Abstract
Acute lung injury (ALI) is a clinically life-threatening form of respiratory failure with a mortality of 30%–40%. Acute respiratory distress syndrome is the aggravated form of ALI. Exosomes are extracellular lipid vesicles ubiquitous in human biofluids with a diameter of 30–150 nm. They can serve as carriers to convey their internal cargo, particularly microRNA (miRNA), to the target cells involved in cellular communication. In disease states, the quantities of exosomes and the cargo generated by cells are altered. These exosomes subsequently function as autocrine or paracrine signals to nearby or distant cells, regulating various pathogenic processes. Moreover, exosomal miRNAs from multiple stem cells can provide therapeutic value for ALI by regulating different signaling pathways. In addition, changes in exosomal miRNAs of biofluids can serve as biomarkers for the early diagnosis of ALI. This study aimed to review the role of exosomal miRNAs produced by different sources participating in various pathological processes of ALI and explore their potential significance in the treatment and diagnosis.
1. Introduction
Acute lung injury (ALI) is one of the most common critical illnesses of the respiratory system. It is an acute hypoxic respiratory insufficiency caused by various direct or indirect causes other than of cardiac origin. Its worsened form is acute respiratory distress syndrome (ARDS).1,2 ARDS was first widely recognized in 1994. The American–European Consensus Conference proposed to diagnose ARDS using four criteria: acute hypoxic episode, bilateral pulmonary infiltrates, pulmonary artery wedge pressure ≤18 mmHg or the absence of clinical manifestations of left atrial hypertension, and a level of the arterial pressure of oxygen/inspiratory fraction of oxygen (PaO2/FiO2) ≤ 200 mmHg.3 ALI was identified using diagnostic criteria similar to those for ARDS except for PaO2/FiO2 ≤ 300 mmHg. In 2012, the Berlin definition revised and supplemented the diagnostic standard for ARDS.4 According to PaO2/FiO2, ARDS was classified as mild (PaO2/FiO2: 200–300 mmHg), moderate (PaO2/FiO2: 100–200 mmHg), or severe (PaO2/FiO2 < 100 mmHg). In the United States, there are approximately 200,000 patients with ALI and 75,000 deaths due to ALI per year.5 Nationwide, there are approximately 3 million cases of ALI per year, which account for 10% of all patients in intensive care units,6 and the mortality rate is 35%–40%.7 ALI is caused by pneumonia, severe infections, trauma, shock, burns, acute pancreatitis, radiation injury, blood transfusion, and other conditions.8 Hyaline membrane formation and widespread pulmonary edema are the pathologic characteristics.9−11 Some patients with severe ALI develop ARDS, which may be accompanied by irreversible pulmonary fibrosis, resulting in pulmonary dysfunction.12 Currently, a new understanding of the pathogenesis of ALI has emerged. The significant imbalance of inflammatory responses in the lungs and whole body is the key theory of the pathogenesis of ALI. The disturbance of the coagulation system, dysregulation of vasoactive chemicals, defective regulation of the alveolar–capillary barrier, and imbalance of oxidative stress and apoptosis are additional pathogenic mechanisms implicated in the regulation.13 Despite decades of research, the primary treatment for ALI is still symptomatic and supportive, and there is no particularly efficient treatment in clinical practice. Drug therapy mainly aims to improve lung inflammation and oxygenation. Commonly used drugs include corticosteroids and inhaled vasodilators.14 Mechanical ventilation is thought to be the only supportive therapy that can enhance the survival rate of patients with ALI.15 However, prolonged mechanical ventilation can result in ventilator-associated lung injury.16,17 To improve a patient’s prognosis and reduce mortality, it is essential to investigate treatments for ALI and markers that can assist in its early diagnosis.
Exosomes are extracellular vesicles (EVs) with diameters from 30 to 150 nm.18 They were thought to be a cellular waste product that only discharged intracellular and membrane components from the cell when first detected during sheep reticulocyte development into mature erythrocytes.19,20 However, it is now considered an emerging intercellular communication vehicle, and their various cargoes (proteins, lipids, lncRNA, miRNA, and mRNA) play a crucial role in the pathophysiological mechanisms,21−23 treatment,24−26 and even diagnosis27,28 of diseases. Since the discovery of miRNAs more than 2 decades ago, researchers have developed novel perspectives of diseases, making miRNAs promising therapeutic targets.29 Multiple types of pulmonary cells cooperate in controlling lung inflammation during ALI.30 Exosomes transmit a diversity of specific miRNAs across cells stably and perform various pathogenic regulatory functions in this process.
Therefore, this paper summarizes the current research on the pathogenesis and treatment of ALI based on the participation of exosomal miRNAs in cellular interactions and describes its potential applications in diagnosis.
2. Exosomes
EVs are important mediators of intercellular communication and play important roles in physiological and pathological processes. They can be divided into exosomes, microvesicles, and apoptotic bodies. The exosome is a type of vesicle secreted by various cells and has a bilayer membrane structure.18,31,32 The International Society for EV (ISEV) has published the latest definition of different subtypes of EVs in “Minimum Information for Studies of Extracellular Vesicles 2018” (MISEV 2018).33 This section discusses the formation, morphology, sources, molecular composition, and functions of exosomes.
2.1. Formation
In 1983, exosomes were first discovered by Pan and Johnstone during the maturation of sheep reticulocytes and were associated with the release of transferrin receptors from sheep reticulocytes into extracellular space.19,34 In 1989, these functional EVs were formally defined by Johnstone as exosomes.35 The biosynthetic pathways of exosomes include the endosomal and plasma membrane pathways, among which the endosomal pathway is widely recognized.36 In the endosomal pathway, they are initially produced as early endosomes produced by the inward budding of cell membranes. Subsequently, intracellular bioactive substances accumulate in the early endosome, forming the late endosome. The late endosome membrane buds inward to generate many small vesicles within the cell, eventually coalescing into multivesicular bodies (MVBs) via the Golgi apparatus. Lysosomes within the cell degrade some MVBs, and the others are fused with the cell membrane and release small vesicles to the extracellular space through exocytosis. These are called exosomes (Figure 1).37 MVB synthesis is the central process of exosome biogenesis, mainly through endosomal sorting complexes required for a transport (ESCRT)-dependent pathway.38 However, several laboratories have found that exosome biogenesis is not substantially reduced after the ESCRT pathway is inhibited.39 Other generation pathways reported so far have been classified as ESCRT-independent mechanisms. Studies have shown that the Rab protein could regulate the occurrence of exosomes through endosomes and plasma membranes, among which Rab27a and Rab27b could participate in the localization of vesicles. Rab27a could dynamically regulate plasma membrane phosphatidylinositol 4,5-bisphosphate (PIP2) to assemble plasma membrane microdomains and participate in membrane germination, while Rab35 could regulate PIP2 levels in cell membranes. Rab11 was involved in exosome formation through calcium-induced MVB fusion.40 In addition, cortactin, Rab27a, and coronin 1b collaboratively modulate cutaneous actin to promote exosome secretion.41
Figure 1.
Biogenesis and molecular composition of exosomes. (Left) Schematic representation of the exosome release into the extracellular space. The generation of exosomes begins with endocytosis of the cell membrane and undergoes several steps within the cell before finally fusing with the cell membrane to be transported to the extracellular space by exocytosis. (Right) Schematic diagram of the major components of exosomes. All exosomes have a typical structure similar to that of a cell, including proteins (tetraspanins, annexin, heat shock proteins, etc.), lipids (ceramide, cholesterol, phosphatidylserine, sphingolipid, etc.), and genetic material (DNA, mRNA, miRNA, circRNA, lncRNA, etc.).
2.2. Morphology
With the application of a scanning electron microscope in transmission mode, transmission electron microscopy (TEM) is widely used for the morphological characterization of exosomes with an imaging resolution of approximately 1 nm. Exosomes are negatively stained and cup-shaped under TEM.42 However, TEM must be operated under vacuum conditions, and the exosome samples must be dyed, fixed, and dehydrated. These procedures impact the actual morphology and size of exosomes. Recently, it has been reported that the morphological characterization of exosomes using cryo-electron microscopy is more representative of the natural morphology of exosomes because there is no need to perform the above procedures on the sample. Under cryo-electron microscopy, exosomes are mostly spherical.43 This provides fresh perspectives on the morphological characteristics of exosomes. Due to the heterogeneity of exosomes, the morphology of exosomes may also be diverse. Hoog et al. found that the morphology of EVs was varied by cryo-electron microscopy, which may be used to distinguish different exosome subgroups.44
2.3. Sources
Almost all living cells can secrete exosomes, especially dendritic cells (DCs), epithelial cells, endothelial cells, and lymphocytes.45 Therefore, exosomes can be extracted from a wide range of biofluids (serum, plasma, alveolar lavage, saliva, urine, peritoneal lavage, and breast milk)46−51 and cell supernatants (stem, immune, and tumor cells).52−55 Recent reports have shown that various edible plants can also produce exosomes, such as grapes,56 apples,57 ginger,58 citrus lemon,59 and broccoli.59 These findings have certainly enriched our understanding of the origin of exosomes.
2.4. Molecular Composition
The molecular composition of exosomes mainly consists of proteins, nucleic acids, lipids, and other immunomodulatory factors. The proteins can be divided into two major groups: the membrane and intramembrane. The membrane proteins mainly include tetraspanins (CD9, CD63, CD81, and CD82), flotillin, annexin, antigen-presenting molecules (major histocompatibility complex I [MHC I] and major histocompatibility complex II [MHC II]), and adhesion molecules. Intramembrane proteins mainly contain heat shock protein (Hsp) family proteins (Hsp27, Hsp60, Hsp90, and Hsp70), ESCRT proteins (ALG-2-interacting protein X [Alxi] and tumor susceptibility gene 101 [TSG101]), cytoskeletal proteins (actin, tubulin, and cofilin), growth factors and cytokines (tumor growth factor-beta [TGF-β], tumor necrosis factor-alpha [TNF-α], and TNF-related apoptosis-inducing ligand [TRAIL]), metabolic enzymes (glyceraldehyde 3-phosphate dehydrogenase [GAPDH], enolase-1, and pyruvate kinase M2 [PKM2]), signal transduction factors (melanoma-associated molecules, ADP ribosylation factor 6 [ARF6], and cell division cycle 42), and adhesion molecules (milk fat globule-EGF factor 8, integrins, and P-selectin). Lipids mainly contain ceramide, cholesterol, sphingolipid, and phosphatidylserine. Nucleic acids mainly include DNA and RNA. RNA has mRNA and noncoding RNA, of which noncoding RNA is dominated by miRNA. Among various exosome molecular components, CD9, CD81, TSG101, CD63, and flotillin are currently considered biomarkers of exosomes because of their stable expression within exosomes.60,61
2.5. Functions
Exosomes are highly stable and immunogenic with low toxicity, which has sparked great interest in them as intercellular communicators. There are three modes of exosome communication action: internalization by the recipient cell, delivering cargo such as proteins, and nucleic acids carried internally to the recipient cell to participate in intracellular signaling. For example, exosomal miR-21 of renal tubular epithelial cell origin activates renal fibroblasts and promotes renal fibrosis by inhibiting the phosphatase and tensin homologue (PTEN)/protein kinase B (AKT) signaling pathway.62 The delivery of syndecan-1 attenuates ALI via the FAK/p190RhoGAP/RhoA/ROCK/nuclear factor kappa B (NF-κB) signaling pathway.63 Second, it binds to the receptor cells through membrane surface proteins and mediates the intracellular signaling cascade response in the receptor cells. For example, DC-derived exosomes can combine with bacterial toll-like receptor ligands to indirectly induce innate immune responses by enhancing the stimulation of bystander DCs.64,65 Third, when exosomes are internalized by recipient cells, they promote the production of new exosome populations by recipient cells.66 In addition to their primary role as carriers mediating biological effects within the target cells, exosomes play an indispensable role in liquid biopsies,27 cell-free vaccine development,67 drug delivery,68 and even regenerative medicine.69
3. Exosomal miRNAs
MicroRNAs are a class of noncoding single-stranded RNA molecules of approximately 22 nucleotides in length encoded by endogenous genes and are highly conserved in plants and animals. They can bind to the 3′ untranslated region or open reading frame of downstream target genes to modulate their expression at the posttranscriptional level and usually act as an inhibitor.70 The biogenesis of miRNAs is well understood by researchers.71,72 In animals, miRNA synthesis requires RNA polymerase II and two types of RNase III proteins (Drosha and Dicer). First, the gene carrying miRNA information is transcribed into pri-miRNA by RNA polymerase II in the nucleus. Second, pri-miRNA is cleaved in the nucleus by the Drosha enzyme into precursor miRNA (pre-miRNA) with an approximately 70 nucleotide length. Then, the pre-miRNA is transferred from the nucleus to the cytoplasm with the help of the exportin-5. In the cytoplasm, the pre-miRNA is shed from the exportin-5 and cleaved by the Dicer enzyme into a mature double-stranded miRNA (mature miRNA) of approximately 20 nucleotides in length.73,74 The mature miRNA eventually forms the RNA-induced silencing complex (RISC) with the Argonaute protein.72 The RISC is a ribonucleoprotein complex that guides miRNA to the target mRNAs to achieve gene silencing.75 In this process, the seven miRNA 5′-end nucleotides are the key to mRNA recognition.76 The miRNA-mediated gene-silencing modalities are mRNA degradation and mRNA translation inhibition. When the miRNA is precisely paired with the target mRNA base, the target mRNA is degraded. However, translational repression will occur when miRNAs are imperfectly paired with target miRNAs. miRNA degradation is irreversible, while mRNA translational repression is reversible because stable mRNAs can be translated again after eliminating translational repression.77 Exosomal miRNAs were found in human serum.78 Based on the peculiarity of miRNAs and the high abundance of miRNAs in exosomes, exosomal miRNAs have been recently shown to regulate various signaling pathways and phenotypes, serving as key regulators in many cancer, inflammatory, and metabolic diseases.79−81 In addition, exosomes can be stably preserved under different conditions, and miRNAs can be stably expressed in exosomes.82−84 Therefore, exosomal miRNAs still have some potential value in disease diagnosis.
