116
Views
3
CrossRef citations to date
0
Altmetric
REVIEW

Therapeutic Benefits of Mesenchymal Stem Cells in Acute Respiratory Distress Syndrome: Potential Mechanisms and Challenges

, , & ORCID Icon
Pages 5235-5246 | Published online: 10 Sep 2022

References

  • Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377(6):562–572. doi:10.1056/NEJMra1608077
  • Ashbaugh DG, Bigelow DB, Petty TL, Levine BE. Acute respiratory distress in adults. Lancet. 1967;2(7511):319–323. doi:10.1016/s0140-6736(67
  • Ranieri VM, Rubenfeld GD, Thompson B, et al.; ARDS Definition Task Force. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307(23):2526–2533. doi:10.1001/jama.
  • Meyer NJ. Future clinical applications of genomics for acute respiratory distress syndrome. Lancet Respir Med. 2013;1(10):793–803. doi:10.1016/S2213-2600(13
  • Bellani G, Laffey JG, Pham T, et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315(8):788–800. doi:10.1001/jama.2016.0291
  • Moss M, Huang DT, et al.; National Heart, Lung, and Blood Institute PETAL Clinical Trials Network. Early neuromuscular blockade in the acute respiratory distress syndrome. N Engl J Med. 2019;380(21):1997–2008. doi:10.1056/NEJMoa1901686.
  • Meyer NJ, Gattinoni L, Calfee CS. Acute respiratory distress syndrome. Lancet. 2021;398(10300):622–637. doi:10.1016/S0140-6736(21
  • Meyer NJ, Calfee CS. Novel translational approaches to the search for precision therapies for acute respiratory distress syndrome. Lancet Respir Med. 2017;5(6):512–523. doi:10.1016/S2213-2600(17
  • Casey JD, Semler MW, Rice TW. Fluid management in acute respiratory distress syndrome. Semin Respir Crit Care Med. 2019;40(1):57–65. doi:10.1055/s-0039-1685206
  • Beitler JR, Thompson BT, Baron RM, et al. Advancing precision medicine for acute respiratory distress syndrome. Lancet Respir Med. 2022;10(1):107–120. doi:10.1016/S2213-2600(21)00157-0
  • Fan E, Brodie D, Slutsky AS. Acute respiratory distress syndrome: advances in diagnosis and treatment. JAMA. 2018;319(7):698–710. doi:10.1001/jama.2017.21907
  • Matthay MA, Zemans RL, Zimmerman GA, et al. Acute respiratory distress syndrome. Nat Rev Dis Primers. 2019;5(1):18. doi:10.1038/s41572-019-0069-0
  • Ervin JN, Rentes VC, Dibble ER, et al. Evidence-based practices for acute respiratory failure and acute respiratory distress syndrome: a systematic review of reviews. Chest. 2020;158(6):2381–2393. doi:10.1016/j.chest.2020.06.080
  • Sweeney RM, McAuley DF. Acute respiratory distress syndrome. Lancet. 2016;388(10058):2416–2430. doi:10.1016/S0140-6736(16
  • Wyncoll DL, Evans TW. Acute respiratory distress syndrome. Lancet. 1999;354(9177):497–501. doi:10.1016/S0140-6736(98
  • Matthay MA. ECMO in severe acute respiratory distress syndrome. Lancet Respir Med. 2019;7(2):106–108. doi:10.1016/S2213-2600(18
  • Opitz B, van Laak V, Eitel J, Suttorp N. Innate immune recognition in infectious and noninfectious diseases of the lung. Am J Respir Crit Care Med. 2010;181(12):1294–1309. doi:10.1164/rccm.200909-1427SO
  • Weiss DJ. Cell-based therapies for acute respiratory distress syndrome. Lancet Respir Med. 2019;7(2):105–106. doi:10.1016/S2213-2600(18
  • Curley GF, Scott JA, Laffey JG. Therapeutic potential and mechanisms of action of mesenchymal stromal cells for acute respiratory distress syndrome. Curr Stem Cell Res Ther. 2014;9(4):319–329. doi:10.2174/1574888x09666140228144812
