key: cord-0827263-2ewmhwu9 authors: Su, Yue; Guo, Haiyan; Liu, Qinghua title: Effects of mesenchymal stromal cell‐derived extracellular vesicles in acute respiratory distress syndrome (ARDS): Current understanding and future perspectives date: 2021-05-06 journal: J Leukoc Biol DOI: 10.1002/jlb.3mr0321-545rr sha: 9db80b8852e6e476302e5a170ab5dfc5812dcec8 doc_id: 827263 cord_uid: 2ewmhwu9 Acute respiratory distress syndrome (ARDS) is a devastating and life‐threatening syndrome that results in high morbidity and mortality. Current pharmacologic treatments and mechanical ventilation have limited value in targeting the underlying pathophysiology of ARDS. Mesenchymal stromal cells (MSCs) have shown potent therapeutic advantages in experimental and clinical trials through direct cell‐to‐cell interaction and paracrine signaling. However, safety concerns and the indeterminate effects of MSCs have resulted in the investigation of MSC‐derived extracellular vesicles (MSC‐EVs) due to their low immunogenicity and tumorigenicity. Over the past decades, soluble proteins, microRNAs, and organelles packaged in EVs have been identified as efficacious molecules to orchestrate nearby immune responses, which attenuate acute lung injury by facilitating pulmonary epithelium repair, reducing acute inflammation, and restoring pulmonary vascular leakage. Even though MSC‐EVs possess similar bio‐functional effects to their parental cells, there remains existing barriers to employing this alternative from bench to bedside. Here, we summarize the current established research in respect of molecular mechanisms of MSC‐EV effects in ARDS and highlight the future challenges of MSC‐EVs for clinical application. to possess the properties of immunomodulation and tissue repair in both experimental acute lung injury (ALI) and sepsis models through the secretion of several growth factors (TNF-α-stimulated gene-6), KGF (keratinocyte growth factor), prostaglandin E2, et al.), and antiinflammatory cytokines (IL-10). [5] [6] [7] Moreover, in an ex vivo model of perfused human lungs injured with live E. coli bacteria, MSCs were also capable of decreasing lung inflammation, clearing alveolar fluid, and enhancing alveolar macrophage phagocytosis. 8 Despite the existence of data that have shown the benefit of MSCs for ARDS, a growing body of evidence is emerging querying the actual effects of MSCs in clinical practice in terms of the outcome of several completed randomized phase I/II clinical trials [9] [10] [11] [12] (Table 1) . Although these early clinical trials have shown that MSCs did not induce prespecified infusion-related adverse events, there was no difference in 28 d mortality between the groups of MSCs and placebo, and the MSC group had higher mean scores of Acute Physiology and Chronic Health Evaluation III, which can provide initial risk stratification and risk estimates for critically ill patients, and higher scores correspond to more severe illness and a higher risk of death. 10, 11 Additionally, it has been proven that the efficacy of MSC engraftment and differentiation is limited 13 and MSCs are short-lived cells, which are no longer viable after 24 h in the damaged lung by i.v. infusion. [14] [15] [16] Of more concern is the fact that MSCs exhibit a high risk of carcinogenesis, and the possibility of tumorigenicity increases when MSCs are expanded in culture, whereas the longterm follow-up of MSC administration is absent in most preclinical experiments and clinical trials. 17, 18 Recently, attention has been drawn to MSC-derived extracellular vesicles (MSC-EVs) as a new frontier in the cell-free treatment regime for ARDS. There are increasing data to suggest that the properties of MSC-EVs are similar to their parental cells in anti-inflammation and tissue homeostasis in damaged cells or diseased organs. Furthermore, the characteristics of no risk of tumorigenicity, and a lower possibility of immunologic rejection and self-replication, make them a promising candidate for the treatment of ARDS. 19 EVs are lipid bilayer-surrounded particles, comprising various subpopulations of released cells, which participate in multiple physiologic and pathologic activities to facilitate intercellular communications and to change the biologic components of recipient cells. 20 MSC-EVs have been demonstrated to have therapeutic benefits for ARDS and severe pneumonia in preclinical studies. The main biofunctionalized benefits of MSC-EVs have been presented in the aspect of attenuating acute inflammation, promoting alveolar epithe-lial regeneration, and enhancing pulmonary endothelial repair. 23 In this context, accumulating evidence aiming at dissecting molecular mechanisms has shown that MSC-EVs as a shuttle of bio-active messengers is capable of wrapping mRNAs, proteins, microRNA (miRNAs), and mitochondria to modulate immune responses and to repair widespread lung damage in the exudative, proliferative, and fibrotic phases of ARDS, which assist in decreasing proinflammatory cytokine production and improving alveolar fluid clearance 12 (Fig. 2 ). The mRNA is a single-stranded molecule that carries genetic information copied from DNA for protein synthesis. 