4. Role of Exosomal miRNAs in the Pathogenesis of ALI
Lung homeostasis is the cornerstone of lung health and depends substantially on the pulmonary microenvironment. Communication among the pulmonary epithelium, endotheliocytes, and immune cells dominantly contributes to maintaining the balance of the pulmonary microenvironment.85,86 The dysfunctions of these cells can alter the pulmonary microenvironment, leading to lung inflammation and cancer development.87−90 Various resident cells from the lung can achieve intercellular communication by secreting exosomes carrying a specific high or low expression of miRNA, providing a novel theoretical foundation for the pathogenesis of ALI (Figure 2).
Figure 2.
Pathogenesis of ALI regulated by exosomal miRNAs. Examples of exosomal miRNAs that play a role in ALI. Exosomes from different sources are involved in ALI by transferring specific miRNAs to regulate relevant genes and pathways, causing changes in target cell phenotype and function.
4.1. Exosomal miRNAs and Macrophages
In the human body, macrophages are a type of immune cell that prevents pathogen invasion and preserves physiological homeostasis. According to the different microenvironments, macrophages can be distinguished into M1 and M2 polarization states. M1-type macrophages can promote inflammatory responses by producing interleukin (IL)-6, TNF-α, IL-12, and other proinflammatory mediators. In contrast, M2-type macrophages have the capacity for anti-inflammatory responses and tissue repair. They produce IL-10, TGF-β, and other anti-inflammatory mediators to inhibit inflammatory responses and accelerate wound healing and revascularization.91 During ALI, exosomes from multiple cellular origins can deliver miRNAs to lung macrophages, causing macrophage activation and generation of inflammatory mediators to hasten lung inflammation. Studies have shown that TNF-α stimulation of neutrophils generates exosomes with a high miR-30d-5p expression that can reach the lungs of mice and induce NF-κB activation by targeting lung macrophage suppressors of cytokine signaling (SOCS) 1/sirtuin 1 (SIRT1), thereby resulting in M1 macrophage polarization and pyroptosis.53 In the lipopolysaccharide (LPS)-induced rat sepsis model, exosomes isolated from bronchoalveolar lavage fluid (BALF) highly expressed miR-92a-3p, which activates NF-κB by targeting the PTEN/AKT signaling pathways in alveolar macrophages, increasing inflammation and alveolar permeability in the rat lung.92 miR-155 is a common regulator of inflammation.93 In a different study of sepsis-induced ALI, elevated serum exosomal miR-155 caused macrophage proliferation and release of proinflammatory mediators by targeting lung macrophage SH2-containing inositol 5′-phosphatase 1 (SHIP1)/SOCS1, respectively. Inhibiting miR-155 may counteract the proinflammatory effects of macrophages.21 However, it has also been reported that miR-155 can alleviate the disease by reducing the formation of neutrophil extracellular traps (NETs) in the lungs of mice with abdominal sepsis through upregulation of peptidyl arginine deiminase 4 (PAD4) and promotion of histone 3 citrullination.94 In addition to increased levels of proinflammatory miRNAs in exosomes, anti-inflammatory miRNA levels are concurrently declining. Zhang et al.95 proved that exosomes isolated from BALF in mice with pneumonia were macrophage-derived. Moreover, the decrease of anti-inflammatory miR-223/142 in exosomes promoted the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome activation in macrophages, which subsequently caused the delivery of proinflammatory mediators (IL-1β and IL-18) to induce ALI. Moreover, a recent study showed that mitochondrial autophagy induced miR-138–5p promoter demethylation and inhibited NLRP3 inflammatory vesicle activation and macrophage pyroptosis, thereby attenuating septic lung injury.96
4.2. Exosomal miRNAs and Epithelial Cells
The alveolar epithelium is an important defense mechanism of the lung against external invasion. It generously covers the alveolar surface and functions as a lung protector, assisting in preserving the structural integrity of the lung during ALI.97 Two types of alveolar epithelial cells (AECs) are resident in the alveolar epithelium, namely, alveolar epithelial type I cells (ACE I) and alveolar epithelial type II cells (ACE II). ACE I, which covers approximately 95%–98% of the alveolar surface area, is majorly involved in the air–blood exchange between the alveoli and the blood. In contrast, ACE II, which occupies approximately 2%–5% of the surface area, can secrete surface-active substances to maintain alveolar surface tension.98,99 Exosomal miRNAs have been implicated in numerous studies as mediating the involvement of AECs in the emergence of ALI. SOCS6 acts as a member of the negative feedback regulation, reducing cytokine signaling by inhibiting multiple activated cytokines and tyrosine kinase receptors.100 Ma et al. cocultured serum exosomes from patients with septic lung injury with BEAS-2B cells and observed that exosomal miR-1298-5p could activate the downstream signal transducer and activator of the transcription 3 (STAT3) signaling pathway by suppressing SOCS6 expression in cells, resulting in the suppression of cell proliferation and an increase of cell permeability. Moreover, overexpression of SOCS6 could alleviate cell damage.101 miR-145 is thought to be a tumor suppressor.102 A study has revealed that miR-145 was significantly downregulated in blood exosomes from septic patients with lung injury and in LPS-treated BEAS-2B cells. Additionally, there was a positive correlation between the degree of miR-145 reduction and the condition of lung injury. Mechanistic studies indicate that miR-145 is significantly reduced in BEAS-2B cells by targeting TGF-β receptors 2, elevating the downstream Smad3, and promoting the inflammatory cytokines’ IL-2 and TNF-α deliverance to cause lung injury.103
4.3. Exosomal miRNAs and Endothelial Cells
Vascular endothelial cells participate in vascular tension formation, local blood flow regulation, immune response, and angiogenesis.104 Vascular endothelial damage due to disruption of the vascular endothelial barrier is a pathological characteristic of ALI.105 miR-1–3p expression drastically increased in the plasma exosomes of rats with septic-associated lung injury and LPS-stimulated human umbilical vein endothelial cells (HUVECs), which promoted apoptosis and cytoskeleton contraction and increased monolayer endothelial cell permeability by inhibiting stress-associated endoplasmic reticulum protein 1 (SERP1) expression in HUVECs.106 In 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) broke out, with a significant negative impact on the economy and global public health. Hypoxic respiratory failure caused by ALI is the leading cause of mortality in patients infected with SARS-CoV-2.107 Increased plasma abundance of exosome-associated neutrophil elastase was related to endothelial cell injury in patients with SARS-CoV-2 and ARDS.108 In addition, the elevation of serum exosomal miR-126 during severe community-acquired pneumonia (SCAP) may be associated with inhibiting pulmonary vascular endothelial cell proliferation and promoting apoptosis by targeting the L-type amino acid transporter 1 (LAT1)/mammalian target of rapamycin (mTOR) signaling axis.109,110
4.4. Exosomal miRNAs and Neutrophils
Neutrophils are first-line defense cells that eliminate pathogenic microorganisms through a nonspecific mechanism. The release of NETs, a unique mechanism of the natural immune response, is an important mode of action to locate and kill pathogens.111 However, NETs play a dual role. In the early stages of sepsis, NETs appear to have a protective function, but as the disease progresses, an enhanced NET release may contribute to thrombosis and multiorgan dysfunction.112 During infectious shock, miR-15b-5p and miR-378a-3p can advance the formation of NETs and aggravate lung injury by targeting neutrophil polycystin 1 (PKD1) and activating the AKT/mTOR autophagy pathway. In addition, the research team revealed for the first time that IκB kinase inhibitors can ameliorate the severity of lung injury during infectious shock by controlling the secretion of platelet-derived exosomes and inhibiting the formation of NETs.113
5. Role of Exosomal miRNAs in ALI Treatment
Despite the multiple targets for ALI/ARDS that have been researched, efficient clinical treatment for ALI is still absent. The main treatment for ALI/ARDS remains symptomatic, including mechanical ventilation, fluid management, corticosteroid supplementation, and inhaled pulmonary vasodilators.14 Although these treatments can relieve the symptoms of patients, they do not solve the underlying problem. Recently, RNA-mediated gene-silencing therapy has opened up a new path for treating ALI/ARDS.114 However, because of the characteristics of naked siRNA, the low efficiency of siRNA delivery to target cells limits the therapeutic performance of siRNA. Therefore, a carrier with good biocompatibility and transport capabilities is urgently needed.115 Exosomes offer excellent potential as carriers for gene therapy and drug delivery because of their unique physicochemical characteristics (small size, high penetration, high deliverability, and minimal immunogenicity).116 For example, exosome-loaded adriamycin exhibits a faster cellular uptake and more severe toxic side effects than free adriamycin and liposome-encapsulated adriamycin.117 Although miRNAs are relatively poor matches to target mRNAs compared to siRNAs, they also have gene-silencing effects.75 Evidence suggests that mesenchymal stem cell (MSC)-derived exosomal miRNAs can form intercellular interactions with resident lung cells and have potential therapeutic value for ALI by targeting different genes and pathways (Table 1).
Table 1. Pathogenesis of Exosomal miRNAs in the Ttreatment of ALI.