  • Matthay MA, McAuley DF, Ware LB. Clinical trials in acute respiratory distress syndrome: challenges and opportunities. Lancet Respir Med. 2017;5(6):524–534. doi:10.1016/S2213-2600(17
  • Lanzoni G, Linetsky E, Correa D, et al. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: a double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl Med. 2021;10(5):660–673. doi:10.1002/sctm.20-0472
  • Walter J, Ware LB, Matthay MA. Mesenchymal stem cells: mechanisms of potential therapeutic benefit in ARDS and sepsis. Lancet Respir Med. 2014;2(12):1016–1026. doi:10.1016/S2213-2600(14
  • Lee JW, Fang X, Gupta N, Serikov V, Matthay MA. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106(38):16357–16362. doi:10.1073/pnas.0907996106
  • Wilson JG, Liu KD, Zhuo H, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3(1):24–32. doi:10.1016/S2213-2600(14)70291-7
  • Matthay MA, Calfee CS, Zhuo H, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised Phase 2a safety trial. Lancet Respir Med. 2019;7(2):154–162. doi:10.1016/S2213-2600(18)30418-1
  • Sipp D, Robey PG, Turner L. Clear up this stem-cell mess. Nature. 2018;561(7724):455–457. doi:10.1038/d41586-018-06756-9
  • Galipeau J, Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824–833. doi:10.1016/j.stem.2018.05.004
  • Nombela-Arrieta C, Ritz J, Silberstein LE. The elusive nature and function of mesenchymal stem cells. Nat Rev Mol Cell Biol. 2011;12(2):126–131. doi:10.1038/nrm3049
  • Horwitz EM, Le Blanc K, Dominici M, et al. Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement. Cytotherapy. 2005;7(5):393–395. doi:10.1080/14653240500319234
  • Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–317. doi:10.1080/14653240600855905
  • McIntosh KR. Evaluation of cellular and humoral immune responses to allogeneic adipose-derived stem/stromal cells. Methods Mol Biol. 2011;702:133–150. doi:10.1007/978-1-61737-960-4_11
  • Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25(11):2739–2749. doi:10.1634/stemcells.2007-0197
  • Weiss DJ, Bertoncello I, Borok Z, et al. Stem cells and cell therapies in lung biology and lung diseases. Proc Am Thorac Soc. 2011;8(3):223–272. doi:10.1513/pats.201012-071DW
  • Lee JW, Fang X, Krasnodembskaya A, Howard JP, Matthay MA. Concise review: mesenchymal stem cells for acute lung injury: role of paracrine soluble factors. Stem Cells. 2011;29(6):913–919. doi:10.1002/stem.643
  • Mei SH, Haitsma JJ, Dos Santos CC, et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med. 2010;182(8):1047–1057. doi:10.1164/rccm.201001-0010OC
  • Rocheteau P, Chatre L, Briand D, et al. Sepsis induces long-term metabolic and mitochondrial muscle stem cell dysfunction amenable by mesenchymal stem cell therapy. Nat Commun. 2015;6:10145. doi:10.1038/ncomms10145
  • Fotino C, Ricordi C, Lauriola V, Alejandro R, Pileggi A. Bone marrow-derived stem cell transplantation for the treatment of insulin-dependent diabetes. Rev Diabet Stud. 2010;7(2):144–157. doi:10.1900/RDS.2010.7.144
  • Hare JM, Fishman JE, Gerstenblith G, et al. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308(22):2369–2379. doi:10.1001/jama.2012.25321
  • Shi M, Zhang Z, Xu R, et al. Human mesenchymal stem cell transfusion is safe and improves liver function in acute-on-chronic liver failure patients. Stem Cells Transl Med. 2012;1(10):725–731. doi:10.5966/sctm.2012-0034
  • Tan J, Wu W, Xu X, et al. Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: a randomized controlled trial. JAMA. 2012;307(11):1169–1177. doi:10.1001/jama.2012.316
