Abstract

Ischemic stroke and acute lung injury are prevalent life-threatening conditions marked by intricate molecular mechanisms and elevated mortality rates. Despite evident pathophysiological distinctions, a notable similarity exists in the gene responses to tissue injury observed in both pathologies. This similarity extends to both protein-encoding RNAs and non-coding RNAs. Extracellular vesicles (EVs) are nano-scale vesicles derived through cell secretion, possessing unique advantages such as high biocompatibility, low immunogenicity, intrinsic cell targeting, and facile chemical and genetic manipulation. Importantly, miRNAs, the most prevalent non-coding RNAs, are selectively concentrated within EVs. Macrophages/microglia serve as immune defense and homeostatic cells, deriving from progenitor cells in the bone marrow. They can be classified into two contrasting types: classical proinflammatory M1 phenotype or alternative anti-inflammatory M2 phenotype. However, there exists a continuum of various intermediate phenotypes between M1 and M2, and macrophages/microglia can transition from one phenotype to another. This review will investigate recent discoveries concerning the impact of EVs derived from macrophages/microglia under various states on the progression of ischemic stroke and acute lung injury. The focus will be on the involvement of miRNAs within these vesicles. The concluding remarks of this review will underscore the clinical possibilities linked to EV-miRNAs, accentuating their potential as both biomarkers and therapeutic targets.

Introduction

Ischemic stroke and acute lung injury are common life-threatening diseases associated with severe inflammation, characterized by complicated molecular mechanisms and high mortality [1–3]. Acute lung injury refers to a sudden onset of hypoxic respiratory insufficiency resulting from diverse factors, both intra and extra-pulmonary. These factors include sepsis, trauma, ischemia-reperfusion, pneumonia, and mechanical ventilation. The condition leads to the breakdown of the barrier formed by capillary endothelial cells and alveolar epithelial cells [4, 5]. Ischemic stroke occurs when cerebrovascular diseases, often linked to vascular occlusions and atherosclerosis, interrupt blood supply to the brain, resulting in ischemic hypoxic necrosis within brain tissues [6, 7]. Ischemic stroke remains among the most incapacitating illnesses and a primary contributor to global mortality [8]. Despite advancements in medical interventions, the worldwide impact of stroke continues to escalate, given the absence of effective treatments capable of mitigating or reversing brain ischemic injury [8]. This initiates a cascade of inflammatory reactions, causing early-stage damage to both the brain and the blood-brain barrier [9, 10]. Despite evident pathophysiological distinctions, there is a significant resemblance in the gene responses to tissue injury across these two pathologies. These similarities extend to both protein-encoding RNAs and non-coding RNAs (ncRNAs) [11, 12].

Targeting ncRNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), in a therapeutic context holds promise for addressing not only ischemic stroke but also acute lung injury [13, 14]. Due to alterations in gene expression, the fundamental aspects of cellular biology and communication undergo significant changes in injured conditions. Cell–cell communication has been demonstrated through various mechanisms, with the latest being the release of extracellular vesicles (EVs) [12]. As EVs transport a diverse range of cargoes, including nucleic acids, lipids, and proteins, they have become a focal point in the fields of diagnosis and treatment [12]. Significantly, miRNAs, the most prevalent ncRNAs [15], are selectively concentrated within EVs [16]. The transferred miRNAs in EVs play a crucial role in regulating diverse aspects of the onset and progression of ischemic stroke and acute lung injury, underscoring their significance as essential components.

In the realm of innate immunity, macrophages/microglia play a crucial role as versatile phagocytes. They express pattern recognition receptors to identify both pathogen-associated molecular patterns and damage-associated molecular patterns. Macrophages/microglia engage in communication with other cells within the tissue through cell-to-cell contact, secretion of molecules, and the release of EVs. Notably, the release of EVs facilitates regulatory processes across distant regions [17–19]. The demonstrated biological role of EV-derived miRNAs from macrophages/microglia in regulating distinct aspects of the onset and progression is evident in ischemic stroke and acute lung injury [20–22]. Nevertheless, there is a scarcity of data regarding the exact mechanisms that underlie this therapeutic approach. This review offers a concise overview of the influence of macrophages/microglia-derived EV-miRNAs in preclinical studies centered on the treatment of ischemic stroke and acute lung injury. The regulation of various signaling pathways in cellular processes is emphasized. Furthermore, we outline the potential clinical applications of EV-miRNAs as diagnostic and therapeutic tools for these two pathologies.

Biology of EVs and miRNAs

The biological characteristics of EVs

EVs were initially identified in plasma by Wolf in the 1960s [23]. Subsequently, various biological fluids have been examined, revealing the presence of vesicles, and in vitro cultured cell lines have demonstrated the release of vesicles to varying degrees [24]. EVs can be categorized into subgroups according to their origin, size, and physicochemical characteristics: microvesicles/endosomes (100–1000 nm), released by the plasma membrane; exosomes (30–150 nm), originating from multivesicular bodies (MVBs); and apoptotic bodies (100–5000 nm), generated through cellular apoptosis [25]. The majority of EVs originate from the direct outward budding and fission of the plasma membrane. First, the plasma membrane gives rise to endocytic vesicles (endosomes). Second, the inward budding of endosomal membranes leads to the formation of small vesicles. These small vesicles, along with various constituents, such as DNA, RNA, bioactive lipids, and proteins, are then assembled into exosomes within larger vesicles known as MVBs. Ultimately, under the regulation of the endosomal sorting complex required for transport (ESCRT) and specific proteins, EVs released into the extracellular environment are accomplished through the fusion of MVBs with the plasma membrane [26]. Upon production in the extracellular space, nanosized EVs can be internalized by recipient cells, functioning as messengers [27].

The biological characteristics of miRNAs

miRNAs are diminutive endogenous RNA molecules, typically 18–25 nucleotides in length, that exhibit a high degree of conservation [15]. Crucially involved in cell differentiation, proliferation, and survival. They achieve this by interacting with complementary target mRNAs, leading to either the inhibition of mRNA translation or its degradation [28, 29]. The biogenesis of miRNA involves a multipartite process, encompassing both nuclear and cytoplasmic phases. These molecules are generally transcribed by RNA polymerases II or III, yielding a lengthy primary miRNA (pri-miRNA) characterized by a cap and a poly-A tail [30]. The pri-miRNA then undergoes cleavage by a microprocessor complex, which consists of the RNA-binding protein DGCR8 and the type III RNase Drosha. This cleavage results in the formation of an 85-nucleotide stem-loop structure known as precursor miRNA (pre-miRNA) [31]. Afterward, RNase III DICER1 processes pre-miRNA into mature ~22-nucleotide microRNA duplexes. Following the unwinding of the duplex, one strand aligns with Ago2 (Argonaute-2) proteins, forming the RNA-induced silencing complex (RISC) [32].