24 Batagov and colleagues showed that most exosomal mRNAs that are enriched with specific 3′-untranslated regions that may present as a competing RNA in targeted cells to modulate the physiologic processes. 25 (1) mRNA: MSC-EVs deliver mRNA fragments of keratinocyte growth factor (KGF) and angiopoietin 1 (Ang-1) have shown great therapeutic effects in decreasing neutrophil infiltration, increasing anti-inflammatory cytokine production, down-regulating proinflammatory cytokine secretion, total protein, and vascular endothelial permeability. (2) miRNA: MSC-EVs are capable of transferring miRNA21, miRNA27a-3p, miR145, and miR146 to alveolar macrophages, contributing to M2 macrophage polarization, STAT3 signaling activation, macrophage phagocytosis promotion, and down-regulate IL-6 secretion. Furthermore, miR100 can be delivered to fibroblast cells by MSC-EVs, resulting in PI3K/protein kinase B (AKT)/NF-kB signaling activation, down-regulation of total protein content, neutrophil counts, and proinflammatory levels. The miRNAs are a class of small noncoding RNAs that are transcribed from DNA sequences, normally 22 nucleotides in length, 33 which exert a critical impact on various biologic activities. Mechanically, miRNAs secreted into EVs or fluids are able to interact with targeted mRNAs, thereby suppressing protein translation. However, in the context of several specific microenvironments, miRNAs also promote or modulate mRNA translation. Accumulating evidence has shown that MSC-EVs transfer miR-21p, miR-27p-3a, miR30b-3p, miR100, miR145, and miR146 to injured or inflamed lung tissue for regulating their biogenesis and homeostasis. MiR21 is one of most investigated and responsive miRNAs in the context of distinct pathologic processes, including acute or chronic lung inflammation, diabetic complications, and myocardial ischemia. It has been demonstrated that miR21 is able to down-regulate STAT3 signaling to suppress LPS-induced ALI by decreasing the IL-6 secretion in murine RAW264.7 cells. 34 In what could be another mechanism for miR21 biologic function in lung disease, miR21 facilitates the pro- Most studies into miR100 in lung diseases concern its antitumor effects and its role as a potential molecular prognostic marker in nonsmall lung cancer. 44, 45 Recently, miR100 has been demonstrated to modulate Additionally, miR100 overexpressed MSC-EVs attenuate the total protein content, neutrophil counts, and the proinflammatory levels in the BALF, and decrease cell apoptosis in the BLM-induced ALI rat model. 47 MiR145 is engaged in normal blood cell development and appears to play a fundamental role in the biogenesis and division of megakaryocytes. 48 Regarding lung diseases, miR145 has been reported to contribute to the pathogenesis of hypoxia-induced pulmonary artery hypertension, and miR145 deficiency has also modulated myofibroblast differentiation to protect BLM-treated lung fibrosis. 49 The miR146 family, consisting of miR146a and miR146b, has been shown to be a well-documented anti-inflammatory miRNA and a neg- Mitochondria, referred to as the powerhouse of the cell, are believed to exert a vital impact on the pathologic processes of the illness by regulating cell metabolism and homeostasis. Optimal performance of cel-lular mitochondria relies on the sophisticated and dynamic system that provides a complete electron transport chain (ETC), an efficient citric acid cycle, and intact mitochondria constituents. Conversely, damaged ETC, excessive superoxide-derived reactive oxygen species production, unbalanced cytosolic and mitochondrial calcium levels, and morphologic changes (mitochondria leak and uncoupling) contribute to mitochondrial dysfunction and cell injury. 59, 60 Of note, mitochondrial bioenergetic failure also plays a key role in the pathogenesis of ARDS. 61 Ever-increasing evidence has suggested mitochondrial dyshomeostasis is implicated in ARDS development and pathophysiology. 62 A preclinical study of ARDS has revealed that significant ATP decline and low arterial oxygenation have been shown in the LPStreated mouse model, which are considered to be associated with prolyl hydroxylases and hypoxia-inducible factor. 63 The preliminary data from Ten et al. have demonstrated that ADP-phosphorylating respiration in mitochondria isolated from mouse injured lungs that were treated with LPS is lower than the control group. 64 To the best of our knowledge, Spees and coworkers have initially A novel human coronavirus, named SARS-CoV-2, was found in some severe pneumonia cases in early December 2019, and within several months had caused a pandemic of respiratory illness termed This epidemic is now spreading in over 200 countries and territories around the world through human-to-human transmission. Recently, a global literature survey has revealed that of hospitalized COVID-19 patients, approximately one-third (33%) develop ARDS, one-fourth (26%) need to be transferred into ICU, and onesixth (16%) cases die in the hospital. 