  • Uccelli A, Laroni A, Ali R, et al. Safety, tolerability, and activity of mesenchymal stem cells versus placebo in multiple sclerosis (MESEMS): a phase 2, randomised, double-blind crossover trial. Lancet Neurol. 2021;20(11):917–929. doi:10.1016/S1474-4422(21)00301-X
  • Xiao Z, Tang F, Zhao Y, et al. Significant improvement of acute complete spinal cord injury patients diagnosed by a combined criteria implanted with NeuroRegen scaffolds and mesenchymal stem cells. Cell Transplant. 2018;27(6):907–915. doi:10.1177/0963689718766279
  • Ciccocioppo R, Bernardo ME, Sgarella A, et al. Autologous bone marrow-derived mesenchymal stromal cells in the treatment of fistulising Crohn’s disease. Gut. 2011;60(6):788–798. doi:10.1136/gut.2010.214841
  • Le Blanc K, Frassoni F, Ball L, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371(9624):1579–1586. doi:10.1016/S0140-6736(08)60690-X
  • Mushahary D, Spittler A, Kasper C, Weber V, Charwat V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A. 2018;93(1):19–31. doi:10.1002/cyto.a.23242
  • Le Blanc K, Mougiakakos D. Multipotent mesenchymal stromal cells and the innate immune system. Nat Rev Immunol. 2012;12(5):383–396. doi:10.1038/nri3209
  • Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant. 2011;20(1):5–14. doi:10.3727/096368910X
  • Jin HJ, Bae YK, Kim M, et al. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013;14(9):17986–18001. doi:10.3390/ijms140917986
  • Estes BT, Diekman BO, Gimble JM, Guilak F. Isolation of adipose-derived stem cells and their induction to a chondrogenic phenotype. Nat Protoc. 2010;5(7):1294–1311. doi:10.1038/nprot.2010.81
  • McIntyre LA, Moher D, Fergusson DA, et al. Efficacy of mesenchymal stromal cell therapy for acute lung injury in preclinical animal models: a systematic review. PLoS One. 2016;11(1):e0147170. doi:10.1371/journal.pone.0147170
  • Zheng G, Huang L, Tong H, et al. Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res. 2014;15(1):39. doi:10.1186/1465-9921-15-39
  • Mahmoudifar N, Doran PM. Mesenchymal stem cells derived from human adipose tissue. Methods Mol Biol. 2015;1340:53–64. doi:10.1007/978-1-4939-2938-2_4
  • Wagner W, Wein F, Seckinger A, et al. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol. 2005;33(11):1402–1416. doi:10.1016/j.exphem.2005.07.003
  • Sabol RA, Bowles AC, Côté A, Wise R, Pashos N, Bunnell BA. Therapeutic potential of adipose stem cells. Adv Exp Med Biol. 2021;1341:15–25. doi:10.1007/5584_2018_248
  • Antunes MA, Abreu SC, Cruz FF, et al. Effects of different mesenchymal stromal cell sources and delivery routes in experimental emphysema. Respir Res. 2014;15(1):118. doi:10.1186/s12931-014-0118-x
  • Russell KC, Lacey MR, Gilliam JK, Tucker HA, Phinney DG, O’Connor KC. Clonal analysis of the proliferation potential of human bone marrow mesenchymal stem cells as a function of potency. Biotechnol Bioeng. 2011;108(11):2716–2726. doi:10.1002/bit.23193
  • Antebi B, Walker KP, Mohammadipoor A, et al. The effect of acute respiratory distress syndrome on bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2018;9(1):251. doi:10.1186/s13287-018-0981-3
  • Danisovic L, Varga I, Polák S, et al. Comparison of in vitro chondrogenic potential of human mesenchymal stem cells derived from bone marrow and adipose tissue. Gen Physiol Biophys. 2009;28(1):56–62.