Source | miRNA | Model construction | Effector cells | Mechanism | Function | Reference |
---|---|---|---|---|---|---|
BMSCs | miR-199a-3p | LPS | Primary mice ATIIC | Increasing of α/γ-ENaC | Inhibited apoptosis, increased cell viability, and restored pulmonary edema | (120) |
BMSCs | miR-30b-3p | LPS | MLE-12 | Targeting SAA3 | Inhibited apoptosis and promoted cell proliferation | (121) |
BMSCs | miR-132-3p | LPS | MLE-12 | Targeting PTEN/PI3K/AKT signaling pathway | Promoted cell proliferation and inhibited apoptosis | (122) |
BMSCs | miR-182-5p, miR-23-3p | LPS | MLE-12 | Targeting Ikbkb/Usp50 and inhibiting NF-κB/Hedgehog signaling pathway | Reduced EMT generation | (124) |
BMSCs | miR-150 | LPS | HUVEC | Targeting MAPK signaling pathway | Inhibited apoptosis | (125) |
BMSCs | miR-384-5p | LPS | NR8383 | Targeting Beclin-1 | Inhibited apoptosis and cellular autophagic stress | (126) |
BMSCs | miR-127-5p | LPS | 293T | Targeting CD64 | Reduced NET formation | (127) |
BMSCs | miR-181-5p | LPS | MDM | Targeting PTEN/pSTAT1/SOCS1 signaling pathway | Promoted macrophage reprogramming | (129) |
BMSCs | miR-425 | High oxygen | RL6-6TN | Targeting PTEN/PI3K/AKT signaling pathway | Increased cell viability and inhibited apoptosis | (130) |
BMSCs | miR-21-5p | I/R | Primary lung endothelial cells | Targeting PTEN/PDCD4 | Inhibited apoptosis | (131) |
BMSCs | miR-202-5p | I/R | MLE-12 | Targeting CMPK2 | Inhibited pyroptosis | (132) |
UC-MSCs | miR-22-3p | LPS | NR8383 | Targeting FZD6 to inhibit NF-κB activation | Inhibited inflammation | (134) |
UC-MSCs | miR-377-3p | LPS | HPAEpiC | Targeting RPTOR | Promoted autophagy | (135) |
UC-MSCs | miR-199a-5p | SM | BEAS-2B | Targeting CAV1/NRF2 signaling pathway | Inhibited oxidative stress | (136) |
UC-MSCs | miR-451 | Burns | – | Targeting TLR4 to inhibit NF-κB | Inhibited inflammation | (137) |
UC-MSCs | miR-451 | Burns | NR8383 | Targeting MIF to activate the PI3K/AKT signaling pathway | Promoted macrophage transformation from M1 to M2 | (138) |
UC-MSCs | miR-146-5p | Pristane | Primary lung macrophages | Inhibiting NOTCH | Promoted macrophage transform from M1 to M2 | (139) |
EPCs | miR-126 | CLP | HUVEC | Targeting SPRED1 to activate the RAF/ERK signaling pathway | Increased endothelial cell permeability, promoted endothelial cell proliferation, migration, and revascularization | (141) |
EPCs | miR-126-3p, miR-126-5p | LPS | SAEC | Targeting PIK3R2 to inhibit HMGB1/VEGF | Maintenance of alveolar epithelial barrier integrity | (142) |
EPCs | miR-382-3p | CLP | – | Targeting BTRC/IκBα/NF-κB | Mediated tissue repair and T-cell immune activity | (143) |
ADSCs | miR-125b-5p | LPS | PMVEC | Targeting Keap1/Nrf2/GPX4 signaling pathway | Inhibited ferroptosis | (146) |
ADSCs | miR-126 | Histone | HUVEC | Targeting PI3K/AKT signaling pathway | Inhibited apoptosis | (150) |
BALF | miR-223-3p | LPS | NR8383 | Targeting STK39 | Increased cell viability, activate autophagy, reduced apoptosis, and inflammation | (151) |
BEAS-2B | miR-103a-3p | LPS | BEAS-2B | Targeting TBL1XR1 to inhibit NF-κB | Inhibited inflammation | (152) |
RLE-6TN | miR-146a | LPS | NR8383 | Targeting TLR4 to inhibit NF-κB | Inhibited inflammation | (153) |
bEnd.3 | miR-125-5p | LPS | – | Targeting TOP2A to elevate VEGF | Inhibited apoptosis | (154) |
A549 | miR-371b-5p | Bleomycin | ATIIC | Targeting PTEN to promote AKT/GSK3β/FOXO phosphorylation | Promoted cell-specific proliferation | (155) |
5.1. BMSC-Derived Exosomal miRNAs and ALI
Bone marrow mesenchymal stem cells (BMSCs) are a type of adult stem cell in the bone marrow other than hematopoietic stem cells. They are used to treat ALI because of their immunomodulatory and regenerative properties, which can lessen the generation of proinflammatory cytokines and promote tissue repair.118 BMSC-derived exosomes (BMSCs-Exo) may contribute to their capacity to cure ALI.119 BMSCs-Exo were reported to reduce LPS-induced ALI by promoting the viability of mouse type II AECs (ATIIC) while inhibiting their apoptosis. Elevated miR-199a-3p in exosomes causes an increase in the α/γ-epithelial sodium channel protein, which helps to restore pulmonary edema.120 By coculturing BMSCs-Exo with the mouse lung epithelial cell line (MLE-12), elevated miR-30b-3p/miR-132-3p in exosomes could target and inhibit the serum amyloid A isoform 3 (SAA3)/TNF receptor associated factor 6 (TRAF6) expression in MLE-12 cells, thereby inhibiting apoptosis and promoting cell proliferation to improve LPS-induced ALI.121,122 Epithelial–mesenchymal transition (EMT) is the conversion of epithelial cells to mesenchymal cells, closely related to the occurrence and progression of idiopathic fibrosis.123 Xiao et al.124 have shown that BMSCs-Exo could inhibit the expression of nuclear factor kappa B kinase subunit beta (IKBKB) and ubiquitin-specific peptidase 50 (Usp50) in MLE-12 cells by delivering miR-182-5p and miR-23-3p, respectively. As binding of IKBKB to Usp50 can cause I kappa B kinase beta (Ikkβ) ubiquitination, inhibition of IKBKB and Usp50 caused a reduction in Ikkβ ubiquitination, which in turn blocked NF-κB and hedgehog signaling pathway activation, reversing EMT progression. In the LPS-induced pulmonary microvascular endothelial cell model, the BMSC-derived exosomal miR-150 can degrade the apoptosis of pulmonary microvascular endothelial cells by inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway activation to maintain the structural integrity of alveoli.125 Furthermore, BMSC-derived exosomal miR-384-5p can reduce macrophage apoptosis and autophagy stress by targeting Beclin-1 of alveolar macrophages to improve survival in ALI rats.126 In animal model studies, BMSC-derived exosomal miR-127-5p inhibited the formation of NETs in sepsis-associated ALI by targeting CD64.127 Macrophage reprogramming has a protective effect on inflammation.128 It was reported that miR-181a-5p in BMSC-derived EVs downregulated PTEN, subsequently producing pSTAT1 and SOCS1. Activating this signaling axis promoted macrophage reprogramming, reduced secretion of TNF-α and IL-8, and inhibited the inflammatory response in ARDS.129
In addition to LPS-induced ALI, BMSCs-Exo has been studied in hyperoxia-induced ALI. High expression of miR-425 in MSCs-Exo can target and inhibit the expression of PTEN in rat alveolar epithelial type II cell lines. PTEN is an antioncogene, and when it is inhibited, the downstream phosphoinositide 3-kinase (PI3K)/AKT inflammatory signaling pathway is activated, resulting in increased cell activity and decreased apoptosis, thus alleviating cell damage.130 Lung ischemia/reperfusion (I/R) is also a cause of ALI. Ji et al. found that BMSCs-Exo had protective effects against oxidative stress-induced ALI in mice by constructing a mouse I/R model in vivo and a hypoxia/reoxygenation model in vitro, which was attributed to the inhibition of endogenous and exogenous apoptosis by exosomal miR-21-5p by inhibiting the PTEN and programmed cell death 4 (PDCD4) in primary mouse pulmonary endothelial cells.131 In another study of a mouse model of I/R, miR-202-5p from BMSCs-Exo could inhibit pyroptosis in lung epithelial cells by targeting cytidine/uridine monophosphate kinase 2 (CMPK2).132
5.2. Umbilical-Cord-Blood MSC-Derived Exosomal miRNAs and ALI
Umbilical-cord-blood MSCs (UCB-MSCs) are typical adult stem cells. They are considered the optimum selection for stem cell therapy because of their noninvasive collection, low immunogenicity, easy in vitro expansion, and ethical compliance compared with other stem cells.133 Numerous studies have pointed to the ability of UCB-MSC-derived exosomes (UCBMSCs-Exo) to mitigate the progression of ALI. Zheng et al.134 found in the LPS-induced macrophage model that the elevated miR-22-3p in UCBMSCs-Exo could target and inhibit frizzled class receptor 6 (FZD6), reduce the cellular inflammatory response and oxidative stress, promote cell proliferation, and inhibit cell apoptosis. In addition, animal experiments have shown that exosomal miR-22-3p can reduce lung inflammation by inhibiting NF-κB activation. Autophagy plays an essential role in tissue repair. Lung inflammation and oxidative stress can be significantly inhibited in pulmonary diseases by activating autophagy. Wei et al.135 found that miR-377-3p in UCBMSCs-Exo could target and inhibit the regulatory-associated protein of mTOR complex 1 (PRTOR) in human AECs and promote cellular autophagy to protect against LPS-induced ALI. Moreover, miR-199a-5p from UCBMSCs-Exo was shown to be a key molecule to alleviate sulfur mustard (SM)-related oxidative stress by regulating the caveolin 1 (CAV1)/nuclear factor erythroid-2–related factor 2 (NRF2) signaling pathway.136 Burns are also a cause of ALI, and burn-induced ALI requires the involvement of functional toll-like receptor 4 (TLR4).8 It was discovered that elevated miR-451 in UCBMSCs-Exo could target and inhibit TLR4 in mouse lung tissue, thereby blocking NF-κB activation and reducing proinflammatory cytokine generation to alleviate burn-induced ALI.137 However, the specific mechanism of action of miR-451 still needs further clarification in in vitro experiments. In another study of burn-induced ALI, the mechanism of the effect of miR-451 in HUC-MSCs-Exo was confirmed at the cellular level, which promoted the transformation of macrophages from M1 to M2 by targeting the macrophage migration inhibitory factor (MIF)/PI3K/AKT signaling pathway.138 Relevant reports have found that UCBMSCs-Exo also has a therapeutic effect on systemic lupus erythematosus (SLE)-associated diffuse alveolar hemorrhage in mice. The exosomes reduced the level of NOTCH expression in diffuse alveolar hemorrhage (DAH) mice via lung tissue via miR-146a-5p, thus reducing lung tissue bleeding and inflammatory cell infiltration. This may be due to miR-146a-5p facilitating the transformation of alveolar macrophages from M1 to M2.139
5.3. Endothelial-Progenitor-Cell-Derived Exosomal miRNAs and ALI
Endothelial progenitor cells (EPCs) are a special type of stem cells and vascular endothelial precursor cells, with a role in normal endothelial function and repair after vascular injury.140 In the ALI, EPCs can migrate to the site of the lesion and improve lung inflammation by participating in vascular endothelial remodeling. Many studies have revealed that miRNAs from EPC-derived exosomes (EPCs-Exo) influence this process. miR-126 is an important regulator of angiogenic signaling that maintains the integrity of vascular endothelial cells and the vascular detail. Xu et al.141 found that miR-126 in EPCs-Exo was transferred to HUVECs and activated the RAF/extracellular regulated protein kinase (ERK) signaling pathway by targeting the sprouty-related EVH1 domain containing 1 (SPRED1) in cells, increasing endothelial cell permeability while promoting endothelial cell proliferation, migration, and angiogenesis to alleviate ALI. Another study found that intratracheal dripping of EPC-Exo with high expression of miR-126-3p and miR-126-5p reduced lung edema and inflammatory cell infiltration and restored alveolar epithelial barrier integrity in mice after 24 and 48 h of LPS induction. This may be connected to suppressing the ALI-related target genes phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2) and high mobility group box 1 (HMGB1)/vascular endothelial growth factor (VEGF) by miR-126-3p/miR-126-5p.142 In addition, miR-382-3p in EPCs-Exo can also target the regulation of a beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC) and an IκBα/NF-κB axis to restore the number of lymphocytes and maintain the balance between Th1 and Th2 cells to alleviate ALI in mice with cecal ligation and puncture (CLP)-induced sepsis.143
5.4. Adipose-Stem-Cell-Derived Exosomal miRNAs and ALI
Adipose stem cells (ADSCs) are mesenchymal ASCs with a multifunctional differentiation potential isolated from adipose tissue. It is of great significance in treating diseases because of its ability to repair tissue cells, resist aging, and improve the subhealth state of the body.144 For instance, human ADSC-derived exosomes (ADSCs-Exo) could inhibit oxidative stress in pulmonary vascular endothelial cells and reduce cell monolayer permeability to alleviate CLP-induced ALI in mice.145 Moreover, miR-125b-5p in the ADSCs-Exo could alleviate ferroptosis of pulmonary vascular endothelial cells (PMVECs) in sepsis-associated ALI by regulating the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor-erythroid 2-related factor 2 (Nrf2)/glutathione peroxidase 4 (GPX4) signaling axis, thereby reducing lung inflammation.146 There is growing evidence that extracellularly released histones are becoming damage-associated molecular patterns, which are thought to contribute to the development of lung injury.126,147−149 Mituzn et al.150 showed that miR-126 in ADSCs-Exo inhibited PI3K/AKT activation in endothelial cells and attenuated histidine-induced ALI.
5.5. Others
In addition to stem cell therapy, the communication of exosomal miRNAs produced by lung resident cells is also a potential therapeutic modality for ALI. It has been suggested that AEC-derived exosomes may also alleviate ALI by acting on macrophages. Nan et al.151 found that BALF-Exo from LPS-induced ALI mice could wrap miR-223-3p to reach alveolar macrophages and negatively regulate serine/threonine kinase 39 (STK39) in the cells. This boosted cell survival, activated autophagy, and reduced apoptosis and inflammation, alleviating ALI. Li et al.152 found that miR-103a-3p was lowly expressed in exosomes generated from serum samples of pediatric patients with pneumonia and the LPS-induced human lung epithelial cell line (BEAS-2B). miR-103a-3p could target transducin of (beta)-like 1 X-linked receptor 1 (TBL1XR1) in BEAS-2B to inhibit NF-κB activation, releasing proinflammatory mediators IL-6 and TNF-α. Traditional Chinese medicine (TCM) is a national treasure of China, and salidroside is one of the effective monomeric components of the TCM Rhodiola rosea L. Zheng et al.153 have shown that salidroside treatment elevated miR-146a in alveolar epithelial-derived exosomes, which targeted and inhibited TLR4 in alveolar macrophages causing NF-κB activation to improve LPS-induced ALI in rats, providing a new theory for the treatment of ALI with TCM. However, exosomal miRNAs produced by cells are not limited to crosstalk with other cells to exert their functions but could also act on themselves. In a mouse model of sepsis constructed by CLP, miR-125b-5p was highly expressed by endothelial cell-derived exosomes, which could inhibit apoptotic injury by targeting and inhibiting their own DNA topoisomerase II alpha (TOP2A) and elevating the vascular endothelial growth factor (VEGF).154 In addition, the ATIIC-derived exosomal miR-371b-5p could promote AKT and its downstream glycogen synthase kinase-3 beta (GSK3β) and forkhead box O (FOXO) phosphorylation by targeting and inhibiting its own PTEN, thus causing ATIIC-specific proliferation and promoting damaged alveolar re-epithelialization to alleviate bleomycin-induced ALI.155
6. Role of Exosomal MiRNAs in the Diagnosis of ALI
Biomarkers are biochemical indicators that can objectively detect and evaluate changes in body structure and are used for disease screening, prediction, and diagnosis.156 However, there is no single biomarker with sufficient sensitivity and specificity for clinical diagnosis of ALI. Therefore, new biomarkers for the early detection of ALI are urgently required. In the past decade, miRNAs have been discovered to engage in pathophysiological processes, including lung injury and repair. Consequently, scientists have been actively investigating their potential for use as biomarkers.157,158 miRNAs have many advantages as biomarkers. First, the internal cargo of exosomes is finely regulated by parental cells in physiological and pathological states. This can reflect changes in parental cells to a certain extent.159 Therefore, the internal cargo of exosomes is likely to be a variable feature that can be captured to predict the functional state of the parent cells. Second, exosomes are relatively stable based on their characteristics, and exosome miRNAs are more resistant to degradation after cryopreservation than cellular miRNAs.160 Most importantly, exosomes are present in almost all biofluids, providing a good biological basis for exosomes as a disease diagnostic tool, especially for noninvasive diagnostics.161 In summary, exosomal miRNAs are superior to circulating or cellular miRNAs alone as diagnostic tools.