miRNAs loading into EVs and subsequent release and uptake

As mentioned above, EVs encapsulate a diverse array of molecules, such as proteins, lipids, DNAs, mRNAs, and miRNAs. Among these components, miRNAs have garnered significant interest for their pivotal regulatory functions in gene expression [33]. Various mechanisms have been suggested for the selection of miRNAs as cargo in EVs. A sumoylated heterogeneous nuclear ribonucleoprotein (hnRNP), primarily comprising hnRNPA2B1, hnRNPA1, and hnRNPC, has been identified as being abundant in miRNAs within EVs. The loading of these miRNAs into MVBs seems to involve an interaction with the hnRNPA2B1 [34]. The second mechanism involves posttranscriptional modifications of miRNAs, specifically 3ʹ end uridylation, which seems to play a role in directly sorting miRNAs into EVs [35]. The third sorting pathway, AGO2, a protein associated with the RNA-induced silencing complex (RISC) responsible for RNA silencing, is believed to regulate the loading of miRNAs into EVs [36]. Following the intracellular loading of miRNAs into EVs and their binding to the cell membrane region, along with plasma membrane fusion, the ultimate release of EVs containing miRNAs occurs. Up to now, it has been suggested that cells internalize EVs either through fusion with the plasma membrane or through the process of endocytosis [37]. Endocytic uptake can be classified into various types of endocytotic processes, such as clathrin- or caveolin-mediated endocytosis, lipid raft-mediated endocytosis, macropinocytosis, and phagocytosis [38].

Macrophages/microglia activation in acute lung injury and ischemic stroke

Macrophage activation after acute lung injury

Recent research indicates that a substantial portion of tissue macrophages is sustained without reliance on blood monocytes. Instead, they originate prenatally from primitive macrophages present within the yolk sac or fetal liver [39, 40]. Macrophages in lung tissue can be classified into two main subpopulations: alveolar macrophages located in the airway lumen, characterized by surface markers CD11blow CD11c++ CD169+, and parenchymal interstitial macrophages, identified by CD11b+ CD11clow CD169. Alveolar macrophages, more prevalent than their interstitial counterparts, serve as the initial defense against foreign invaders, playing a crucial role in host defense and maintaining immune homeostasis in the local microenvironment of lung tissue [41, 42]. Macrophages are versatile cells with the capacity to transition through a spectrum of phenotypes and activation states, primarily influenced by the surrounding microenvironment. Broadly speaking, two primary macrophage phenotypes have been proposed: classically activated or pro-inflammatory (M1) macrophages and alternatively activated or anti-inflammatory (M2) macrophages [43]. In terms of phenotype, cytokines primarily released by Th1 cells, such as IFN-γ and TNF-α, along with lipopolysaccharide and granulocyte-macrophage colony-stimulating factor, are potent inducers that promote the polarization of macrophages into the M1 phenotype. In addition, M1 macrophages display heightened expression levels of CD16, CD32, CD40, CD64, CD68, CD80, CD86, MHCII, TLR4, and iNOS. They also produce increased quantities of pro-inflammatory Th1 cytokines (e.g. IL-6, IL-12, IL-1β, and TNF-α), monocyte chemotactic protein 1, macrophage inflammatory protein 2, cyclooxygenase 2, and chemokines (e.g. CCL2, CCL5, CCL8) [44, 45]. These elements play a central role in the clearance of intracellular pathogens and the recruitment and activation of T and B cells. On the contrary, M2 macrophages, also known as ‘selectively activated macrophages’, arise in response to macrophage colony-stimulating factors, immune complexes, Th2 cytokines such as IL-4 and IL-13, as well as anti-inflammatory cytokines like IL-10 and TGF-β [46, 47]. M2 macrophages primarily showcase expression of CD163, CD206, CD64, and CD209 and release anti-inflammatory cytokines, including IL-8, IL-10, and IL-13 as well as chemokines such as CCL1-4, CCL13-14, and CCL22-24 [47–49]. This expression profile contributes to their anti-inflammatory effects, support for tissue remodeling, facilitation of tumor development, and elimination of parasites. Depending on distinct microenvironments and stimuli, M2 macrophages can be categorized into four subtypes: M2a, M2b, M2c, and M2d [50]. M2a macrophages arise from non-polarized macrophages upon stimulation with IL-4 and IL-13, associating with allergic reactions [50, 51]. The M2b subset, distinguished by high expression of CD14 and CD80, can be activated by either LPS or IL-1β, resulting in the production of inflammatory cytokines [50]. M2c macrophages, induced by IL-10, TGF-β, and glucocorticoids, play a crucial role in suppressing immune responses and facilitating tissue repair [52, 53]. M2d macrophages, also known as tumor-associated macrophages, are triggered by the adenosine A2 receptor, leukemia inhibitory factor, and IL-6. Their primary function involves dampening inflammatory reactions while promoting angiogenesis and supporting tumor growth [54, 55].

Microglia activation after ischemic brain injury

Microglia represent specialized macrophages exclusive to the central nervous system. Microglia arise from primitive myeloid progenitor cells that exit the yolk sac around 8.5–9 days of embryonic development [56]. Subsequently, they migrate into the neural tube through primitive blood flow to the central nervous system. During this process, they undergo genealogy-specific gene expression changes and mature into fully differentiated microglia [57]. Under normal conditions, microglia in a resting state usually exhibit a highly branched morphology, often referred to as the resting state [56]. Serving as integral components of the innate immune system, these microglia actively survey the surrounding environment, staying vigilant for indications of damage, which can encompass damage-associated molecular patterns. Upon detecting signals of ischemia in the central nervous system microenvironment, microglia undergo morphological changes, marked by cell body hypertrophy and the retraction of cell processes. Subsequently, they promptly activate specific genes essential for initiating an inflammatory process. This inflammatory response is orchestrated with the goal of containing the progression of damage and promoting repair. The activation of microglia was traditionally classified based on intracellular dynamics and protein production into two categories: classical pro-inflammatory activation (M1) and alternative anti-inflammatory activation (M2). M1-type microglia exhibit distinctive features, producing pro-inflammatory cytokines such as IL-1β, IL-6, IL-12, IL-17, IL-18, IL-23, and TNF-α [56]. They generate redox molecules like phagocytic oxidase and iNOS, express chemokines such as CCL2 and CXCL10, and generate substantial amounts of reactive oxygen species [58, 59]. These combined factors actively contribute to the stimulation of inflammatory responses and play a role in neurotoxicity. On the flip side, M2-type microglia liberate anti-inflammatory cytokines like TGF-β, IL-4, IL-10, IL-13, and growth factors such as vascular endothelial growth factor, brain-derived neurotrophic factor, insulin-like growth factor, and platelet-derived growth factor [60, 61]. This function is geared towards mitigating inflammation and promoting tissue recovery. The M2 phenotypes are subdivided into three states: M2a, M2b, and M2c. Despite sharing common biochemical functions, these states vary regarding the stimuli that activate them, marker expression, and mechanisms of action. M2a microglia demonstrate a robust association with IL-13 and IL-4, exerting potent anti-inflammatory effects. These cells also exhibit substantial production of arginase-1, Ym-1, CD206, Fizz1, and different scavenger receptors [62, 63]. In contrast, the M2b phenotype, activated by the fusion of TLR and FCγ receptors, lacks the capacity to generate the latter [63, 64]. The M2c phenotype, also recognized as deactivated microglia, is linked to the promotion of tissue regeneration during later stages of disease progression when inflammation is diminishing [65, 66]. In animal models of ischemic stroke, the activation of microglia dynamically fluctuates over time and space during distinct stages, showing a correlation with the severity of the ischemic stroke [67]. Following a stroke, M1-type microglia are predominantly concentrated in the penumbra within the first 24 hours, and there is a noteworthy increase in the level of its marker CD68 starting from 24 hours post middle cerebral artery occlusion. Subsequently, later point, M1-type microglia can be observed in both the penumbra and the ischemic center [68]. In the early stages of stroke, the presence of M2-type microglia is confined to the ischemic core area, peaking around 3–5 days post middle cerebral artery occlusion. As time progresses, this expression diminishes, transitioning into M1-type microglia within the infarct margin area. Between days 14 and 28, the abundance of M2-type microglia has returned to its pre-injury baseline [69, 70]. In contrast, M1-type microglia continue to show a sustained increase over the 14-day period following middle cerebral artery occlusion. Moreover, Fig. 1 illustrates the polarization process between the M1 and M2 phenotypes of macrophage/microglia, highlighting the distinct signaling pathways and functional roles associated with each phenotype.