  • Afflerbach AK, Kiri MD, Detinis T, Maoz BM. Mesenchymal stem cells as a promising cell source for integration in novel in vitro models. Biomolecules. 2020;10(9):1306. doi:10.3390/biom10091306
  • Bloor AJC, Patel A, Griffin JE, et al. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: a Phase I, multicenter, open-label, dose-escalation study. Nat Med. 2020;26(11):1720–1725. doi:10.1038/s41591-020-1050-x
  • Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med. 2000;342(18):1334–1349. doi:10.1056/NEJM200005043421806
  • Ortiz LA, Dutreil M, Fattman C, et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci U S A. 2007;104(26):11002–11007. doi:10.1073/pnas.0704421104
  • Danchuk S, Ylostalo JH, Hossain F, et al. Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-α-induced protein 6. Stem Cell Res Ther. 2011;2(3):27. doi:10.1186/scrt68
  • Ionescu L, Byrne RN, van Haaften T, et al. Stem cell conditioned medium improves acute lung injury in mice: in vivo evidence for stem cell paracrine action. Am J Physiol Lung Cell Mol Physiol. 2012;303(11):L967–977. doi:10.1152/ajplung.00144.2011
  • Wang L, Li Y, Xu M, et al. Regulation of inflammatory cytokine storms by mesenchymal stem cells. Front Immunol. 2021;12:726909. doi:10.3389/fimmu.2021.726909
  • Wick KD, Leligdowicz A, Zhuo H, Ware LB, Matthay MA. Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight. 2021;6(12):e148983. doi:10.1172/jci.insight.148983
  • Millar JE, von Bahr V, Malfertheiner MV, et al. Administration of mesenchymal stem cells during ECMO results in a rapid decline in oxygenator performance. Thorax. 2019;74(2):194–196. doi:10.1136/thoraxjnl-2017-211439
  • Matthay MA. Therapeutic potential of mesenchymal stromal cells for acute respiratory distress syndrome. Ann Am Thorac Soc. 2015;Suppl 1(Suppl1):S54–57. doi:10.1513/AnnalsATS.201406-254MG
  • Chinnadurai R, Copland IB, Garcia MA, et al. Cryopreserved mesenchymal stromal cells are susceptible to T-cell mediated apoptosis which is partly rescued by IFNγ licensing. Stem Cells. 2016;34(9):2429–2442. doi:10.1002/stem.2415
  • Galleu A, Riffo-Vasquez Y, Trento C, et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation. Sci Transl Med. 2017;9(416):eaam7828. doi:10.1126/scitranslmed.aam7828
  • Bhattacharya J, Matthay MA. Regulation and repair of the alveolar-capillary barrier in acute lung injury. Annu Rev Physiol. 2013;75:593–615. doi:10.1146/annurev-physiol-030212-183756
  • Byrnes D, Masterson CH, Artigas A, Laffey JG. Mesenchymal stem/stromal cells therapy for sepsis and acute respiratory distress syndrome. Semin Respir Crit Care Med. 2021;42(1):20–39. doi:10.1055/s-0040-1713422
  • Wang F, Fang B, Qiang X, Shao J, Zhou L. The efficacy of mesenchymal stromal cell-derived therapies for acute respiratory distress syndrome-a meta-analysis of preclinical trials. Respir Res. 2020;21(1):307. doi:10.1186/s12931-020-01574-y
  • Zhang X, Chen J, Xue M, et al. Overexpressing p130/E2F4 in mesenchymal stem cells facilitates the repair of injured alveolar epithelial cells in LPS-induced ARDS mice. Stem Cell Res Ther. 2019;10(1):74. doi:10.1186/s13287-019-1169-1
  • Dutra Silva J, Su Y, Calfee CS, et al. Mesenchymal stromal cell extracellular vesicles rescue mitochondrial dysfunction and improve barrier integrity in clinically relevant models of ARDS. Eur Respir J. 2021;58(1):2002978. doi:10.1183/13993003.02978-2020
