguren A, Gómez-Álvarez M, Francés-Herrero E, Romeu M, Ferrero H, Seli E, et al. Human umbilical cord-based therapeutics: Stem cells and blood derivatives for female reproductive medicine. Int J Mol Sci 2022; 23:15942-15971.
19. Eggenhofer E, Luk F, Dahlke MH, Hoogduijn MJ. The life and fate of mesenchymal stem cells. Front Immunol 2014; 5:148-153.
20. Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS, et al. An official American Thoracic Society workshop report: Features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol 2011; 44:725-738.
21. Piekarska K, Urban-Wójciuk Z, Kurkowiak M, Pelikant-Małecka I, Schumacher A, Sakowska J, et al. Mesenchymal stem cells transfer mitochondria to allogeneic Tregs in an HLA-dependent manner improving their immunosuppressive activity. Nat Commun 2022; 13:856-875.
22. Shi Y, Hu G, Su J, Li W, Chen Q, Shou P, et al. Mesenchymal stem cells: A new strategy for immunosuppression and tissue repair.  Cell Res 2010; 20:510-518.
23. Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem 2019; 88:487-514.
24. Walker S, Busatto S, Pham A, Tian M, Suh A, Carson K, et al. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics 2019; 9:8001-8017.
25. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics 2021; 11:3183-3195.
26.    Salunkhe S, Dheeraj, Basak M, Chitkara D, Mittal A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: Strategies and significance. J Control Release 2020; 326:599-614.
27. Grange C, Tapparo M, Bruno S, Chatterjee D, Quesenberry PJ, Tetta C, et al. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int J Mol Med 2014; 33:1055-1063.
28. Duan L, Xu L, Xu X, Qin Z, Zhou X, Xiao Y, et al. Exosome-mediated delivery of gene vectors for gene therapy. Nanoscale 2021; 13:1387-1397.
29. Krylova SV, Feng D. The machinery of exosomes: Biogenesis, release, and uptake. Int J Mol Sci 2023; 24:1337-1351.
30. Wang W, Zhu N, Yan T, Shi YN, Chen J, Zhang CJ, et al. The crosstalk: Exosomes and lipid metabolism. Cell Commun Signal 2020; 18:119-130.
31. Boskabady M, Khazdair MR, Bargi R, Saadat S, Memarzia A, Mohammadian Roshan N, et al. Thymoquinone ameliorates lung inflammation and pathological changes observed in lipopolysaccharide-induced lung injury. Evid Based Complement Alternat Med 2021; 2021:1-10.
32. Arab Z, Hosseini M, Marefati N, Beheshti F, Anaeigoudari A, Sadeghnia HR, et al. Neuroprotective and memory enhancing effects of Zataria multiflora in lipopolysaccharide-treated rats. Vet Res Forum 2022; 13:101-110.
33. Gu T, Zhang Z, Liu J, Chen L, Tian Y, Xu W, et al. Chlorogenic acid alleviates LPS-induced inflammation and oxidative stress by modulating CD36/AMPK/PGC-1α in RAW264.7 macrophages. Int J Mol Sci 2023; 24:13516-13528 .
34. Kumar J, Haldar C, Verma R. Melatonin ameliorates LPS-induced testicular nitro-oxidative stress (iNOS/TNFα) and inflammation (NF-kB/COX-2) via modulation of SIRT-1. Reprod Sci 2021; 28:3417-3430.
35. He Y, Li Z, Xu T, Luo D, Chi Q, Zhang Y, et al. Polystyrene nanoplastics deteriorate LPS-modulated duodenal permeability and inflammation in mice via ROS drived-NF-κB/NLRP3 pathway. Chemosphere 2022; 307:135662.
36. Li J, Lu K, Sun F, Tan S, Zhang X, Sheng W, et al. Panaxydol attenuates ferroptosis against LPS-induced acute lung injury in mice by Keap1-Nrf2/HO-1 pathway. J Transl Med 2021; 19:96-109.
37. Luan R, Ding D, Yang J. The protective effect of natural medicines against excessive inflammation and oxidative stress in acute lung injury by regulating the Nrf2 signaling pathway. Front Pharmacol 2022; 13:1039022.
38. Mohamed GA, Ibrahim SRM, El-Agamy DS, Elsaed WM, Sirwi A, Asfour HZ, et al. Terretonin as a new protective agent against sepsis-induced acute lung injury: Impact on SIRT1/Nrf2/NF-κBp65/NLRP3 signaling. Biology (Basel) 2021; 10:1219-1236.
39. Zheng Y, Liu J, Chen P, Lin L, Luo Y, Ma X, et al. Exosomal miR-22-3p from human umbilical cord blood-derived mesenchymal stem cells protects against lipopolysaccharid-induced acute lung injury. Life Sci 2021; 269:119004.
40. Boskabadi J, Mokhtari-Zaer A, Abareshi A, Khazdair MR, Emami B, Mohammadian Roshan N, et al. The effect of captopril on lipopolysaccharide-induced lung inflammation. Exp Lung Res 2018; 44:191-200.
41. Mokrá D. Acute lung injury - from pathophysiology to treatment. Physiol Res 2020; 69:S353-S366.
42. Boskabadi J, Askari VR, Hosseini M, Boskabady MH. Immunomodulatory properties of captopril, an ACE inhibitor, on LPS-induced lung inflammation and fibrosis as well as oxidative stress. Inflammopharmacology 2018; 27:639-647.
43. Afonina IS, Zhong Z, Karin M, Beyaert R. Limiting inflammation-the negative regulation of NF-κB and the NLRP3 inflammasome. Nat Immunol 2017; 18:861-869.
44. Saadat S, Beheshti F, Askari VR, Hosseini M, Mohamadian Roshan N, Boskabady MH. Aminoguanidine affects systemic and lung inflammation induced by lipopolysaccharide in rats. Respir Res 2019; 20:96-108.
45. Gholamnezhad Z, Safarian B, Esparham A, Mirzaei M, Esmaeilzadeh M, Boskabady MH. The modulatory effects of exercise on lipopolysaccharide-induced lung inflammation and injury: A systemic review. Life Sci 2022; 293:120306.