: illustrates the polarization process between the M1 and M2 phenotypes of macrophage/microglia, highlighting the distinct signaling pathways and functional roles associated with each phenotype. The polarization of macrophage/microglia into the M1 or M2 phenotype is a dynamic process influenced by the surrounding microenvironment and specific signaling molecules. Microglia that adopt the M1 phenotype are typically activated in response to proinflammatory stimuli such as interferon-gamma or lipopolysaccharide. On the other hand, macrophage/microglia that polarize towards the M2 phenotype are typically induced by anti-inflammatory signals such as interleukin-4 or interleukin-13. Created with Biorender.com
Figure 1

: illustrates the polarization process between the M1 and M2 phenotypes of macrophage/microglia, highlighting the distinct signaling pathways and functional roles associated with each phenotype. The polarization of macrophage/microglia into the M1 or M2 phenotype is a dynamic process influenced by the surrounding microenvironment and specific signaling molecules. Microglia that adopt the M1 phenotype are typically activated in response to proinflammatory stimuli such as interferon-gamma or lipopolysaccharide. On the other hand, macrophage/microglia that polarize towards the M2 phenotype are typically induced by anti-inflammatory signals such as interleukin-4 or interleukin-13. Created with Biorender.com

The potential differences between macrophage and microglia

While macrophages and microglia share many functional similarities as immune cells involved in inflammation and tissue repair, they also exhibit distinct differences in origin, function, and behavior that are important to clarify. Macrophages are derived from circulating monocytes, which originate in the bone marrow and migrate to various tissues throughout the body, where they differentiate into macrophages [71, 72]. These cells are highly adaptable and can be recruited to sites of injury or infection anywhere in the body, differentiating into various phenotypes, such as the pro-inflammatory M1 or the anti-inflammatory M2, depending on environmental signals [71–73]. This plasticity allows them to participate in a wide range of immune responses across different tissues. In contrast, microglia are resident immune cells of the central nervous system (CNS), originating from yolk sac progenitors during early development [74]. They remain in the CNS throughout life, providing critical immune surveillance and responding to CNS-specific injuries or diseases. Microglia have a more restricted range of functions compared to macrophages, being involved in maintaining homeostasis, pruning synapses, and responding to CNS-specific conditions [75, 76]. Upon activation, microglia can also polarize into M1-like or M2-like states, but their response is finely tuned to the unique environment of the CNS, including the presence of neurons and the blood-brain barrier [75]. Additionally, macrophages secrete a broad range of cytokines, chemokines, and growth factors that influence immune responses throughout the body, with their secretion profile being influenced by the tissue environment and signals they receive [77]. Microglia, while also capable of secreting cytokines and other factors, have a secretion profile specifically adapted to the CNS [77]. For example, they are involved in the production of neurotrophic factors that support neuron survival and function, a role less common in peripheral macrophages [77]. Moreover, macrophages are exposed to a wide variety of signals from different tissues and pathogens, leading to diverse responses depending on the context [78, 79]. Microglia, however, are more sensitive to subtle changes in the CNS environment, such as fluctuations in neurotransmitter levels or the presence of misfolded proteins, which is crucial for their role in CNS-specific diseases like neurodegenerative disorders [78–80]. These distinctions between macrophages and microglia reflect their specialization for roles in different parts of the body and are important to consider when evaluating their roles in diseases and therapeutic strategies, particularly in the context of EV and miRNA research, where the source and behavior of these cells can significantly influence outcomes.

The emerging role of EV-miRNAs from macrophage in acute lung injury

Critical pulmonary conditions, such as acute lung injury, present a substantial danger to critically ill patients, affecting around 200 000 individuals annually in the USA [81–83]. This condition comprises approximately 10% of all patients admitted to intensive care units and is linked to a considerable mortality rate, reaching around 40% [84, 85]. Acute lung injury manifests as a consequence of the pathophysiological response to a harmful trigger, leading to compromised pulmonary gas exchange attributed to widespread damage in the alveoli. The absence of prognostic biomarkers has impeded detection methods and the development of effective therapies for acute lung injury. Utilizing quantitative miRNA-based approaches in systemic circulation has been suggested as a way to improve diagnostic strategies and facilitate pharmacotherapy in cases of acute lung injury. It has been elucidated that several miRNAs, specifically miR-34a [86], miR-300 [87], miR-132 [88], miR-155 [89], miR-887-3p [90], miR-34a-5p [91], and miR-1246 [92], exhibit decreased levels in acute lung injury conditions. Conversely, others, such as miR-27a [93], miR-16 [94], miR-182 [95], miR-145-5p [96], and miR-140 [97], show increased expression at defined time points. The intricate balance between protective and adverse effects following acute lung injury is governed by a precisely orchestrated process regulated by various factors. These factors include but are not limited to, TLR4, FOXO3, Ang1, IκBα, FOXO3, SOCS1, SIRT1, and ACE2. Furthermore, with regard to EVs, it has been clarified that EVs originating from various stem cells, such as human adipose mesenchymal stem cells, human umbilical cord mesenchymal stem cells, and bone marrow mesenchymal stem cells, exhibit promise in the treatment of acute lung injury [98]. This potential is attributed to their favorable biological characteristics, ease of cultivation, low immunogenicity, and abundant sources.

In discussions surrounding EV-derived miRNAs from macrophages, a growing body of literature underscores their significance in regulating the prognosis of acute lung injury. A comprehensive search strategy was employed to identify relevant studies on the impact of macrophage-derived EVs, including macrophage-derived exosomes, in the treatment of acute lung injury. Databases such as PubMed, the Cochrane Library (last accessed in Nov 2023), and pertinent platforms like Web of Science and EMBASE (covering the period from 1990 to Nov 2023) were queried. The search utilized keywords such as ‘lung injury’, ‘extracellular vesicles’, or ‘exosomes’, in conjunction with ‘macrophage’ or ‘microRNA’, employing Boolean logic. This process adopted the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement as guidelines. Additionally, a manual check of the reference lists in the identified studies was conducted to identify other potential qualifying studies. This iterative process was repeated until no further publications could be obtained. In this section, a total of six publications were discovered [22, 99–102], originating from Belgium, Italy, and China, and conducted between 2020 and 2023. EVs carrying miRNAs from macrophages in different states have been identified, and these miRNAs, such as miR-1249-5p [99], miR-15a-5p22, miR-29a-3p22, miR-143-3p22, miR-122-5p22, miR-370 [100], miR-7219–3p [101], miR-155-5p [102], and miR-142-3p [103], exhibit either promotive or inhibitory effects on the prognosis of acute lung injury, as outlined in Table 1.