With the continuous advancement and application of various bioassay technologies, Sandfeld-Paulsen et al.162 identified various highly expressed exosomal proteins using mass spectrometry analysis of plasma exosome proteomics in patients with lung cancer. These proteins could be used as a diagnostic tool for lung cancer independently of pathological staging and histological subtypes. In addition, the authors also demonstrated that various exosomal miRNAs of humoral origin can be used as early diagnostic markers for ALI to some extent. Parzibut et al.163 compared the expression of plasma exosomal miRNA in 8 patients with ARDS and 10 healthy subjects using small RNA sequencing analysis and identified 12 differentially expressed miRNAs. Among these differentially expressed miRNAs, seven miRNAs (miR-221-3p, miR-24-3p, miR-130a-3p, Let-7d-3p, miR-1273a, miR-98-3p, and miR-193a-5p) were proved to distinguish ARDS and hemorrhagic shock well using receiver operating characteristic curve analysis (area under the curve >0.8). Recent studies have found that EVs with CD14+ in BALF can also serve as a new biomarker for ARDS. Elevated counts of EVs with CD14+/CD81+ in BALF of patients with sepsis-associated ARDS are associated with the increased mortality of patients with ARDS.164 SCAP usually leads to high mortality in ARDS. Higher levels of miR-146a, miR-126, miR-27a, and miR-155 were found in serum exosomes of patients with SCAP than in the non-ARDS group, and the combination of the four was predictive of ARDS. In addition, the authors also found that miR-126 could predict 28-day mortality in patients with SCAP.109 In a recent asthma study, miR-126 was highly expressed in serum exosomes of patients with allergic asthma and lung tissues of asthmatic mice, which has some reference value for diagnosing bronchial asthma.165 Moreover, the amount of miR-122-5p in plasma EVs was proportional to the number of inflammatory cells in the blood of patients with asthma, and it has the potential to distinguish different subtypes of asthma.166 Sputum is also the direct body fluid for respiratory disease detection. miR-142-3p, miR-223-3p, and miR-629-3p were increased in the sputum of patients with severe asthma, and this was associated with increased sputum neutrophils.167 It would be interesting to prove that these miRNAs are enriched in exosomes. Harmful gas inhalation is an etiology of ALI. In ozone-induced ALI mice, differential miRNAs in EVs isolated from BALF increased with increasing ozone concentration compared with controls, among which miR-22-3p is expected to be a marker of ALI. In addition, comprehensive analysis suggests that the occurrence of ALI induced by miR-22-3p could be by targeting macrophages.168 Local vascular inflammation due to intimal tear and pseudotumor formation in acute type A aortic dissection (ATAAD) can progress to systemic inflammatory syndrome, leading to ALI. miR-485 was upregulated, and miR-206 was downregulated in plasma exosomes of patients with ALI compared with those with ATAAD without ALI and is expected to be a marker of ALI in patients with ATAAD.169
7. Opportunities and Challenges for Exosomal miRNA in ALI
In previous studies of ALI, exosomes seem to be a new therapeutic agent for gene and drug delivery. They are also helpful in early diagnosis and improve the prognosis (Table 2). However, there are still many challenges to using exosomes for practical clinical applications: (1) The mechanism of action of exosomal miRNAs is mostly based on animal/cellular models; it needs more validation in clinical samples. (2) Prospective studies of clinical samples are mostly based on small samples and lack validation in large cohort studies. (3) The regulation mechanism of miRNA in ALI is complex and affected by many factors. Further elucidation of the mechanism of action of exosomal miRNAs in disease states is needed. (4) The isolation of exosomes is uneven and relatively costly, which undoubtedly increases the cost of treating patients if applied to clinical treatment. Therefore, to apply exosome therapy to clinical practice, we must continue exploring exosome isolation technology to find an efficient, rapid, low-cost, and standardized extraction method. (5) EVs include several subtypes, and different EVs may have different effects; therefore, there is an urgent need for specific ways to accurately distinguish different subtypes of EVs and ensure the purity of exosomes. (6) Although exosomes as a diagnostic tool in liquid biopsies are easy to obtain and noninvasive, the changes of miRNA in exosomes in diseased states are diverse and even change continuously with the development of ALI. Combining multiple exosomal miRNAs with existing mature diagnostic indicators may help in the early diagnosis of ALI and improve the sensitivity and specificity of ALI diagnosis. (7) Differences in exosomes secreted by different cells under different environments/stimuli and how to prepare stable decoy exosomes for disease treatment must also be addressed.
Table 2. Exosomal miRNAs Associated with ALI Diagnosis.
Diseases | Biofluids | miRNAs | Changes | Reference |
---|---|---|---|---|
ARDS | Plasma | miR-130a-3p, miR-98-3p, miR-221-3p, miR-193a-5p, miR-24-3p, Let-7d-3p, miR-1273a, miR-193a-5p | Upregulated | (163) |
SCAP | Serum | miR-146a, miR-126, miR-27a, miR-155 | Upregulated | (109) |
Serum/lung tissue | miR-126 | Upregulated | (165) | |
ASTHMA | Plasma | miR-122-5p | Upregulated | (166) |
Sputum | miR-142-3p, miR-223-3p, miR-629-3p | Upregulated | (167) | |
ALI | BALF | miR-22-3p | Upregulated | (168) |
ATAAD | Serum | miR-485, miR-206 | Upregulated, Downregulated | (169) |
8. Conclusion
In conclusion, exosomal miRNAs have a multifaceted regulatory role in ALI. As a new class of regulators, miRNAs reach effector cells under exosomal load to mediate intercellular crosstalk. These miRNAs activate the inflammatory signaling axis by regulating downstream target genes, promoting changes in the phenotype and function of target cells to promote disease progression. Moreover, MSCs are a class of pluripotents with self-renewal and multidirectional differentiation capabilities. The exosomes produced by MSCs from different sources achieve anti-inflammatory, antiapoptotic, and antioxidative stress effects through miRNAs, which are expected to be applied in clinical cell-free therapy. In addition, exosome substances are protected from degradation and may be useful in diagnosing ALI by analyzing the composition of miRNAs with specificity in humoral exosomes. Although the research based on miRNA delivery is only the tip of the iceberg, with the continuous progress of technology and the gradual deepening of research, exosomal miRNAs are expected to provide a new strategy for preventing, diagnosing, and treating ALI.
Acknowledgments
The authors thank Dr. Ge Peng and Dr. Yao Jiaqi for their significant contributions to the revision of this paper.
Glossary
Abbreviations Used
- ALI
acute lung injury
- ARDS
acute respiratory distress syndrome
- PaO2
arterial pressure of oxygen
- FiO2
inspiratory fraction of oxygen
- EVs
extracellular vesicles
- ISEV
International Society for Extracellular Vesicles
- MISEV
Minimum Information for Studies of Extracellular Vesicles
- MVBs
multivesicular bodies
- ESCRT
endosomal sorting complexes required for transport
- PIP2
phosphatidylinositol 4,5-bisphosphate
- SEM
scanning electron microscope
- TEM
transmission electron microscopy
- DCs
dendritic cells
- MHC I
major histocompatibility complex I
- MHC II
major histocompatibility complex II
- Hsp
heat shock protein
- Alix
ALG-2-interacting protein X
- TSG101
tumor susceptibility gene 101
- TGF-β
tumor growth factor-beta
- TNF-α
tumor necrosis factor-alpha
- TRAIL
TNF-related apoptosis-inducing ligand
- GAPDH
glyceraldehyde 3-phosphate dehydrogenase
- PKM2
pyruvate kinase M2
- PTEN
phosphatase and tensin homologue
- AKT
protein kinase B
- NF-κB
nuclear factor kappa B
- pre-miRNA
precursor miRNA
- mature miRNA
mature double-stranded miRNA
- RISC
RNA-induced silencing complex
- IL
interleukin
- SOCS1
suppressor of cytokine signaling
- SIRT1
sirtuin 1
- LPS
lipopolysaccharide
- BALF
bronchoalveolar lavage fluid
- SHIP1
SH2-containing inositol 5′-phosphatase 1
- NETs
neutrophil extracellular traps
- PAD4
peptidyl arginine deiminase 4
- NLRP3
NLR family pyrin domain containing 3
- AECs
alveolar epithelial cells
- ACE I
epithelial type I cells
- ACE II
alveolar epithelial type II cells
- STAT3
signal transducer and activator of transcription 3
- HUVECs
human umbilical vein endothelial cells
- SERP1
stress-associated endoplasmic reticulum protein 1
- SARS-CoV-2
severe acute respiratory syndrome coronavirus 2
- SCAP
severe community-acquired pneumonia
- LAT1
L-type amino acid transporter 1
- mTOR
mammalian target of rapamycin
- PKD1
polycystin 1
- MSCs
mesenchymal stem cells
- BMSCs
bone marrow mesenchymal stem cells
- BMSCs-Exo
BMSC-derived exosomes
- ATIIC
type II AECs
- SAA3
serum amyloid A isoform 3
- TRAF6
TNF receptor associated factor 6
- EMT
epithelial–mesenchymal transition
- IKBKB
inhibition of nuclear factor kappa B kinase subunit beta
- Usp50
ubiquitin-specific peptidase 50
- Ikkβ
IkappaB kinase beta
- MAPK
mitogen-activated protein kinase
- PI3K
phosphoinositide 3-kinase
- I/R
ischemia/reperfusion
- PDCD4
programmed cell death 4
- CMPK2
cytidine/uridine monophosphate kinase 2
- UCB-MSCs
umbilical cord blood mesenchymal stem cells
- UCBMSCs-Exo
UCB-MSC-derived exosomes
- FZD6
frizzled class receptor 6
- PRTOR
regulatory-associated protein of mTOR complex 1
- SM
sulfur mustard
- CAV1
caveolin 1
- NRF2
nuclear factor erythroid-2-related factor 2
- TLR4
toll-like receptor 4
- MIF
migration inhibitory factor
- SLE
systemic lupus erythematosus
- DAH
diffuse alveolar hemorrhage
- EPCs
endothelial progenitor cells
- EPCs-Exo
endothelial progenitor cell derived exosomes
- ERK
extracellular regulated protein kinases
- SPRED1
sprouty-related EVH1 domain containing 1
- PIK3R2
phosphoinositide-3-kinase regulatory subunit 2
- HMBG1
high mobility group box 1
- VEGF
vascular endothelial growth factor
- BTRC
beta-transducin repeat containing E3 ubiquitin protein ligase
- CLP
cecal ligation and puncture
- ADSCs
adipose stem cells
- ADSCs-Exo
ADSC-derived exosomes
- PMVECs
pulmonary vascular endothelial cells
- Keap1
Kelch-like ECH-associated protein 1
- Nrf2
nuclear factor–erythroid 2-related factor 2
- GPX4
glutathione peroxidase 4
- STK39
serine/threonine kinase 39
- TBL1XR1
transducin (beta)-like 1 X-linked receptor 1
- TCM
traditional Chinese medicine
- TOP2A
DNA topoisomerase II alpha
- VEGF
vascular endothelial growth factor
- GSK3β
glycogen synthase kinase-3 beta
- FOXO
forkhead box O
- ATAAD
acute type A aortic dissection
Author Contributions
# These authors contributed equally to this work. Bowen Lan and Xuanchi Dong cowrote the original manuscript and drew the graphs. Qi Yang and Yalan Luo reviewed and edited the original manuscript. Zhe Chen and Haiyun Wen checked the references. Hailong Chen provided professional guidance and serious supervision.