  • Marshall R, Bellingan G, Laurent G. The acute respiratory distress syndrome: fibrosis in the fast lane. Thorax. 1998t;53(10):815–817. doi:10.1136/thx.53.10.815
  • Jung YJ, Park YY, Huh JW, Hong SB. The effect of human adipose-derived stem cells on lipopolysaccharide-induced acute respiratory distress syndrome in mice. Ann Transl Med. 2019;7(22):674. doi:10.21037/atm.2019.10.48
  • Moroncini G, Paolini C, Orlando F, et al. Mesenchymal stromal cells from human umbilical cord prevent the development of lung fibrosis in immunocompetent mice. PLoS One. 2018;13(6):e0196048. doi:10.1371/journal.pone.0196048
  • Shen T, Xia L, Dong W, et al. A systematic review and meta-analysis: safety and efficacy of mesenchymal stem cells therapy for heart failure. Curr Stem Cell Res Ther. 2021;16(3):354–365. doi:10.2174/1574888X15999200820171432
  • Chen T, Yang T, Zhang W, Shao J. The therapeutic potential of mesenchymal stem cells in treating osteoporosis. Biol Res. 2021;54(1):42. doi:10.1186/s40659-021-00366-y
  • Nikitina V, Astrelina T, Nugis V, et al. Clonal chromosomal and genomic instability during human multipotent mesenchymal stromal cells long-term culture. PLoS One. 2018;13(2):e0192445. doi:10.1371/journal.pone
  • Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell. 2015;17(1):11–22. doi:10.1016/j.stem.2015.06.007
  • Kaffash Farkhad N, Sedaghat A, Reihani H, et al. Mesenchymal stromal cell therapy for COVID-19-induced ARDS patients: a successful phase 1, control-placebo group, clinical trial. Stem Cell Res Ther. 2022;13(1):283. doi:10.1186/s13287-022-02920-1
  • Monsel A, Hauw-Berlemont C, Mebarki M, et al. Treatment of COVID-19-associated ARDS with mesenchymal stromal cells: a multicenter randomized double-blind trial. Crit Care. 2022;26(1):48. doi:10.1186/s13054-022-03930-4
  • Ahn SY, Chang YS, Kim JH, Sung SI, Park WS. Two-Year Follow-Up Outcomes of Premature Infants Enrolled in the Phase I Trial of Mesenchymal Stem Cells Transplantation for Bronchopulmonary Dysplasia. J Pediatr. 2017;185:49–54. doi:10.1016/j.jpeds.2017.02.061
  • Ahn SY, Chang YS, Lee MH, et al. Stem cells for bronchopulmonary dysplasia in preterm infants: a randomized controlled phase II trial. Stem Cells Transl Med. 2021;10(8):1129–1137. doi:10.1002/sctm.20-0330
  • Cruz FF, Weiss DJ, Rocco PR. Prospects and progress in cell therapy for acute respiratory distress syndrome. Expert Opin Biol Ther. 2016;16(11):1353–1360. doi:10.1080/14712598.2016.1218845
  • Antebi B, Mohammadipoor A, Batchinsky AI, Cancio LC. The promise of mesenchymal stem cell therapy for acute respiratory distress syndrome. J Trauma Acute Care Surg. 2018;84(1):183–191. doi:10.1097/TA.0000000000001713
  • Donders R, Bogie JFJ, Ravanidis S, et al. Human wharton’s jelly-derived stem cells display a distinct immunomodulatory and proregenerative transcriptional signature compared to bone marrow-derived stem cells. Stem Cells Dev. 2018;27(2):65–84. doi:10.1089/scd.2017.0029
  • Silva JD, Lopes-Pacheco M, Paz AHR, et al. Mesenchymal stem cells from bone marrow, adipose tissue, and lung tissue differentially mitigate lung and distal organ damage in experimental acute respiratory distress syndrome. Crit Care Med. 2018;46(2):e132–e140. doi:10.1097/CCM.0000000000002833
  • Ren H, Zhang Q, Wang J, Pan R. Comparative effects of umbilical cord- and menstrual blood-derived MSCs in repairing acute lung injury. Stem Cells Int. 2018;2018:7873625. doi:10.1155/2018/7873625