Table 1:

preclinical studies assessing the effects of microRNA transferred via EVs-derived from macrophage in lung injury

Author, yearmiRNAExpressionPurificationMacrophage typeFunctionTarget
Zhu et al. 2022 [99]miR-1249-5pDownregulationUltracentrifugeCell line RAW264.7Regulate influenza A virus-induced acute
lung injury in RAW246.7 cells.
SLC4A1 and NF-κB
Albano et al. 2023 [22]miR-15a-5p, miR-29a-3p, miR-143-3p, miR-122-5pDownregulation:
miR-122-5p
Upregulation:
miR-15a-5p, 29a-3p, 143-3p
UltracentrifugePBDE-47 and LPSModulate the expression of markers of epithelial integrity, epithelial mesenchymal transition, inflammation, and muco-secretion.Not report
Li et al. 2021 [100]miR-370DownregulationUltracentrifugeM2 macrophageAlleviate asthma progression in asthma-like mouse and cell models.FGF1/MAPK/STAT1
Niu et al. 2022 [101]miR-7219–3pUpregulationUltracentrifugeCell line RAW264.7Mediate fibroblast trans-differentiation in silicosis.SPRY1
Xu et al. 2023 [102]miR-155-5pUpregulationPurification kitCell line RAW264.7Widespread macrophage M1 polarization in hypervirulent Klebsiella pneumoniae-induced acute lung injuryMSK1/p38-MAPK
Guiot et al. 2020 [103]miR-142-3pUpregulationUltracentrifugeCell line RAW264.7Attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3pTGFβ-R1
Author, yearmiRNAExpressionPurificationMacrophage typeFunctionTarget
Zhu et al. 2022 [99]miR-1249-5pDownregulationUltracentrifugeCell line RAW264.7Regulate influenza A virus-induced acute
lung injury in RAW246.7 cells.
SLC4A1 and NF-κB
Albano et al. 2023 [22]miR-15a-5p, miR-29a-3p, miR-143-3p, miR-122-5pDownregulation:
miR-122-5p
Upregulation:
miR-15a-5p, 29a-3p, 143-3p
UltracentrifugePBDE-47 and LPSModulate the expression of markers of epithelial integrity, epithelial mesenchymal transition, inflammation, and muco-secretion.Not report
Li et al. 2021 [100]miR-370DownregulationUltracentrifugeM2 macrophageAlleviate asthma progression in asthma-like mouse and cell models.FGF1/MAPK/STAT1
Niu et al. 2022 [101]miR-7219–3pUpregulationUltracentrifugeCell line RAW264.7Mediate fibroblast trans-differentiation in silicosis.SPRY1
Xu et al. 2023 [102]miR-155-5pUpregulationPurification kitCell line RAW264.7Widespread macrophage M1 polarization in hypervirulent Klebsiella pneumoniae-induced acute lung injuryMSK1/p38-MAPK
Guiot et al. 2020 [103]miR-142-3pUpregulationUltracentrifugeCell line RAW264.7Attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3pTGFβ-R1

FGF1: Fibroblast Growth Factor 1; MAPK: Mitogen-Activated Protein Kinase; miR: microRNA; MSK1: Mitogen- and Stress-Activated Protein Kinase 1; NF-κB: Nuclear Factor kappa B; SLC4A1: Solute Carrier Family 4 Member 1; SPRY1: Sprouty 1; STAT1: Signal Transducer and Activator of Transcription 1; TGFβ-R1: Transforming Growth Factor Beta Receptor 1.

Table 1:

preclinical studies assessing the effects of microRNA transferred via EVs-derived from macrophage in lung injury

Author, yearmiRNAExpressionPurificationMacrophage typeFunctionTarget
Zhu et al. 2022 [99]miR-1249-5pDownregulationUltracentrifugeCell line RAW264.7Regulate influenza A virus-induced acute
lung injury in RAW246.7 cells.
SLC4A1 and NF-κB
Albano et al. 2023 [22]miR-15a-5p, miR-29a-3p, miR-143-3p, miR-122-5pDownregulation:
miR-122-5p
Upregulation:
miR-15a-5p, 29a-3p, 143-3p
UltracentrifugePBDE-47 and LPSModulate the expression of markers of epithelial integrity, epithelial mesenchymal transition, inflammation, and muco-secretion.Not report
Li et al. 2021 [100]miR-370DownregulationUltracentrifugeM2 macrophageAlleviate asthma progression in asthma-like mouse and cell models.FGF1/MAPK/STAT1
Niu et al. 2022 [101]miR-7219–3pUpregulationUltracentrifugeCell line RAW264.7Mediate fibroblast trans-differentiation in silicosis.SPRY1
Xu et al. 2023 [102]miR-155-5pUpregulationPurification kitCell line RAW264.7Widespread macrophage M1 polarization in hypervirulent Klebsiella pneumoniae-induced acute lung injuryMSK1/p38-MAPK
Guiot et al. 2020 [103]miR-142-3pUpregulationUltracentrifugeCell line RAW264.7Attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3pTGFβ-R1
Author, yearmiRNAExpressionPurificationMacrophage typeFunctionTarget
Zhu et al. 2022 [99]miR-1249-5pDownregulationUltracentrifugeCell line RAW264.7Regulate influenza A virus-induced acute
lung injury in RAW246.7 cells.
SLC4A1 and NF-κB
Albano et al. 2023 [22]miR-15a-5p, miR-29a-3p, miR-143-3p, miR-122-5pDownregulation:
miR-122-5p
Upregulation:
miR-15a-5p, 29a-3p, 143-3p
UltracentrifugePBDE-47 and LPSModulate the expression of markers of epithelial integrity, epithelial mesenchymal transition, inflammation, and muco-secretion.Not report
Li et al. 2021 [100]miR-370DownregulationUltracentrifugeM2 macrophageAlleviate asthma progression in asthma-like mouse and cell models.FGF1/MAPK/STAT1
Niu et al. 2022 [101]miR-7219–3pUpregulationUltracentrifugeCell line RAW264.7Mediate fibroblast trans-differentiation in silicosis.SPRY1
Xu et al. 2023 [102]miR-155-5pUpregulationPurification kitCell line RAW264.7Widespread macrophage M1 polarization in hypervirulent Klebsiella pneumoniae-induced acute lung injuryMSK1/p38-MAPK
Guiot et al. 2020 [103]miR-142-3pUpregulationUltracentrifugeCell line RAW264.7Attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3pTGFβ-R1

FGF1: Fibroblast Growth Factor 1; MAPK: Mitogen-Activated Protein Kinase; miR: microRNA; MSK1: Mitogen- and Stress-Activated Protein Kinase 1; NF-κB: Nuclear Factor kappa B; SLC4A1: Solute Carrier Family 4 Member 1; SPRY1: Sprouty 1; STAT1: Signal Transducer and Activator of Transcription 1; TGFβ-R1: Transforming Growth Factor Beta Receptor 1.