This study was supported by the National Natural Science Foundation of China (Nos. 82074158 and 82274311) and the National Key R&D Program of China (No. 2019YFE0119300).
The authors declare no competing financial interest.
References
- Cardinal-Fernández P.; Lorente J. A.; Ballén-Barragán A.; Matute-Bello G. Acute Respiratory Distress Syndrome and Diffuse Alveolar Damage. New Insights on a Complex Relationship. Ann. Am. Thorac Soc. 2017, 14 (6), 844–850. 10.1513/AnnalsATS.201609-728PS. [DOI] [PubMed] [Google Scholar]
- Ashbaugh D. G.; Bigelow D. B.; Petty T. L.; Levine B. E. Acute respiratory distress in adults. Lancet 1967, 2 (7511), 319–323. 10.1016/s0140-6736(67)90168-7. [DOI] [PubMed] [Google Scholar]
- Bernard G. R.; Artigas A.; Brigham K. L.; Carlet J.; Falke K.; Hudson L.; Lamy M.; Legall J. R.; Morris A.; Spragg R. The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am. J. Respir Crit Care Med. 1994, 149, 818–824. 10.1164/ajrccm.149.3.7509706. [DOI] [PubMed] [Google Scholar]
- Ranieri V. M.; Rubenfeld G. D.; Thompson B. T.; Ferguson N. D.; Caldwell E.; Fan E.; Camporota L.; Slutsky A. S. Acute respiratory distress syndrome: the Berlin Definition. Jama 2012, 307 (23), 2526–2533. 10.1001/jama.2012.5669. [DOI] [PubMed] [Google Scholar]
- Rubenfeld G. D.; Caldwell E.; Peabody E.; Weaver J.; Martin D. P.; Neff M.; Stern E. J.; Hudson L. D. Incidence and outcomes of acute lung injury. N Engl J. Med. 2005, 353 (16), 1685–1693. 10.1056/NEJMoa050333. [DOI] [PubMed] [Google Scholar]
- Bellani G.; Laffey J. G.; Pham T.; Fan E.; Brochard L.; Esteban A.; Gattinoni L.; van Haren F.; Larsson A.; McAuley D. F.; 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. 10.1001/jama.2016.0291. [DOI] [PubMed] [Google Scholar]
- Fan E.; Brodie D.; Slutsky A. S. Acute Respiratory Distress Syndrome: Advances in Diagnosis and Treatment. Jama 2018, 319 (7), 698–710. 10.1001/jama.2017.21907. [DOI] [PubMed] [Google Scholar]
- Krzyzaniak M.; Cheadle G.; Peterson C.; Loomis W.; Putnam J.; Wolf P.; Baird A.; Eliceiri B.; Bansal V.; Coimbra R. Burn-induced acute lung injury requires a functional Toll-like receptor 4. Shock 2011, 36 (1), 24–29. 10.1097/SHK.0b013e318212276b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nanchal R. S.; Truwit J. D. Recent advances in understanding and treating acute respiratory distress syndrome. F1000Res 2018, 7, 1322. 10.12688/f1000research.15493.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camporota L.; Cronin J. N.; Busana M.; Gattinoni L.; Formenti F. Pathophysiology of coronavirus-19 disease acute lung injury. Curr. Opin Crit Care 2022, 28 (1), 9–16. 10.1097/MCC.0000000000000911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Standiford T. J.; Ward P. A. Therapeutic targeting of acute lung injury and acute respiratory distress syndrome. Transl Res. 2016, 167 (1), 183–191. 10.1016/j.trsl.2015.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang K.; Wang Y.; Cao Y.; Wang H.; Zhou Y.; Gao L.; Zeng Z.; Cheng M.; Jin X.; Chen J.; et al. Lumican is elevated in the lung in human and experimental acute respiratory distress syndrome and promotes early fibrotic responses to lung injury. J. Transl Med. 2022, 20 (1), 392. 10.1186/s12967-022-03597-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matuschak G. M.; Lechner A. J. Acute lung injury and the acute respiratory distress syndrome: pathophysiology and treatment. Mo Med. 2010, 107 (4), 252–258. [PMC free article] [PubMed] [Google Scholar]
- Lewis S. R.; Pritchard M. W.; Thomas C. M.; Smith A. F. Pharmacological agents for adults with acute respiratory distress syndrome. Cochrane Database Syst. Rev. 2019, 7 (7), Cd004477. 10.1002/14651858.CD004477.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luh S. P.; Chiang C. H. Acute lung injury/acute respiratory distress syndrome (ALI/ARDS): the mechanism, present strategies and future perspectives of therapies. J. Zhejiang Univ Sci. B 2007, 8 (1), 60–69. 10.1631/jzus.2007.B0060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beitler J. R.; Malhotra A.; Thompson B. T. Ventilator-induced Lung Injury. Clin Chest Med. 2016, 37 (4), 633–646. 10.1016/j.ccm.2016.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cruz F. F.; Ball L.; Rocco P. R. M.; Pelosi P. Ventilator-induced lung injury during controlled ventilation in patients with acute respiratory distress syndrome: less is probably better. Expert Rev. Respir Med. 2018, 12 (5), 403–414. 10.1080/17476348.2018.1457954. [DOI] [PubMed] [Google Scholar]
- Crescitelli R.; Lässer C.; Lötvall J. Isolation and characterization of extracellular vesicle subpopulations from tissues. Nat. Protoc 2021, 16 (3), 1548–1580. 10.1038/s41596-020-00466-1. [DOI] [PubMed] [Google Scholar]
- Pan B. T.; Johnstone R. M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell 1983, 33 (3), 967–978. 10.1016/0092-8674(83)90040-5. [DOI] [PubMed] [Google Scholar]
- Johnstone R. M.; Adam M.; Hammond J. R.; Orr L.; Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262 (19), 9412–9420. 10.1016/S0021-9258(18)48095-7. [DOI] [PubMed] [Google Scholar]
- Jiang K.; Yang J.; Guo S.; Zhao G.; Wu H.; Deng G. Peripheral Circulating Exosome-Mediated Delivery of miR-155 as a Novel Mechanism for Acute Lung Inflammation. Mol. Ther 2019, 27 (10), 1758–1771. 10.1016/j.ymthe.2019.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Juan C. X.; Mao Y.; Cao Q.; Chen Y.; Zhou L. B.; Li S.; Chen H.; Chen J. H.; Zhou G. P.; Jin R. Exosome-mediated pyroptosis of miR-93-TXNIP-NLRP3 leads to functional difference between M1 and M2 macrophages in sepsis-induced acute kidney injury. J. Cell Mol. Med. 2021, 25 (10), 4786–4799. 10.1111/jcmm.16449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lv L. L.; Feng Y.; Wu M.; Wang B.; Li Z. L.; Zhong X.; Wu W. J.; Chen J.; Ni H. F.; Tang T. T.; et al. Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury. Cell Death Differ. 2020, 27 (1), 210–226. 10.1038/s41418-019-0349-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dad H. A.; Gu T. W.; Zhu A. Q.; Huang L. Q.; Peng L. H. Plant Exosome-like Nanovesicles: Emerging Therapeutics and Drug Delivery Nanoplatforms. Mol. Ther 2021, 29 (1), 13–31. 10.1016/j.ymthe.2020.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan L.; Xu L.; Xu X.; Qin Z.; Zhou X.; Xiao Y.; Liang Y.; Xia J. Exosome-mediated delivery of gene vectors for gene therapy. Nanoscale 2021, 13 (3), 1387–1397. 10.1039/D0NR07622H. [DOI] [PubMed] [Google Scholar]
- Song Y.; Wang B.; Zhu X.; Hu J.; Sun J.; Xuan J.; Ge Z. Human umbilical cord blood-derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol. Toxicol 2021, 37 (1), 51–64. 10.1007/s10565-020-09530-8. [DOI] [PubMed] [Google Scholar]
- Yu W.; Hurley J.; Roberts D.; Chakrabortty S. K.; Enderle D.; Noerholm M.; Breakefield X. O.; Skog J. K. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann. Oncol 2021, 32 (4), 466–477. 10.1016/j.annonc.2021.01.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang B.; Chen Y.; Shi J. Exosome Biochemistry and Advanced Nanotechnology for Next-Generation Theranostic Platforms. Adv. Mater. 2019, 31 (2), e1802896. 10.1002/adma.201802896. [DOI] [PubMed] [Google Scholar]
- Rupaimoole R.; Slack F. J. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov 2017, 16 (3), 203–222. 10.1038/nrd.2016.246. [DOI] [PubMed] [Google Scholar]
- Zhou H.; Fan E. K.; Fan J. Cell-Cell Interaction Mechanisms in Acute Lung Injury. Shock 2021, 55 (2), 167–176. 10.1097/SHK.0000000000001598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shao H.; Im H.; Castro C. M.; Breakefield X.; Weissleder R.; Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem. Rev. 2018, 118 (4), 1917–1950. 10.1021/acs.chemrev.7b00534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai J. J.; Chau Z. L.; Chen S. Y.; Hill J. J.; Korpany K. V.; Liang N. W.; Lin L. H.; Lin Y. H.; Liu J. K.; Liu Y. C.; et al. Exosome Processing and Characterization Approaches for Research and Technology Development. Adv. Sci. (Weinh) 2022, 9 (15), e2103222. 10.1002/advs.202103222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Théry 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 (1), 1535750. 10.1080/20013078.2018.1535750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan B. T.; Johnstone R. Selective externalization of the transferrin receptor by sheep reticulocytes in vitro. Response to ligands and inhibitors of endocytosis. J. Biol. Chem. 1984, 259 (15), 9776–9782. 10.1016/S0021-9258(17)42767-0. [DOI] [PubMed] [Google Scholar]
- Johnstone R. M.; Bianchini A.; Teng K. Reticulocyte maturation and exosome release: transferrin receptor containing exosomes shows multiple plasma membrane functions. Blood 1989, 74 (5), 1844–1851. 10.1182/blood.V74.5.1844.bloodjournal7451844. [DOI] [PubMed] [Google Scholar]
- Catalano M.; O’Driscoll L. Inhibiting extracellular vesicles formation and release: a review of EV inhibitors. J. Extracell Vesicles 2020, 9 (1), 1703244. 10.1080/20013078.2019.1703244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pegtel D. M.; Gould S. J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. 10.1146/annurev-biochem-013118-111902. [DOI] [PubMed] [Google Scholar]
- Han Q. F.; Li W. J.; Hu K. S.; Gao J.; Zhai W. L.; Yang J. H.; Zhang S. J. Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer. Mol. Cancer 2022, 21 (1), 207. 10.1186/s12943-022-01671-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edgar J. R.; Eden E. R.; Futter C. E. Hrs- and CD63-dependent competing mechanisms make different sized endosomal intraluminal vesicles. Traffic 2014, 15 (2), 197–211. 10.1111/tra.12139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ostrowski M.; Carmo N. B.; Krumeich S.; Fanget I.; Raposo G.; Savina A.; Moita C. F.; Schauer K.; Hume A. N.; Freitas R. P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12 (1), 19–30. 10.1038/ncb2000. [DOI] [PubMed] [Google Scholar]
- Sinha S.; Hoshino D.; Hong N. H.; Kirkbride K. C.; Grega-Larson N. E.; Seiki M.; Tyska M. J.; Weaver A. M. Cortactin promotes exosome secretion by controlling branched actin dynamics. J. Cell Biol. 2016, 214 (2), 197–213. 10.1083/jcb.201601025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jung M. K.; Mun J. Y. Sample Preparation and Imaging of Exosomes by Transmission Electron Microscopy. J. Vis Exp 2018, 10.3791/56482-v. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cizmar P.; Yuana Y. Detection and Characterization of Extracellular Vesicles by Transmission and Cryo-Transmission Electron Microscopy. Methods Mol. Biol. 2017, 1660, 221–232. 10.1007/978-1-4939-7253-1_18. [DOI] [PubMed] [Google Scholar]
- Höög J. L.; Lötvall J. Diversity of extracellular vesicles in human ejaculates revealed by cryo-electron microscopy. J. Extracell Vesicles 2015, 4, 28680. 10.3402/jev.v4.28680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H.; Wang L.; Li C.; Yu Y.; Yi Y.; Wang J.; Chen D. Exosome-Induced Regulation in Inflammatory Bowel Disease. Front Immunol 2019, 10, 1464. 10.3389/fimmu.2019.01464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tao L.; Zhou J.; Yuan C.; Zhang L.; Li D.; Si D.; Xiu D.; Zhong L. Metabolomics identifies serum and exosomes metabolite markers of pancreatic cancer. Metabolomics 2019, 15 (6), 86. 10.1007/s11306-019-1550-1. [DOI] [PubMed] [Google Scholar]