71 Previous studies, including clinical trials, have shown that MSCs or MSC-EVs are beneficial to H1N1, H5N1, H9N2, or H7N9-induced ALI or pneumonia, presenting a potent effect on decreasing proinflammatory cytokine secretion and suppressing immune cell recruitment in the lungs [72] [73] [74] [75] [76] [77] (Table 2) . Due to there being no precise and specific antiviral medicine for this emerging illness, MSCs were applied in several critically ill COVID-19 patients, contributing to the decline of plasma C reactive protein (CRP), aspartic aminotransferase, creatine kinase activity, and myoglobin. Furthermore, combined with symptomatic and supportive treatment, chest CT manifestation and SARS-CoV-2 nucleic acid detection improved markedly or returned to negative after 2 wk. [78] [79] [80] [81] Although the positive results mentioned earlier supported the admin- and neutrophil count decreased, whereas the average CD3+, CD4+, and CD8+ lymphocyte count increased. However, the paper did not mention how the ExoFlo was developed and manufactured, and there appears to be no difference in mortality between this cohort and that of the global survey (Table 3) . To date, even though several clinical trials are ongoing to determine the effects of MSC-EVs for COVID19, the International Society of Cell and Gene Therapy and ISEV strongly urge that the decision to apply of MSC-EVs for COVID should be taken carefully due to the limited preclinical and clinical data, and they also sug-gest any use of EVs should be carefully evaluated through rational clinical trial design. 83 The The authors declare no conflicts of interest. Acute respiratory distress syndrome The acute respiratory distress syndrome: pathophysiology, current clinical practice, and emerging therapies Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries Acute respiratory distress syndrome: Advances in diagnosis and treatment Mesenchymal stromal cells and macrophages in sepsis: new insights Therapeutic potential of mesenchymal stromal cells in the treatment of ARDS. Transfusion Mesenchymal stem cells: mechanisms of potential therapeutic benefit in ARDS and sepsis Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial Primary analysis of a phase 1/2 study to assess multistem cell therapy, a regenerative advanced therapy medicinal product (ATMP), in acute respiratory distress syndrome (MUST-ARDS). B14. Late Break Clin Trials Paracrine factors of multipotent stromal cells ameliorate lung injury in an elastase-induced emphysema model Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion The life and fate of mesenchymal stem cells Mesenchymal stromal (stem) cell therapy fails to improve outcomes in experimental severe influenza Concise review: dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth Defining the risks of mesenchymal stromal cell therapy Exosomes derived from mesenchymal stem cells Shedding light on the cell biology of extracellular vesicles Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases Extracellular vesicles: exosomes, microvesicles, and friends Mesenchymal stem cells-derived extracellular vesicles in acute respiratory distress syndrome: a review of current literature and potential future treatment options The roles of RNA in the synthesis of protein Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3′-untranslated regions Human KGF is FGFrelated with properties of a paracrine effector of epithelial cell growth Inducible expression of keratinocyte growth factor (KGF) in mice inhibits lung epithelial cell death induced by hyperoxia Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice Signaling and functions of angiopoietin-1 in vascular protection Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin Mesenchymal stem cell-based angiopoietin-1 gene therapy for acute lung injury induced by lipopolysaccharide in mice Mesenchymal stem cell microvesicles attenuate acute lung injury in mice partly mediated by Ang-1 mRNA Overview of microRNA biogenesis, mechanisms of actions, and circulation Liang-Ge-San, a classic traditional chinese medicine formula, attenuates lipopolysaccharide-induced acute lung injury through up-regulating miR-21 Long non-coding RNA NKILA weakens TNF-α-induced inflammation of MRC-5 cells by miR-21 upregulation Mesenchymal stromal cells-derived exosomes alleviate ischemia/reperfusion injury in mouse lung by transporting anti-apoptotic miR-21-5p MicroRNA-27a/b controls endothelial cell repulsion and angiogenesis by targeting semaphorin 6A Regulation of angiogenesis and choroidal neovascularization by members of microRNA-23∼27∼24 clusters Paclitaxel alleviated liver injury of septic mice by alleviating inflammatory response via microRNA-27a/TAB3/NF-κB signaling pathway Mesenchymal stem cell-derived extracellular vesicles alleviate acute lung injury via transfer of miR-27a-3p * The miR-30 family inhibits pulmonary vascular hyperpermeability in the premetastatic phase by direct targeting of Skp2 Inhibition and role of let-7d in idiopathic pulmonary fibrosis Exosomes derived from microRNA-30b-3p-overexpressing mesenchymal stem cells protect against