In a lung injury associated with an asthma mouse model induced by ovalbumin, EVs derived from M2 macrophages markedly mitigated ovalbumin-induced fibrosis and inflammatory responses in the lung tissues of mice [100]. Additionally, they inhibited abnormal proliferation, invasion, and the production of fibrosis-related proteins in primary mouse airway smooth muscle cells treated with platelet-derived growth factor [100]. The administration of ovalbumin in mice or the treatment of airway smooth muscle cells with platelet-derived growth factor-BB led to a decrease in the expression of miR-370. Interestingly, the introduction of M2 macrophage-derived EVs reversed the inhibitory effect on miR-370 expression in both vivo and vitro. Conversely, the downregulation of miR-370 by Lv-miR-370 inhibitor counteracted the protective effects of M2 macrophage-derived EVs in asthma-like mouse and cell models. Collectively, miR-370 was involved in the therapeutic effects of M2 macrophage-derived EVs by alleviating asthma progression [100]. In the context of lung injury associated with silicosis, Niu et al. [101] demonstrated that exposure to silica stimulates macrophages to release an increased quantity of EVs. These vesicles play a crucial role in fibroblast-to-myofibroblast transition, and this impact is, at least in part, mediated by miR-7219-3p carried within EVs. The authors additionally observed that these effects were attained by inhibiting SPRY1 and activating ERK/MAPK phosphorylation. Herein, this phenomenon could be reversed after treatment with a miR-7219-3p inhibitor and antagomir. In a lung injury associated with fibrosis, Guiot et al. [103] showed that EVs derived from macrophages alleviate fibrosis in airway epithelial cells by delivering the antifibrotic miR-142-3p. miRNAs within EVs originating from various cells can also modulate macrophage polarization and contribute to the advancement of lung injury. For example, in a lung injury induced by influenza A virus, Zhu et al. [99] indicated that miR-1249-5p derived from mouse lung epithelial cell-EVs modulates acute lung injury in macrophages by targeting SLC4A1. In addition, macrophages under stimulation can autonomously influence their polarization by releasing EV-derived miRNAs. As indicated by Xu et al. [102], they revealed that EVs miR-155-5p originating from macrophages activated by hypervirulent Klebsiella pneumoniae can impact the M1 polarization and inflammatory response of quiescent macrophages. This occurs by activating the p38-MAPK signaling pathway, thereby influencing lung inflammation and tissue damage.

The emerging role of EV-miRNAs derived from microglia in ischemic stroke

Numerous investigators exploring EV-miRNAs from macrophages have unveiled a fresh perspective on the interplay in acute lung injury, as previously mentioned. Additionally, the pivotal involvement of miRNAs encapsulated in EVs in ischemic stroke has been acknowledged. In the initial phase, a specific group of miRNAs, including miR-22, miR-26a, miR-124, miR-146a, miR-181c, miR-183, miR-203, miR-210, and miR-424, exhibit an elevation following ischemic stroke [104]. Conversely, others such as miR-130a, miR-150, miR-155, miR-182, and miR-320 are diminished during this early stage [104]. Meanwhile, EVs possess the capability to be internalized by adjacent or distant cells due to their diverse targets. These EVs modulate gene expression through posttranscriptional regulation, with the miRNAs they carry serving as potent factors for repair. A growing body of evidence indicates that miRNAs are preferentially concentrated in EVs, exerting biological functions by modulating specific facets of ischemic stroke. Consequently, they play a role in cell death, apoptosis, and neuroinflammation [20, 21, 105–111], as outlined in Table 2 and depicted in Figure 2. These aspects are further explored in the subsequent sections.

Table 2:

preclinical studies assessing the effects of microRNA transferred via EVs-derived from microglia in stroke

Author, yearmiRNAExpressionPurificationMicroglial statusFunctionTarget
Li et al. 2021 [20]miR-124UpregulationUltracentrifugationM2-BV2Reduce glial scar formation and promote ischemic stroke recovery.STAT3
Li et al. 2022 [21]miR-23a-5pUpregulationUltracentrifugationM2-BV2Promote white matter repair and functional ischemic stroke recovery.Olig3
Li et al. 2023 [105]miR-212-5pUpregulationUltracentrifugationMicrogliaPromote synaptic plasticity and attenuate axonal degeneration.PlexinA2
Song et al. 2019 [106]miR-124UpregulationUltracentrifugationM2-BV2Attenuate ischemic brain injury and promote neuronal survival.USP14
Song et al. 2023 [107]miR-124UpregulationUltracentrifugationM2-BV2Enhance neural stem cell proliferation and differentiation.AAK1/Notch
Wang et al. 2023 [108]miR-9-5pDownregulationUltracentrifugationBV2Attenuate LPS-induced microglia cell inflammation.NO, IL-6, TNF-α
Wei et al. 2021 [109]miR-383-3pUpregulationUltracentrifugationM2 microgliaPromote necroptosis of neurons in intracerebral hemorrhage rats.ATF4
Xie et al. 2020 [110]miR‑424-5pUpregulationUltracentrifugationOGD-treated microgliaAggravate BBB destruction and neurological damage.FGF2/STAT3
Zhang et al. 2022 [111]miR-137UpregulationUltracentrifugationM2-BV2Alleviate neuronal apoptosis and ischemia-reperfusion brain injury.Notch1
Author, yearmiRNAExpressionPurificationMicroglial statusFunctionTarget
Li et al. 2021 [20]miR-124UpregulationUltracentrifugationM2-BV2Reduce glial scar formation and promote ischemic stroke recovery.STAT3
Li et al. 2022 [21]miR-23a-5pUpregulationUltracentrifugationM2-BV2Promote white matter repair and functional ischemic stroke recovery.Olig3
Li et al. 2023 [105]miR-212-5pUpregulationUltracentrifugationMicrogliaPromote synaptic plasticity and attenuate axonal degeneration.PlexinA2
Song et al. 2019 [106]miR-124UpregulationUltracentrifugationM2-BV2Attenuate ischemic brain injury and promote neuronal survival.USP14
Song et al. 2023 [107]miR-124UpregulationUltracentrifugationM2-BV2Enhance neural stem cell proliferation and differentiation.AAK1/Notch
Wang et al. 2023 [108]miR-9-5pDownregulationUltracentrifugationBV2Attenuate LPS-induced microglia cell inflammation.NO, IL-6, TNF-α
Wei et al. 2021 [109]miR-383-3pUpregulationUltracentrifugationM2 microgliaPromote necroptosis of neurons in intracerebral hemorrhage rats.ATF4
Xie et al. 2020 [110]miR‑424-5pUpregulationUltracentrifugationOGD-treated microgliaAggravate BBB destruction and neurological damage.FGF2/STAT3
Zhang et al. 2022 [111]miR-137UpregulationUltracentrifugationM2-BV2Alleviate neuronal apoptosis and ischemia-reperfusion brain injury.Notch1

AAK1: adaptor-associated protein kinase 1; BBB: blood-brain barrier; miR: microRNAs; OGD: oxygen-glucose deprivation; USP14: ubiquitin-specific protease 14.