- Jiang X.; Li J.; Zhang B.; Hu J.; Ma J.; Cui L.; Chen Z. J. Differential expression profile of plasma exosomal microRNAs in women with polycystic ovary syndrome. Fertil Steril 2021, 115 (3), 782–792. 10.1016/j.fertnstert.2020.08.019. [DOI] [PubMed] [Google Scholar]
- Zareba L.; Szymanski J.; Homoncik Z.; Czystowska-Kuzmicz M. EVs from BALF-Mediators of Inflammation and Potential Biomarkers in Lung Diseases. Int. J. Mol. Sci. 2021, 22, 3651. 10.3390/ijms22073651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li K.; Lin Y.; Luo Y.; Xiong X.; Wang L.; Durante K.; Li J.; Zhou F.; Guo Y.; Chen S.; et al. A signature of saliva-derived exosomal small RNAs as predicting biomarker for esophageal carcinoma: a multicenter prospective study. Mol. Cancer 2022, 21 (1), 21. 10.1186/s12943-022-01499-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Street J. M.; Koritzinsky E. H.; Glispie D. M.; Star R. A.; Yuen P. S. Urine Exosomes: An Emerging Trove of Biomarkers. Adv. Clin Chem. 2017, 78, 103–122. 10.1016/bs.acc.2016.07.003. [DOI] [PubMed] [Google Scholar]
- Roman-Canal B.; Tarragona J.; Moiola C. P.; Gatius S.; Bonnin S.; Ruiz-Miró M.; Sierra J. E.; Rufas M.; González E.; Porcel J. M.; et al. EV-associated miRNAs from peritoneal lavage as potential diagnostic biomarkers in colorectal cancer. J. Transl Med. 2019, 17 (1), 208. 10.1186/s12967-019-1954-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiong Y. Y.; Gong Z. T.; Tang R. J.; Yang Y. J. The pivotal roles of exosomes derived from endogenous immune cells and exogenous stem cells in myocardial repair after acute myocardial infarction. Theranostics 2021, 11 (3), 1046–1058. 10.7150/thno.53326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiao Y.; Zhang T.; Zhang C.; Ji H.; Tong X.; Xia R.; Wang W.; Ma Z.; Shi X. Exosomal miR-30d-5p of neutrophils induces M1 macrophage polarization and primes macrophage pyroptosis in sepsis-related acute lung injury. Crit Care 2021, 25 (1), 356. 10.1186/s13054-021-03775-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei K.; Ma Z.; Yang F.; Zhao X.; Jiang W.; Pan C.; Li Z.; Pan X.; He Z.; Xu J.; et al. M2 macrophage-derived exosomes promote lung adenocarcinoma progression by delivering miR-942. Cancer Lett. 2022, 526, 205–216. 10.1016/j.canlet.2021.10.045. [DOI] [PubMed] [Google Scholar]
- Pritchard A.; Tousif S.; Wang Y.; Hough K.; Khan S.; Strenkowski J.; Chacko B. K.; Darley-Usmar V. M.; Deshane J. S. Lung Tumor Cell-Derived Exosomes Promote M2Macrophage Polarization. Cells 2020, 9 (5), 1303. 10.3390/cells9051303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ju S.; Mu J.; Dokland T.; Zhuang X.; Wang Q.; Jiang H.; Xiang X.; Deng Z. B.; Wang B.; Zhang L.; et al. Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol. Ther 2013, 21 (7), 1345–1357. 10.1038/mt.2013.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fujita D.; Arai T.; Komori H.; Shirasaki Y.; Wakayama T.; Nakanishi T.; Tamai I. Apple-Derived Nanoparticles Modulate Expression of Organic-Anion-Transporting Polypeptide (OATP) 2B1 in Caco-2 Cells. Mol. Pharmaceutics 2018, 15 (12), 5772–5780. 10.1021/acs.molpharmaceut.8b00921. [DOI] [PubMed] [Google Scholar]
- Zhang M.; Xiao B.; Wang H.; Han M. K.; Zhang Z.; Viennois E.; Xu C.; Merlin D. Edible Ginger-derived Nano-lipids Loaded with Doxorubicin as a Novel Drug-delivery Approach for Colon Cancer Therapy. Mol. Ther 2016, 24 (10), 1783–1796. 10.1038/mt.2016.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raimondo S.; Naselli F.; Fontana S.; Monteleone F.; Lo Dico A.; Saieva L.; Zito G.; Flugy A.; Manno M.; Di Bella M. A.; et al. Citrus limon-derived nanovesicles inhibit cancer cell proliferation and suppress CML xenograft growth by inducing TRAIL-mediated cell death. Oncotarget 2015, 6 (23), 19514–19527. 10.18632/oncotarget.4004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colombo M.; Raposo G.; Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev Biol. 2014, 30, 255–289. 10.1146/annurev-cellbio-101512-122326. [DOI] [PubMed] [Google Scholar]
- Cordonnier M.; Chanteloup G.; Isambert N.; Seigneuric R.; Fumoleau P.; Garrido C.; Gobbo J. Exosomes in cancer theranostic: Diamonds in the rough. Cell Adh Migr 2017, 11 (2), 151–163. 10.1080/19336918.2016.1250999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao S.; Li W.; Yu W.; Rao T.; Li H.; Ruan Y.; Yuan R.; Li C.; Ning J.; Li S.; et al. Exosomal miR-21 from tubular cells contributes to renal fibrosis by activating fibroblasts via targeting PTEN in obstructed kidneys. Theranostics 2021, 11 (18), 8660–8673. 10.7150/thno.62820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C.; Guo F.; Chang M.; Zhou Z.; Yi L.; Gao C.; Huang X.; Huan J. Exosome-delivered syndecan-1 rescues acute lung injury via a FAK/p190RhoGAP/RhoA/ROCK/NF-κB signaling axis and glycocalyx enhancement. Exp. Cell Res. 2019, 384 (1), 111596. 10.1016/j.yexcr.2019.111596. [DOI] [PubMed] [Google Scholar]
- Sobo-Vujanovic A.; Munich S.; Vujanovic N. L. Dendritic-cell exosomes cross-present Toll-like receptor-ligands and activate bystander dendritic cells. Cell Immunol 2014, 289 (1–2), 119–127. 10.1016/j.cellimm.2014.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guan S.; Li Q.; Liu P.; Xuan X.; Du Y. Umbilical cord blood-derived dendritic cells loaded with BGC823 tumor antigens and DC-derived exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumor immunity in vitro and in vivo. Cent Eur. J. Immunol 2014, 39 (2), 142–151. 10.5114/ceji.2014.43713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiménez-Alesanco A.; Marcuello M.; Pastor-Jiménez M.; López-Puerto L.; Bonjoch L.; Gironella M.; Carrascal M.; Abian J.; de-Madaria E.; Closa D. Acute pancreatitis promotes the generation of two different exosome populations. Sci. Rep 2019, 9 (1), 19887. 10.1038/s41598-019-56220-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barros F. M.; Carneiro F.; Machado J. C.; Melo S. A. Exosomes and Immune Response in Cancer: Friends or Foes?. Front Immunol 2018, 9, 730. 10.3389/fimmu.2018.00730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pei W.; Li X.; Bi R.; Zhang X.; Zhong M.; Yang H.; Zhang Y.; Lv K. Exosome membrane-modified M2 macrophages targeted nanomedicine: Treatment for allergic asthma. J. Controlled Release 2021, 338, 253–267. 10.1016/j.jconrel.2021.08.024. [DOI] [PubMed] [Google Scholar]
- Zhao T.; Sun F.; Liu J.; Ding T.; She J.; Mao F.; Xu W.; Qian H.; Yan Y. Emerging Role of Mesenchymal Stem Cell-derived Exosomes in Regenerative Medicine. Curr. Stem Cell Res. Ther 2019, 14 (6), 482–494. 10.2174/1574888x14666190228103230. [DOI] [PubMed] [Google Scholar]
- Ferragut Cardoso A. P.; Banerjee M.; Nail A. N.; Lykoudi A.; States J. C. miRNA dysregulation is an emerging modulator of genomic instability. Semin Cancer Biol. 2021, 76, 120–131. 10.1016/j.semcancer.2021.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michlewski G.; Cáceres J. F. Post-transcriptional control of miRNA biogenesis. Rna 2019, 25 (1), 1–16. 10.1261/rna.068692.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ha M.; Kim V. N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15 (8), 509–524. 10.1038/nrm3838. [DOI] [PubMed] [Google Scholar]
- García-López J.; Brieño-Enríquez M. A.; Del Mazo J. MicroRNA biogenesis and variability. Biomol Concepts 2013, 4 (4), 367–380. 10.1515/bmc-2013-0015. [DOI] [PubMed] [Google Scholar]
- Kim V. N. MicroRNA precursors in motion: exportin-5 mediates their nuclear export. Trends Cell Biol. 2004, 14 (4), 156–159. 10.1016/j.tcb.2004.02.006. [DOI] [PubMed] [Google Scholar]
- Peng Y.; Croce C. M. The role of MicroRNAs in human cancer. Signal Transduct Target Ther 2016, 1, 15004. 10.1038/sigtrans.2015.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng D.; Huo M.; Li B.; Wang W.; Piao H.; Wang Y.; Zhu Z.; Li D.; Wang T.; Liu K. The Role of Exosomes and Exosomal MicroRNA in Cardiovascular Disease. Front Cell Dev Biol. 2020, 8, 616161. 10.3389/fcell.2020.616161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Macfarlane L. A.; Murphy P. R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genomics 2010, 11 (7), 537–561. 10.2174/138920210793175895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montecalvo A.; Larregina A. T.; Shufesky W. J.; Stolz D. B.; Sullivan M. L.; Karlsson J. M.; Baty C. J.; Gibson G. A.; Erdos G.; Wang Z.; et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 2012, 119 (3), 756–766. 10.1182/blood-2011-02-338004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noonin C.; Thongboonkerd V. Exosome-inflammasome crosstalk and their roles in inflammatory responses. Theranostics 2021, 11 (9), 4436–4451. 10.7150/thno.54004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B.; Cao Y.; Sun M.; Feng H. Expression, regulation, and function of exosome-derived miRNAs in cancer progression and therapy. Faseb j 2021, 35 (10), e21916. 10.1096/fj.202100294RR. [DOI] [PubMed] [Google Scholar]
- Isaac R.; Reis F. C. G.; Ying W.; Olefsky J. M. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab 2021, 33 (9), 1744–1762. 10.1016/j.cmet.2021.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Im H.; Lee K.; Weissleder R.; Lee H.; Castro C. M. Novel nanosensing technologies for exosome detection and profiling. Lab Chip 2017, 17 (17), 2892–2898. 10.1039/c7lc00247e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalra H.; Adda C. G.; Liem M.; Ang C. S.; Mechler A.; Simpson R. J.; Hulett M. D.; Mathivanan S. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics 2013, 13 (22), 3354–3364. 10.1002/pmic.201300282. [DOI] [PubMed] [Google Scholar]
- Ge Q.; Zhou Y.; Lu J.; Bai Y.; Xie X.; Lu Z. miRNA in plasma exosome is stable under different storage conditions. Molecules 2014, 19 (2), 1568–1575. 10.3390/molecules19021568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Worrell J. C.; MacLeod M. K. L. Stromal-immune cell crosstalk fundamentally alters the lung microenvironment following tissue insult. Immunology 2021, 163 (3), 239–249. 10.1111/imm.13319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Waal A. M.; Hiemstra P. S.; Ottenhoff T. H.; Joosten S. A.; van der Does A. M. Lung epithelial cells interact with immune cells and bacteria to shape the microenvironment in tuberculosis. Thorax 2022, 77 (4), 408–416. 10.1136/thoraxjnl-2021-217997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao X.; Wu Y.; Shen F.; MuLaTiAize Y.; Xinhua N. Osimertinib Improves the Immune Microenvironment of Lung Cancer by Downregulating PD-L1 Expression of Vascular Endothelial Cells and Enhances the Antitumor Effect of Bevacizumab. J. Oncol 2022, 2022, 1531353. 10.1155/2022/1531353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christenson J. L.; Williams M. M.; Richer J. K. The underappreciated role of resident epithelial cell populations in metastatic progression: contributions of the lung alveolar epithelium. Am. J. Physiol Cell Physiol 2022, 323 (6), C1777–c1790. 10.1152/ajpcell.00181.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McQuattie-Pimentel A. C.; Ren Z.; Joshi N.; Watanabe S.; Stoeger T.; Chi M.; Lu Z.; Sichizya L.; Aillon R. P.; Chen C. I. The lung microenvironment shapes a dysfunctional response of alveolar macrophages in aging. J. Clin Invest 2021, 10.1172/jci140299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ancey P. B.; Contat C.; Boivin G.; Sabatino S.; Pascual J.; Zangger N.; Perentes J. Y.; Peters S.; Abel E. D.; Kirsch D. G.; et al. GLUT1 Expression in Tumor-Associated Neutrophils Promotes Lung Cancer Growth and Resistance to Radiotherapy. Cancer Res. 2021, 81 (9), 2345–2357. 10.1158/0008-5472.Can-20-2870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yunna C.; Mengru H.; Lei W.; Weidong C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. 10.1016/j.ejphar.2020.173090. [DOI] [PubMed] [Google Scholar]