  • Asmussen S, Ito H, Traber DL, et al. Human mesenchymal stem cells reduce the severity of acute lung injury in a sheep model of bacterial pneumonia. Thorax. 2014;69(9):819–825. doi:10.1136/thoraxjnl-2013-204980
  • Devaney J, Horie S, Masterson C, et al. Human mesenchymal stromal cells decrease the severity of acute lung injury induced by E. coli in the rat. Thorax. 2015;70(7):625–635. doi:10.1136/thoraxjnl-2015-206813
  • Qin H, Zhao A. Mesenchymal stem cell therapy for acute respiratory distress syndrome: from basic to clinics. Protein Cell. 2020;11(10):707–722. doi:10.1007/s13238-020-00738-2
  • Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. Mesenchymal stem cell secretome: toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci. 2017;18(9):1852. doi:10.3390/ijms18091852
  • de Witte SFH, Luk F, Sierra Parraga JM, et al. Immunomodulation by therapeutic mesenchymal stromal cells (MSC) is triggered through phagocytosis of MSC by monocytic cells. Stem Cells. 2018;36(4):602–615. doi:10.1002/stem.2779
  • Tang XD, Shi L, Monsel A, et al. Mesenchymal stem cell microvesicles attenuate acute lung injury in mice partly mediated by ang-1 mRNA. Stem Cells. 2017;35(7):1849–1859. doi:10.1002/stem.2619
  • Curley GF, Hayes M, Ansari B, et al. Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat. Thorax. 2012;67(6):496–501. doi:10.1136/thoraxjnl-2011-201059
  • Witwer KW, Van Balkom BWM, Bruno S, et al. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. J Extracell Vesicles. 2019;8(1):1609206. doi:10.1080/20013078.2019.1609206
  • Park KS, Svennerholm K, Shelke GV, et al. Mesenchymal stromal cell-derived nanovesicles ameliorate bacterial outer membrane vesicle-induced sepsis via IL-10. Stem Cell Res Ther. 2019;10(1):231. doi:10.1186/s13287-019-1352-4
  • Monsel A, Zhu YG, Gennai S, et al. Therapeutic effects of human mesenchymal stem cell-derived microvesicles in severe pneumonia in mice. Am J Respir Crit Care Med. 2015;192(3):324–336. doi:10.1164/rccm.201410-1765OC
  • Gorman E, Millar J, McAuley D, O’Kane C. Mesenchymal stromal cells for acute respiratory distress syndrome (ARDS), sepsis, and COVID-19 infection: optimizing the therapeutic potential. Expert Rev Respir Med. 2021;15(3):301–324. doi:10.1080/17476348.2021.1848555
  • Lu H, Cook T, Poirier C, et al. Pulmonary retention of adipose stromal cells following intravenous delivery is markedly altered in the presence of ARDS. Cell Transplant. 2016;25(9):1635–1643. doi:10.3727/096368915X690189
  • Dunbar H, Weiss DJ, Rolandsson Enes S, Laffey JG, English K. The Inflammatory Lung Microenvironment; a Key Mediator in MSC Licensing. Cells. 2021;10(11):2982. doi:10.3390/cells10112982
  • Rolandsson Enes S, Hampton TH, Barua J, et al. Healthy versus inflamed lung environments differentially affect mesenchymal stromal cells. Eur Respir J. 2021;58(4):2004149. doi:10.1183/13993003.04149-2020
  • Islam D, Huang Y, Fanelli V, et al. Identification and modulation of microenvironment is crucial for effective mesenchymal stromal cell therapy in acute lung injury. Am J Respir Crit Care Med. 2019;199(10):1214–1224. doi:10.1164/rccm.201802-0356OC
  • Cogle CR, Guthrie SM, Sanders RC, et al. An overview of stem cell research and regulatory issues. Mayo Clin Proc. 2003;78(8):993–1003. doi:10.4065/78.8.993