Table 2:

preclinical studies assessing the effects of microRNA transferred via EVs-derived from microglia in stroke

Author, yearmiRNAExpressionPurificationMicroglial statusFunctionTarget
Li et al. 2021 [20]miR-124UpregulationUltracentrifugationM2-BV2Reduce glial scar formation and promote ischemic stroke recovery.STAT3
Li et al. 2022 [21]miR-23a-5pUpregulationUltracentrifugationM2-BV2Promote white matter repair and functional ischemic stroke recovery.Olig3
Li et al. 2023 [105]miR-212-5pUpregulationUltracentrifugationMicrogliaPromote synaptic plasticity and attenuate axonal degeneration.PlexinA2
Song et al. 2019 [106]miR-124UpregulationUltracentrifugationM2-BV2Attenuate ischemic brain injury and promote neuronal survival.USP14
Song et al. 2023 [107]miR-124UpregulationUltracentrifugationM2-BV2Enhance neural stem cell proliferation and differentiation.AAK1/Notch
Wang et al. 2023 [108]miR-9-5pDownregulationUltracentrifugationBV2Attenuate LPS-induced microglia cell inflammation.NO, IL-6, TNF-α
Wei et al. 2021 [109]miR-383-3pUpregulationUltracentrifugationM2 microgliaPromote necroptosis of neurons in intracerebral hemorrhage rats.ATF4
Xie et al. 2020 [110]miR‑424-5pUpregulationUltracentrifugationOGD-treated microgliaAggravate BBB destruction and neurological damage.FGF2/STAT3
Zhang et al. 2022 [111]miR-137UpregulationUltracentrifugationM2-BV2Alleviate neuronal apoptosis and ischemia-reperfusion brain injury.Notch1
Author, yearmiRNAExpressionPurificationMicroglial statusFunctionTarget
Li et al. 2021 [20]miR-124UpregulationUltracentrifugationM2-BV2Reduce glial scar formation and promote ischemic stroke recovery.STAT3
Li et al. 2022 [21]miR-23a-5pUpregulationUltracentrifugationM2-BV2Promote white matter repair and functional ischemic stroke recovery.Olig3
Li et al. 2023 [105]miR-212-5pUpregulationUltracentrifugationMicrogliaPromote synaptic plasticity and attenuate axonal degeneration.PlexinA2
Song et al. 2019 [106]miR-124UpregulationUltracentrifugationM2-BV2Attenuate ischemic brain injury and promote neuronal survival.USP14
Song et al. 2023 [107]miR-124UpregulationUltracentrifugationM2-BV2Enhance neural stem cell proliferation and differentiation.AAK1/Notch
Wang et al. 2023 [108]miR-9-5pDownregulationUltracentrifugationBV2Attenuate LPS-induced microglia cell inflammation.NO, IL-6, TNF-α
Wei et al. 2021 [109]miR-383-3pUpregulationUltracentrifugationM2 microgliaPromote necroptosis of neurons in intracerebral hemorrhage rats.ATF4
Xie et al. 2020 [110]miR‑424-5pUpregulationUltracentrifugationOGD-treated microgliaAggravate BBB destruction and neurological damage.FGF2/STAT3
Zhang et al. 2022 [111]miR-137UpregulationUltracentrifugationM2-BV2Alleviate neuronal apoptosis and ischemia-reperfusion brain injury.Notch1

AAK1: adaptor-associated protein kinase 1; BBB: blood-brain barrier; miR: microRNAs; OGD: oxygen-glucose deprivation; USP14: ubiquitin-specific protease 14.

: examining the impact of microRNA transferred through extracellular vesicles released by microglia in ischemic stroke reveals a broad range of effects. Originating predominantly from microglial cells in the extracellular space, these vesicles primarily influence processes such as cell differentiation, autophagy, apoptosis, necroptosis, regeneration, and inflammation through diverse pathways. These vesicles can interact with multiple cell types, including neurons, astrocytes, stem cells, and oligodendrocytes. MicroRNAs assume crucial roles in various pathways within this context. Created with Biorender.com
Figure 2

: examining the impact of microRNA transferred through extracellular vesicles released by microglia in ischemic stroke reveals a broad range of effects. Originating predominantly from microglial cells in the extracellular space, these vesicles primarily influence processes such as cell differentiation, autophagy, apoptosis, necroptosis, regeneration, and inflammation through diverse pathways. These vesicles can interact with multiple cell types, including neurons, astrocytes, stem cells, and oligodendrocytes. MicroRNAs assume crucial roles in various pathways within this context. Created with Biorender.com

The occurrence of cell death in neurological diseases results from a variety of cellular processes, encompassing mitochondrial dysfunction, protein aggregation, the generation of free radicals, excitotoxicity, and inflammation [112]. Numerous studies have revealed that EV-miRNAs derived from microglia are involved in cell death regulation. For example, as indicated by Zhang et al. [111], in the LDH assay, it was observed that a microglia-conditioned medium led to a decrease in LDH activity, signifying reduced cellular injury in neurons following OGD treatment. Interestingly, Zhang et al. [111] noted that the introduction of GW4869, an inhibitor of EV secretion, partially reversed the impact of the microglia-conditioned medium on OGD-treated neurons. These findings collectively suggest the potential involvement of microglia-derived EVs in a neuroprotective mechanism against OGD. Furthermore, LDH assays revealed that neurons, when cocultured with microglia-derived EVs for 24 hours followed by OGD, displayed a significant enhancement in cell viability. Importantly, it was observed that miR-137 was associated with the decreased infarct volume and behavioral deficits in ischemic mice mediated by microglia-derived EVs. As such, Li et al. [105] indicated that focal ischemic stroke alters EV-miR-212-5p derived from microglia, with a potential implication of miR-212-5p in promoting neuronal survival and contributing to functional recovery. Furthermore, the miRNAs in these EVs can also promote the survival rates of oligodendrocytes. As revealed by Li et al. [21], M2-type microglia-EVs were found to enhance oligodendrogenesis in a model of MCAO, promoting the proliferation, survival, and differentiation of oligodendrocyte precursor cells in vitro OGD model. MiR-23a-5p was identified as an enriched component in these EVs, exerting a positive influence on the proliferation, survival, and maturation of oligodendrocyte precursor cells. Knocking down miR-23a-5p in these EVs reversed the beneficial effects observed both in vitro and in vivo. To facilitate the restoration of neurological function following a stroke, the miRNAs contained within these EVs can additionally impede the proliferation of specific cells, such as astrocytes [106]. EVs derived from M2 microglia attenuated the formation of glial scars and facilitated post-stroke recovery. These vesicles were notably enriched in miR-124. Additionally, treatment with M2 microglial EVs resulted in reduced expression of the astrocyte proliferation gene Signal Transducer and Activator of Transcription 3, a target of miR-124. This treatment also led to decreased levels of glial fibrillary acidic protein, inhibiting astrocyte proliferation both in vitro and in vivo. Knocking down miR-124 in such EVs hindered their impact on glial scar formation and impaired the recovery process after a stroke [106]. It is worth noting that not all the miRNAs in these EVs are neuroprotective by regulating cell survival. Xie et al. [110] showed that EVs derived from OGD-activated microglia exacerbated OGD-induced damage to brain microvascular endothelial cells, including reduced viability and integrity, as well as loss of vascular formation. However, these detrimental effects were significantly mitigated by inhibiting miR-424-5p. Furthermore, the overexpression of miR-424-5p notably intensified OGD-induced damage and increased permeability in brain microvascular endothelial cells.