- Liu F.; Peng W.; Chen J.; Xu Z.; Jiang R.; Shao Q.; Zhao N.; Qian K. Exosomes Derived From Alveolar Epithelial Cells Promote Alveolar Macrophage Activation Mediated by miR-92a-3p in Sepsis-Induced Acute Lung Injury. Front Cell Infect Microbiol 2021, 11, 646546. 10.3389/fcimb.2021.646546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahesh G.; Biswas R. MicroRNA-155: A Master Regulator of Inflammation. J. Interferon Cytokine Res. 2019, 39 (6), 321–330. 10.1089/jir.2018.0155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corcoran K.; Jabbour M.; Bhagwandin C.; Deymier M. J.; Theisen D. L.; Lybarger L. Ubiquitin-mediated regulation of CD86 protein expression by the ubiquitin ligase membrane-associated RING-CH-1 (MARCH1). J. Biol. Chem. 2011, 286 (43), 37168–37180. 10.1074/jbc.M110.204040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D.; Lee H.; Wang X.; Groot M.; Sharma L.; Dela Cruz C. S.; Jin Y. Apotential role of microvesicle-containing miR-223/142 in lung inflammation. Thorax 2019, 74 (9), 865–874. 10.1136/thoraxjnl-2018-212994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu F.; Yang Y.; Peng W.; Zhao N.; Chen J.; Xu Z.; Cui Y.; Qian K. Mitophagy-promoting miR-138–5p promoter demethylation inhibits pyroptosis in sepsis-associated acute lung injury. Inflamm Res. 2023, 72 (2), 329–346. 10.1007/s00011-022-01675-y. [DOI] [PubMed] [Google Scholar]
- Whitsett J. A.; Alenghat T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat. Immunol 2015, 16 (1), 27–35. 10.1038/ni.3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guillot L.; Nathan N.; Tabary O.; Thouvenin G.; Le Rouzic P.; Corvol H.; Amselem S.; Clement A. Alveolar epithelial cells: master regulators of lung homeostasis. Int. J. Biochem. Cell Biol. 2013, 45 (11), 2568–2573. 10.1016/j.biocel.2013.08.009. [DOI] [PubMed] [Google Scholar]
- Stone K. C.; Mercer R. R.; Gehr P.; Stockstill B.; Crapo J. D. Allometric relationships of cell numbers and size in the mammalian lung. Am. J. Respir. Cell Mol. Biol. 1992, 6 (2), 235–243. 10.1165/ajrcmb/6.2.235. [DOI] [PubMed] [Google Scholar]
- Yang Z.; Huang S.; Zhao L. Suppressor of cytokine signaling 6 in cancer development and therapy: Deciphering its emerging and suppressive roles. Cytokine Growth Factor Rev. 2022, 64, 21–32. 10.1016/j.cytogfr.2022.02.001. [DOI] [PubMed] [Google Scholar]
- Ma J.; Xu L. Y.; Sun Q. H.; Wan X. Y.; Bing Li. Inhibition of miR-1298–5p attenuates sepsis lung injury by targeting SOCS6. Mol. Cell. Biochem. 2021, 476 (10), 3745–3756. 10.1007/s11010-021-04170-w. [DOI] [PubMed] [Google Scholar]
- Kent O. A.; McCall M. N.; Cornish T. C.; Halushka M. K. Lessons from miR-143/145: the importance of cell-type localization of miRNAs. Nucleic Acids Res. 2014, 42 (12), 7528–7538. 10.1093/nar/gku461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao X.; Zhang C.; Zhang X.; Chen Y.; Zhang H. MiR-145 negatively regulates TGFBR2 signaling responsible for sepsis-induced acute lung injury. Biomed Pharmacother 2019, 111, 852–858. 10.1016/j.biopha.2018.12.138. [DOI] [PubMed] [Google Scholar]
- Krüger-Genge A.; Blocki A.; Franke R. P.; Jung F. Vascular Endothelial Cell Biology: An Update. Int. J. Mol. Sci. 2019, 10.3390/ijms20184411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hao Y.; Wang Z.; Frimpong F.; Chen X. Calcium-Permeable Channels and Endothelial Dysfunction in Acute Lung Injury. Curr. Issues Mol. Biol. 2022, 44 (5), 2217–2229. 10.3390/cimb44050150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao M.; Yu T.; Liu D.; Shi Y.; Yang P.; Zhang J.; Wang J.; Liu Y.; Zhang X. Sepsis plasma-derived exosomal miR-1–3p induces endothelial cell dysfunction by targeting SERP1. Clin Sci. (Lond) 2021, 135 (2), 347–365. 10.1042/cs20200573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polidoro R. B.; Hagan R. S.; de Santis Santiago R.; Schmidt N. W. Overview: Systemic Inflammatory Response Derived From Lung Injury Caused by SARS-CoV-2 Infection Explains Severe Outcomes in COVID-19. Front Immunol 2020, 11, 1626. 10.3389/fimmu.2020.01626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lascano J.; Oshins R.; Eagan C.; Wadood Z.; Qiang X.; Flagg T.; Scindia Y.; Mehrad B.; Brantly M.; Khodayari N. Correlation of alpha-1 antitrypsin levels and exosome associated neutrophil elastase endothelial injury in subjects with SARS-CoV2 infection. PLoS One 2022, 17 (9), e0274427. 10.1371/journal.pone.0274427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu X.; Wu C.; Gu W.; Ji H.; Zhu L. Serum Exosomal MicroRNAs Predict Acute Respiratory Distress Syndrome Events in Patients with Severe Community-Acquired Pneumonia. Biomed Res. Int. 2019, 2019, 3612020. 10.1155/2019/3612020. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Sun Y.; Liu X. L.; Zhang D.; Liu F.; Cheng Y. J.; Ma Y.; Zhou Y. J.; Zhao Y. X. Platelet-Derived Exosomes Affect the Proliferation and Migration of Human Umbilical Vein Endothelial Cells Via miR-126. Curr. Vasc Pharmacol 2019, 17 (4), 379–387. 10.2174/1570161116666180313142139. [DOI] [PubMed] [Google Scholar]
- Szturmowicz M.; Demkow U. Neutrophil Extracellular Traps (NETs) in Severe SARS-CoV-2 Lung Disease. Int. J. Mol. Sci. 2021, 10.3390/ijms22168854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li R. H. L.; Tablin F. AComparative Review of Neutrophil Extracellular Traps in Sepsis. Front Vet Sci. 2018, 5, 291. 10.3389/fvets.2018.00291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiao Y.; Li W.; Wang W.; Tong X.; Xia R.; Fan J.; Du J.; Zhang C.; Shi X. Platelet-derived exosomes promote neutrophil extracellular trap formation during septic shock. Crit Care 2020, 24 (1), 380. 10.1186/s13054-020-03082-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 (2), 600–620. 10.1016/j.apsb.2021.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanasty R.; Dorkin J. R.; Vegas A.; Anderson D. Delivery materials for siRNA therapeutics. Nat. Mater. 2013, 12 (11), 967–977. 10.1038/nmat3765. [DOI] [PubMed] [Google Scholar]
- Tian T.; Zhang H. X.; He C. P.; Fan S.; Zhu Y. L.; Qi C.; Huang N. P.; Xiao Z. D.; Lu Z. H.; Tannous B. A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018, 150, 137–149. 10.1016/j.biomaterials.2017.10.012. [DOI] [PubMed] [Google Scholar]
- Schindler C.; Collinson A.; Matthews C.; Pointon A.; Jenkinson L.; Minter R. R.; Vaughan T. J.; Tigue N. J. Exosomal delivery of doxorubicin enables rapid cell entry and enhanced in vitro potency. PLoS One 2019, 14 (3), e0214545. 10.1371/journal.pone.0214545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng Y.; Xu Q.; Yang Y.; Shi W.; Meng W.; Zhang H.; He X.; Sun M.; Chen Y.; Zhao J.; et al. The therapeutic effects of bone marrow-derived mesenchymal stromal cells in the acute lung injury induced by sulfur mustard. Stem Cell Res. Ther 2019, 10 (1), 90. 10.1186/s13287-019-1189-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaspi H.; Semo J.; Abramov N.; Dekel C.; Lindborg S.; Kern R.; Lebovits C.; Aricha R. MSC-NTF (NurOwn®) exosomes: a novel therapeutic modality in the mouse LPS-induced ARDS model. Stem Cell Res. Ther 2021, 12 (1), 72. 10.1186/s13287-021-02143-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen L.; Hou Y.; Du D.; Cui Y.; Nie H.; Ding Y. MiR-199a-3p in mouse bone marrow mesenchymal stem cell exosomes increases epithelial sodium channel expression in lung injury. Fundam Clin Pharmacol 2022, 10.1111/fcp.12807. [DOI] [PubMed] [Google Scholar]
- Yi X.; Wei X.; Lv H.; An Y.; Li L.; Lu P.; Yang Y.; Zhang Q.; Yi H.; Chen G. Exosomes derived from microRNA-30b-3p-overexpressing mesenchymal stem cells protect against lipopolysaccharide-induced acute lung injury by inhibiting SAA3. Exp. Cell Res. 2019, 383 (2), 111454. 10.1016/j.yexcr.2019.05.035. [DOI] [PubMed] [Google Scholar]
- Liu J. H.; Li C.; Cao L.; Zhang C. H.; Zhang Z. H. Exosomal miR-132–3p from mesenchymal stem cells alleviated LPS-induced acute lung injury by repressing TRAF6. Autoimmunity 2021, 54 (8), 493–503. 10.1080/08916934.2021.1966768. [DOI] [PubMed] [Google Scholar]
- 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-κB/NLRP3-mediated epithelial-mesenchymal transition and inflammation. Cell Death Dis 2020, 11 (11), 978. 10.1038/s41419-020-03178-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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-κB and Hedgehog pathways in LPS-induced acute lung injury. Cell Death Dis 2020, 11 (10), 863. 10.1038/s41419-020-03034-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J.; Xu D.; Yu Z.; Fu Z.; Lv Z.; Meng L.; Zhao X. Exosomal miR-150 partially attenuated acute lung injury by mediating microvascular endothelial cells and MAPK pathway. Biosci Rep 2022, 10.1042/bsr20203363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X.; Gao C.; Wang Y.; Niu L.; Jiang S.; Pan S. BMSC-Derived Exosomes Ameliorate LPS-Induced Acute Lung Injury by miR-384–5p-Controlled Alveolar Macrophage Autophagy. Oxid Med. Cell Longev 2021, 2021, 9973457. 10.1155/2021/9973457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng X.; Zhang F.; Zhao F.; Li L.; Huang H.; Li L.; Yi Y.; Yin H.; Xu J. Exosomal miR-127–5p from BMSCs alleviated sepsis-related acute lung injury by inhibiting neutrophil extracellular trap formation. Int. Immunopharmacol 2023, 123, 110759. 10.1016/j.intimp.2023.110759. [DOI] [PubMed] [Google Scholar]
- Peace C. G.; O’Neill L. A. The role of itaconate in host defense and inflammation. J. Clin Invest 2022, 10.1172/jci148548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su Y.; Silva J. D.; Doherty D.; Simpson D. A.; Weiss D. J.; Rolandsson-Enes S.; McAuley D. F.; O’Kane C. M.; Brazil D. P.; Krasnodembskaya A. D. Mesenchymal stromal cells-derived extracellular vesicles reprogramme macrophages in ARDS models through the miR-181a-5p-PTEN-pSTAT5-SOCS1 axis. Thorax 2023, 78 (6), 617–630. 10.1136/thoraxjnl-2021-218194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y.; Li J.; Yuan R.; Deng Z.; Wu X. Bone marrow mesenchymal stem cell-derived exosomes alleviate hyperoxia-induced lung injury via the manipulation of microRNA-425. Arch. Biochem. Biophys. 2021, 697, 108712. 10.1016/j.abb.2020.108712. [DOI] [PubMed] [Google Scholar]
- Li J. W.; Wei L.; Han Z.; Chen Z. Mesenchymal stromal cells-derived exosomes alleviate ischemia/reperfusion injury in mouse lung by transporting anti-apoptotic miR-21–5p. Eur. J. Pharmacol. 2019, 852, 68–76. 10.1016/j.ejphar.2019.01.022. [DOI] [PubMed] [Google Scholar]