lipopolysaccharide-induced acute lung injury by inhibiting SAA3 MicroRNA-100 is a potential molecular marker of non-small cell lung cancer and functions as a tumor suppressor by targeting polo-like kinase 1 Overexpression of miR-100 inhibits cell proliferation, migration, and chemosensitivity in human glioblastoma through FGFR3 Long noncoding RNA HAGLROS regulates cell apoptosis and autophagy in lipopolysaccharides-induced WI-38 cells via modulating miR-100/NF-κB axis Microvesicles derived from human Wharton's jelly mesenchymal stem cells enhance autophagy and ameliorate acute lung injury via delivery of miR-100 MicroRNAs in erythroid and megakaryocytic differentiation and megakaryocyte-erythroid progenitor lineage commitment A role for miR-145 in pulmonary arterial hypertension: evidence from mouse models and patient samples MiR-145 regulates myofibroblast differentiation and lung fibrosis MiR-145 negatively regulates TGFBR2 signaling responsible for sepsis-induced acute lung injury Mesenchymal stem cellderived extracellular vesicles decrease lung injury in mice Rapid changes in microRNA-146a expression negatively regulate the IL-1β-induced inflammatory response in human lung alveolar epithelial cells MicroRNA-146a and microRNA-146b expression and anti-inflammatory function in human airway smooth muscle Pharmacological studies of the mechanism and function of interleukin-1β-induced miRNA-146a expression in primary human airway smooth muscle Combined effects of PVT1 and MiR-146a single nucleotide polymorphism on the lung function of smokers with chronic obstructive pulmonary disease GSKJ4 protects mice against early sepsis via reducing proinflammatory factors and up-regulating MiR-146a Exosomal miR-146a contributes to the enhanced therapeutic efficacy of interleukin-1β-primed mesenchymal stem cells against sepsis. Stem Cells Mitochondria and Critical Illness Mitochondria in lung disease Polydatin mediates Parkin-dependent mitophagy and protects against mitochondria-dependent apoptosis in acute respiratory distress syndrome Mitochondrial alarmins are tissue mediators of ventilator-induced lung injury and ARDS Enhancement of glycolysis by inhibition of oxygen-sensing prolyl hydroxylases protects alveolar epithelial cells from acute lung injury Mitochondrial bioenergetics and pulmonary dysfunction: current progress and future directions Mitochondrial transfer between cells can rescue aerobic respiration Mitochondrial transfer from bonemarrow-derived stromal cells to pulmonary alveoli protects against acute lung injury Mitochondrial transfer via tunneling nanotubes is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer Hypercapnic acidosis induces mitochondrial dysfunction and impairs the ability of mesenchymal stem cells to promote distal lung epithelial repair COVID-19 infection: origin, transmission, and characteristics of human coronaviruses Incidence of ARDS and outcomes in hospitalized patients with COVID-19: a global literature survey. Crit Care Mesenchymal stromal cell treatment prevents H9N2 avian influenza virus-induced acute lung injury in mice Human mesenchymal stromal cells reduce influenza A H5N1-associated acute lung injury in vitro and in vivo Therapeutic implications of human umbilical cord mesenchymal stromal cells in attenuating influenza A (H5N1) virus-associated acute lung injury Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model Influenza causes prolonged disruption of the alveolar-capillary barrier in mice unresponsive to mesenchymal stem cell therapy Clinical study of mesenchymal stem cell treatment for acute respiratory distress syndrome induced by epidemic influenza A (H7N9) infection : a hint for COVID-19 treatment Transplantation of ACE2-Mesenchymal stem cells improves the outcome of patients with covid-19 pneumonia Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trial Adiposederived mesenchymal stromal cells for the treatment of patients with severe SARS-CoV-2 pneumonia requiring mechanical ventilation. A proof of concept study Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19 International Society for Extracellular Vesicles and International Society for Cell and Gene Therapy statement on extracellular vesicles from mesenchymal stromal cells and other cells: considerations for potential therapeutic agents to suppress coronavirus Marrow stromal cells as stem cells for nonhematopoietic tissues Single-cell RNA-seq highlights heterogeneity in human primary Wharton's jelly mesenchymal stem/stromal cells cultured in vitro Extracellular vesicles derived from different sources of mesenchymal stem cells: therapeutic effects and translational potential Differential effects of extracellular vesicles from aging and young mesenchymal stem cells in acute lung injury Therapeutic efficacy of human mesenchymal stromal cells in the repair of established ventilator induced lung injury in the rat Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria Effects of mesenchymal stromal cell-derived extracellular vesicles in acute respiratory distress syndrome (ARDS): current understanding and future perspectives