Among the diverse pathways leading to programmed cell death [113, 114], apoptosis constitutes a significant portion of cell demise associated with brain injury [114]. This process efficiently eliminates damaged cells resulting from DNA damage or during developmental stages [115]. Apoptosis is crucial for maintaining the homeostasis of normal tissues, and researchers have identified that EV-miRNAs from microglia play essential roles in the regulation of cellular apoptosis. As indicated by Zhang et al. [111] utilizing the TUNEL assay to identify apoptotic changes in neurons subjected to control conditions, OGD, OGD with M2-phenotype microglia EVs, mitigation of neuronal apoptosis induced by OGD was observed. Notably, RNA-seq analysis revealed an upregulation of miR-137 in EVs derived from M2-phenotype microglia. To elucidate the role of miR-137, it was found that the expression of cleaved caspase-3 significantly decreased in neurons treated with such EVs following OGD. This effect was partially reversed when neurons were treated with EVs derived from microglia subjected to a miR-137 inhibitor. Additionally, TUNEL assay results demonstrated that EVs with reduced miR-137 expression partially counteracted the pro-apoptotic impact of these EVs after OGD. In addition to apoptosis, the regulation of EV-miRNAs from microglia on necroptosis has been reported [109]. Programmed cell death takes on different characteristics in the forms of apoptosis and necroptosis. Necroptosis is orchestrated by the receptor-interacting proteins RIP1/RIP3 and the substrate of RIP3/MLKL [116]. These proteins collaborate to form a necrosis signaling complex, actively participating in the process of necroptosis [116]. Wei et al. demonstrated that the EVs derived from activated microglia play a mediating role in promoting neuronal necroptosis through the inhibition of ATF4 expression via miR-383-3p in stroke [109]. This also indicates that EV-miRNAs in microglial cells are not all protective.

The inflammatory response presents a dual role following ischemia, acting as a double-edged sword by exacerbating secondary brain injury while also fostering the recovery of neurological function. This dual effect highlights the association between inflammation, pathogenesis, and the prognosis of ischemia [117]. Numerous studies have demonstrated the close association of EV-miRNAs from microglial cells with the regulation of inflammation in the context of ischemia. The research revealed that Xiaoxuming decoction cutting possesses anti-neuroinflammatory properties [108]. Small RNA-seq analysis of EVs identified miR-9-5p as a crucial miRNA in Xiaoxuming decoction cutting’s mechanism for addressing neuroinflammation. Inhibition of miR-9-5p was observed to decrease the expression of inflammatory factors, including IL-1β, IL-6, iNOS, and TNF-α. Consequently, Xiaoxuming decoction cutting modulates the inflammatory response by influencing the expression of miR-9-5p in EVs derived from microglia [108].

EV-miRNAs: emerging candidates as novel biomarkers and therapeutic targets

EV-mediated miRNAs as potential biomarkers

EVs serve as mediators of cell-to-cell communication, facilitating the transport and delivery of their cargo to target cells. The contents of these vesicles serve as a molecular bioprinter, reflecting the characteristics of their parent cells. Consequently, EVs found in blood and other biofluids hold promise as potential noninvasive biomarkers for the early diagnosis and prognosis of various diseases. EVs, acting as natural carriers of miRNAs, possess several noteworthy attributes. Effectively harnessing the delivery of these vesicles poses a considerable challenge, given the need to attain heightened stability, even at room temperature over extended periods, while simultaneously minimizing toxicity and mitigating off-target effects [118]. Furthermore, EVs are plentiful in various body fluids such as blood, urine, and cerebrospinal fluids, which not only enables straightforward isolation but also bestows non-invasive advantages. The changes in expression of numerous miRNAs subsequent to ischemic stroke or acute lung injury, proposed as potential biomarkers, emphasize the possibility of manipulating EV-derived miRNAs to elucidate the progression of both injury conditions. Under ischemic stroke conditions, it is already known that miR-15, miR-16, miR-17-5p, miR-30a, and miR-126 from plasma, serum, or peripheral leukocytes were significantly downregulated in ischemic stroke patients [119], and the upregulated miR-16, miR-15, miR-17-5p, miR-29b, and miR-132 derived from the plasma of ischemic stroke patients also served as diagnostic biomarkers [119]. Interestingly, EVs as delivery carriers for miRNAs have also been identified as biomarkers for diagnosis. miR-134, miR-21-5p, miR-30a-5p, miR-223, miR-9, and miR-124 related to EVs and originating from the plasma or serum of stroke patients show an upregulated expression. Conversely, miR-422a and miR-125b-2-3p exhibit downregulated expression in the same context [120–124]. Under acute lung injury, in a study by Parzibut et al. [125], the expression of plasma EV-miRNAs was compared between 8 patients with acute respiratory distress syndrome and 10 healthy subjects using small RNA sequencing analysis. Notably, among the differentially expressed miRNAs, namely miR-221-3p, miR-24-3p, miR-130a-3p, Let-7d-3p, miR-1273a, miR-98-3p, and miR-193a-5p, were demonstrated to effectively distinguish between acute respiratory distress syndrome and hemorrhagic shock through receiver operating characteristic curve analysis. Elevated levels of miR-146a, miR-126, miR-27a, and miR-155 were identified in serum EVs from individuals with severe community-acquired pneumonia-related acute lung injury compared to those in the control group [126]. Despite traditional markers being widely used for clinical diagnosis, low sensitivity, and specificity limitations exist. A combination of conventional markers with EV-miRNAs is illustrated to boost diagnostic accuracy [13].