- Sun Z. L.; You T.; Zhang B. H.; Liu Y.; Liu J. Bone marrow mesenchymal stem cell-derived exosomes miR-202–5p inhibited pyroptosis to alleviate lung ischemic-reperfusion injury by targeting CMPK2. Kaohsiung J. Med. Sci. 2023, 39 (7), 688–698. 10.1002/kjm2.12688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li T.; Xia M.; Gao Y.; Chen Y.; Xu Y. Human umbilical cord mesenchymal stem cells: an overview of their potential in cell-based therapy. Expert Opin Biol. Ther 2015, 15 (9), 1293–1306. 10.1517/14712598.2015.1051528. [DOI] [PubMed] [Google Scholar]
- Zheng Y.; Liu J.; Chen P.; Lin L.; Luo Y.; Ma X.; Lin J.; Shen Y.; Zhang L. 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. 10.1016/j.lfs.2020.119004. [DOI] [PubMed] [Google Scholar]
- Wei X.; Yi X.; Lv H.; Sui X.; Lu P.; Li L.; An Y.; Yang Y.; Yi H.; Chen G. MicroRNA-377–3p released by mesenchymal stem cell exosomes ameliorates lipopolysaccharide-induced acute lung injury by targeting RPTOR to induce autophagy. Cell Death Dis 2020, 11 (8), 657. 10.1038/s41419-020-02857-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong C.; Gu Z.; Zhang X.; Xu Q.; Mao G.; Pei Z.; Meng W.; Cen J.; Liu J.; He X.; et al. HMSCs exosome-derived miR-199a-5p attenuates sulfur mustard-associated oxidative stress via the CAV1/NRF2 signalling pathway. J. Cell Mol. Med. 2023, 27 (15), 2165–2182. 10.1111/jcmm.17803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J. S.; Du J.; Cheng X.; Zhang X. Z.; Li Y.; Chen X. L. Exosomal miR-451 from human umbilical cord mesenchymal stem cells attenuates burn-induced acute lung injury. J. Chin Med. Assoc 2019, 82 (12), 895–901. 10.1097/jcma.0000000000000189. [DOI] [PubMed] [Google Scholar]
- Liu J.; Xing F.; Fu Q.; He B.; Jia Z.; Du J.; Li Y.; Zhang X.; Chen X. hUC-MSCs exosomal miR-451 alleviated acute lung injury by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway. Environ. Toxicol 2022, 10.1002/tox.23639. [DOI] [PubMed] [Google Scholar]
- Chen X.; Su C.; Wei Q.; Sun H.; Xie J.; Nong G. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Diffuse Alveolar Hemorrhage Associated with Systemic Lupus Erythematosus in Mice by Promoting M2Macrophage Polarization via the microRNA-146a-5p/NOTCH1 Axis. Immunol Invest 2022, 1–19. 10.1080/08820139.2022.2090261. [DOI] [PubMed] [Google Scholar]
- Mao M.; Xu X.; Zhang Y.; Zhang B.; Fu Z. H. Endothelial progenitor cells: the promise of cell-based therapies for acute lung injury. Inflamm Res. 2013, 62 (1), 3–8. 10.1007/s00011-012-0570-3. [DOI] [PubMed] [Google Scholar]
- Wu X.; Liu Z.; Hu L.; Gu W.; Zhu L. Exosomes derived from endothelial progenitor cells ameliorate acute lung injury by transferring miR-126. Exp. Cell Res. 2018, 370 (1), 13–23. 10.1016/j.yexcr.2018.06.003. [DOI] [PubMed] [Google Scholar]
- Zhou Y.; Li P.; Goodwin A. J.; Cook J. A.; Halushka P. V.; Chang E.; Zingarelli B.; Fan H. Exosomes from endothelial progenitor cells improve outcomes of the lipopolysaccharide-induced acute lung injury. Crit Care 2019, 23 (1), 44. 10.1186/s13054-019-2339-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y.; Luo T.; Li H.; Zhao X.; Zhou M.; Cheng M. Protective effect of endothelial progenitor cell-derived exosomal microRNA-382–3p on sepsis-induced organ damage and immune suppression in mice. Am. J. Transl Res. 2022, 14 (10), 6856–6873. [PMC free article] [PubMed] [Google Scholar]
- Mazini L.; Rochette L.; Admou B.; Amal S.; Malka G. Hopes and Limits of Adipose-Derived Stem Cells (ADSCs) and Mesenchymal Stem Cells (MSCs) in Wound Healing. Int. J. Mol. Sci. 2020, 10.3390/ijms21041306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai W. X.; Shen K.; Cao T.; Wang J.; Zhao M.; Wang K. J.; Zhang Y.; Han J. T.; Hu D. H.; Tao K. [Effects of exosomes from human adipose-derived mesenchymal stem cells on pulmonary vascular endothelial cells injury in septic mice and its mechanism]. Zhonghua Shao Shang Za Zhi 2022, 38 (3), 266–275. 10.3760/cma.j.cn501120-20211020-00362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen K.; Wang X.; Wang Y.; Jia Y.; Zhang Y.; Wang K.; Luo L.; Cai W.; Li J.; Li S.; et al. miR-125b-5p in adipose derived stem cells exosome alleviates pulmonary microvascular endothelial cells ferroptosis via Keap1/Nrf2/GPX4 in sepsis lung injury. Redox Biol. 2023, 62, 102655. 10.1016/j.redox.2023.102655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J.; Baalachandran R.; Li Y.; Zhang C. O.; Ke Y.; Karki P.; Birukov K. G.; Birukova A. A. Circulating extracellular histones exacerbate acute lung injury by augmenting pulmonary endothelial dysfunction via TLR4-dependent mechanism. Am. J. Physiol Lung Cell Mol. Physiol 2022, 323 (3), L223–l239. 10.1152/ajplung.00072.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y.; Wan D.; Luo X.; Song T.; Wang Y.; Yu Q.; Jiang L.; Liao R.; Zhao W.; Su B. Circulating Histones in Sepsis: Potential Outcome Predictors and Therapeutic Targets. Front Immunol 2021, 12, 650184. 10.3389/fimmu.2021.650184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richards C. M.; McRae S. A.; Ranger A. L.; Klegeris A. Extracellular histones as damage-associated molecular patterns in neuroinflammatory responses. Rev. Neurosci 2022, 10.1515/revneuro-2022-0091. [DOI] [PubMed] [Google Scholar]
- Mizuta Y.; Akahoshi T.; Guo J.; Zhang S.; Narahara S.; Kawano T.; Murata M.; Tokuda K.; Eto M.; Hashizume M.; et al. Exosomes from adipose tissue-derived mesenchymal stem cells ameliorate histone-induced acute lung injury by activating the PI3K/Akt pathway in endothelial cells. Stem Cell Res. Ther 2020, 11 (1), 508. 10.1186/s13287-020-02015-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He N.; Tan H.; Deng X.; Shu L.; Qing B.; Liang H. MiR-223–3p-loaded exosomes from bronchoalveolar lavage fluid promote alveolar macrophage autophagy and reduce acute lung injury by inhibiting the expression of STK39. Hum Cell 2022, 10.1007/s13577-022-00762-w. [DOI] [PubMed] [Google Scholar]
- Li R.; Liang P.; Yuan J.; He F. Exosomal miR-103a-3p ameliorates lipopolysaccharide-induced immune response in BEAS-2B cells via NF-κB pathway by targeting transducin β-like 1X related protein 1. Clin. Exp. Pharmacol. Physiol. 2020, 47 (4), 620–627. 10.1111/1440-1681.13241. [DOI] [PubMed] [Google Scholar]
- Zheng L.; Su J.; Zhang Z.; Jiang L.; Wei J.; Xu X.; Lv S. Salidroside regulates inflammatory pathway of alveolar macrophages by influencing the secretion of miRNA-146a exosomes by lung epithelial cells. Sci. Rep 2020, 10 (1), 20750. 10.1038/s41598-020-77448-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang L.; Ni J.; Shen G.; Xia Z.; Zhang L.; Xia S.; Pan S.; Qu H.; Li X. Upregulation of endothelial cell-derived exosomal microRNA-125b-5p protects from sepsis-induced acute lung injury by inhibiting topoisomerase II alpha. Inflamm Res. 2021, 70 (2), 205–216. 10.1007/s00011-020-01415-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quan Y.; Wang Z.; Gong L.; Peng X.; Richard M. A.; Zhang J.; Fornage M.; Alcorn J. L.; Wang D. Exosome miR-371b-5p promotes proliferation of lung alveolar progenitor type II cells by using PTEN to orchestrate the PI3K/Akt signaling. Stem Cell Res. Ther 2017, 8 (1), 138. 10.1186/s13287-017-0586-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin Y.; Qian F.; Shen L.; Chen F.; Chen J.; Shen B. Computer-aided biomarker discovery for precision medicine: data resources, models and applications. Brief Bioinform 2019, 20 (3), 952–975. 10.1093/bib/bbx158. [DOI] [PubMed] [Google Scholar]
- He B.; Zhao Z.; Cai Q.; Zhang Y.; Zhang P.; Shi S.; Xie H.; Peng X.; Yin W.; Tao Y.; et al. miRNA-based biomarkers, therapies, and resistance in Cancer. Int. J. Biol. Sci. 2020, 16 (14), 2628–2647. 10.7150/ijbs.47203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Backes C.; Meese E.; Keller A. Specific miRNA Disease Biomarkers in Blood, Serum and Plasma: Challenges and Prospects. Mol. Diagn Ther 2016, 20 (6), 509–518. 10.1007/s40291-016-0221-4. [DOI] [PubMed] [Google Scholar]
- Kilchert C.; Wittmann S.; Vasiljeva L. The regulation and functions of the nuclear RNA exosome complex. Nat. Rev. Mol. Cell Biol. 2016, 17 (4), 227–239. 10.1038/nrm.2015.15. [DOI] [PubMed] [Google Scholar]
- Salido-Guadarrama I.; Romero-Cordoba S.; Peralta-Zaragoza O.; Hidalgo-Miranda A.; Rodríguez-Dorantes M. MicroRNAs transported by exosomes in body fluids as mediators of intercellular communication in cancer. Onco Targets Ther 2014, 7, 1327–1338. 10.2147/ott.S61562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu D.; Li Y.; Wang M.; Gu J.; Xu W.; Cai H.; Fang X.; Zhang X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21 (1), 56. 10.1186/s12943-022-01509-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sandfeld-Paulsen B.; Jakobsen K. R.; Bæk R.; Folkersen B. H.; Rasmussen T. R.; Meldgaard P.; Varming K.; Jørgensen M. M.; Sorensen B. S. Exosomal Proteins as Diagnostic Biomarkers in Lung Cancer. J. Thorac Oncol 2016, 11 (10), 1701–1710. 10.1016/j.jtho.2016.05.034. [DOI] [PubMed] [Google Scholar]
- Parzibut G.; Henket M.; Moermans C.; Struman I.; Louis E.; Malaise M.; Louis R.; Misset B.; Njock M. S.; Guiot J. ABlood Exosomal miRNA Signature in Acute Respiratory Distress Syndrome. Front Mol. Biosci 2021, 8, 640042. 10.3389/fmolb.2021.640042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahida R. Y.; Price J.; Lugg S. T.; Li H.; Parekh D.; Scott A.; Harrison P.; Matthay M. A.; Thickett D. R. CD14-positive extracellular vesicles in bronchoalveolar lavage fluid as a new biomarker of acute respiratory distress syndrome. Am. J. Physiol Lung Cell Mol. Physiol 2022, 322 (4), L617–l624. 10.1152/ajplung.00052.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao M.; Li Y. P.; Geng X. R.; Zhao M.; Ma S. B.; Yang Y. H.; Deng Z. H.; Luo L. M.; Pan X. Q. Expression Level of MiRNA-126 in Serum Exosomes of Allergic Asthma Patients and Lung Tissues of Asthmatic Mice. Curr. Drug Metab 2019, 20 (10), 799–803. 10.2174/1389200220666191011114452. [DOI] [PubMed] [Google Scholar]
- Bahmer T.; Krauss-Etschmann S.; Buschmann D.; Behrends J.; Watz H.; Kirsten A. M.; Pedersen F.; Waschki B.; Fuchs O.; Pfaffl M. W.; et al. RNA-seq-based profiling of extracellular vesicles in plasma reveals a potential role of miR-122–5p in asthma. Allergy 2021, 76 (1), 366–371. 10.1111/all.14486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maes T.; Cobos F. A.; Schleich F.; Sorbello V.; Henket M.; De Preter K.; Bracke K. R.; Conickx G.; Mesnil C.; Vandesompele J.; et al. Asthma inflammatory phenotypes show differential microRNA expression in sputum. J. Allergy Clin Immunol 2016, 137 (5), 1433–1446. 10.1016/j.jaci.2016.02.018. [DOI] [PubMed] [Google Scholar]
- Smith G. J.; Tovar A.; Kanke M.; Wang Y.; Deshane J. S.; Sethupathy P.; Kelada S. N. P. Ozone-induced changes in the murine lung extracellular vesicle small RNA landscape. Physiol Rep 2021, 9 (18), e15054. 10.14814/phy2.15054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C.; Bai H.; Zhang L.; Zhang Y.; Chen X.; Shi R.; Zhang G.; Xu Q.; Lin G. Differential expression profile of plasma exosomal microRNAs in acute type A aortic dissection with acute lung injury. Sci. Rep 2022, 12 (1), 11667. 10.1038/s41598-022-15859-3. [DOI] [PMC free article] [PubMed] [Google Scholar]