Utilizing EVs as carriers for miRNA-based therapy

EV-miRNAs also emerge as promising targets for therapeutic interventions in the management of conditions such as ischemic stroke and acute lung injury. EVs serve as protective carriers for miRNA cargo, shielding them from endogenous RNases and facilitating their delivery to target cells [127]. Generally, miRNA-based therapy aims to achieve therapeutic objectives by either replenishing or inhibiting the sequences of target miRNAs. The essential stages of miRNA-based therapy encompass identifying optimal miRNA targets, deciphering miRNA chemistry, refining delivery methods, conducting preclinical testing, and progressing to clinical trials. Under ischemic stroke conditions, in the work of Cai et al. [128], the expression of miR-542-3p was upregulated in EVs derived from mesenchymal stem cells through the transfection of miR-542-3p mimics. Administration of EV-miR-542-3p increased the expression of miR-542-3p and provided protection against ischemia-induced damage in mice subjected to middle cerebral artery occlusion and HA1800 cells exposed to hypoxia. This protective effect was achieved through the reduction of inflammation. Under acute lung injury conditions, the primary approach to treating acute lung injury continues to be symptomatic, involving interventions such as mechanical ventilation, fluid management, corticosteroid supplementation, and the use of inhaled pulmonary vasodilators. While these treatments can alleviate patient symptoms, they do not address the root cause of the condition. There is evidence indicating that miRNAs derived from EVs of mesenchymal stem cells can engage in intercellular interactions with resident lung cells, suggesting potential therapeutic benefits for acute lung injury [129]. EV-associated miR-384-5p, originating from bone marrow-derived mesenchymal stem cells, has demonstrated the capacity to mitigate macrophage apoptosis and autophagy stress in rats with acute lung injury [130]. This effect is achieved by targeting Beclin-1 in alveolar macrophages, ultimately contributing to improved survival. Additionally, in another study, EV-miR-127-5p derived from bone marrow-derived mesenchymal stem cells has been observed to impede the formation of neutrophil extracellular traps in acute lung injury associated with sepsis by targeting CD64. More importantly, EVs commonly contain CD47, an integrin-associated transmembrane protein that acts as a ‘don’t eat me’ signal, offering protection against phagocytosis [131–134]. Research findings indicate that EVs with an overexpression of CD47 demonstrate an extended half-life in circulation when compared to EVs lacking CD47. This observation suggests that the presence of CD47 on EVs contributes to their increased stability and prolonged presence in the bloodstream.

Challenges and potential risks of using EVs in clinical settings

The article presents a compelling case for the use of EVs as diagnostic and therapeutic tools, particularly in the context of neuroinflammatory conditions. However, translating these promising findings into clinical practice involves several significant challenges and limitations that require careful consideration. One of the primary clinical challenges lies in the standardization and isolation techniques for EVs [135, 136]. The inherent heterogeneity of EVs, combined with the lack of universally accepted protocols for their isolation and characterization, complicates the ability to achieve consistent and reproducible results [135, 136]. This variability can significantly impact the purity, yield, and functionality of EV preparations, making it difficult to ensure consistency in clinical applications where it is paramount. Additionally, the scalability and manufacturing of EVs for clinical use present another substantial hurdle. Producing EVs in sufficient quantities under good manufacturing practice conditions is not only technically demanding but also costly, potentially limiting the accessibility, and widespread adoption of EV-based therapies [12]. The safety profile of EVs also remains an area of concern [12]. Although EVs hold promise as therapeutic agents, they can carry a variety of bioactive molecules, including miRNAs, proteins, and lipids, some of which could inadvertently induce off-target effects, such as promoting oncogenesis or unwanted immune responses [12]. Moreover, the long-term effects of EV administration are not well understood, raising concerns about potential adverse outcomes over time. Regulatory and ethical concerns further complicate the clinical translation of EVs. The regulatory landscape for EV-based therapies is complex and still evolving, with stringent requirements for demonstrating safety, efficacy, and consistency [137]. Navigating these regulatory hurdles could be time-consuming and may delay the clinical implementation of these technologies. Ethical issues also arise, particularly regarding the source of EVs, especially if they are derived from human stem cells or other tissues [137]. Ensuring ethical compliance in terms of donor consent, potential exploitation, and the use of human-derived materials is crucial for the responsible development of EV-based therapies. Furthermore, there are limitations in current research that must be addressed. Much of the existing knowledge about EVs and their therapeutic potential is derived from in vitro studies or animal models [16]. Translating these findings to human patients presents challenges due to differences in physiology and the complexity of human diseases, and the efficacy and safety observed in preclinical studies may not always translate directly to clinical success [16]. Additionally, variability in EV isolation techniques, experimental conditions, and even patient populations can lead to inconsistent results, making it difficult to draw definitive conclusions about their therapeutic potential [12, 16]. This variability underscores the need for standardized methods and large-scale clinical trials to validate the efficacy of EV-based therapies. In summary, while the potential of EVs as diagnostic and therapeutic tools is promising, there are significant clinical challenges and limitations that need to be addressed before these technologies can be widely adopted in clinical practice. Further research, standardized protocols, and rigorous clinical trials will be essential to overcome these challenges and fully realize the therapeutic potential of EVs.

Conclusion

EVs play a pivotal role in the progression of ischemic stroke and acute lung injury, with their cargo holding significant importance. Enclosed miRNAs within EVs contribute to biological functions by influencing various aspects thereby decisively impacting tissue progression. Nevertheless, there are substantial challenges to address before EV-associated miRNAs can be effectively utilized as clinical assays for such disorders. Further exploration is essential to devise techniques for generating pure and homogeneous EVs. Additionally, addressing variations in EVs secreted by diverse cells under different environmental or stimulatory conditions, and developing methods to prepare stable decoy EVs for disease treatment, are additional challenges that require careful consideration. Significant hurdles persist in the clinical translation of EVs, particularly regarding the large-scale isolation and preparation of EVs. Ensuring the reproducibility of EV preparations under specific culturing conditions is also a substantial challenge. Additionally, the development of potency assays is essential to guarantee that the activity characteristics of EVs remain consistent across different preparations before their administration to human patients [138, 139]. The nuclear RNA exosome complex stands out as one of the most versatile RNA-degradation mechanisms in eukaryotes [140]. Hence, it is highly valuable to showcase how the regulation of the nuclear RNA exosome complex can be influenced by modulating the levels of exosomal miRNAs [16]. The functionality of EV-miRNAs is predominantly studied through animal and cellular models; however, additional validation in clinical samples is imperative.

Abbreviations

    Abbreviations
     
  • Ago2

    argonaute-2

  •  
  • circRNAs

    circular RNAs

  •  
  • CNS

    central nervous system

  •  
  • ESCRT

    endosomal sorting complex required for transport

  •  
  • EVs

    extracellular vesicles

  •  
  • lncRNAs

    long noncoding RNAs

  •  
  • MCAO

    middle cerebral artery occlusion

  •  
  • miRNAs

    microRNAs

  •  
  • MVBs

    multivesicular bodies

  •  
  • ncRNAs

    non-coding RNAs

  •  
  • OGD

    oxygen-glucose deprivation

  •  
  • pre-miRNA

    precursor miRNA

  •  
  • pri-miRNA

    primary miRNA

  •  
  • PRISMA

    preferred reporting items for systematic reviews and meta-analyses

  •  
  • RISC

    RNA-induced silencing complex

Conflict of interest

None declared.

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

Author contributions

Xianbin Wang (Conceptualization, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Validation, Visualization, Writing—original draft, Writing—review & editing), Ting Wang (Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Writing—original draft, Writing—review & editing), Dong Zhu (Data curation, Formal analysis, Investigation, Software, Validation, Visualization, Writing—original draft), Jing Wang (Data curation, Funding acquisition, Methodology, Project administration, Software, Validation, Writing—original draft), and Weijie Han (Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing—original draft, Writing—review & editing)

References

1.

Bindal
P
,
Kumar
V
,
Kapil
L
,
Singh
C
,
Singh
A.
Therapeutic management of ischemic stroke
.
Naunyn-Schmiedeberg’s Arch Pharmacol
2023
,
397
,
2651
2679
. doi: https://doi.org/

2.

Deng
X
,
Hu
Z
,
Zhou
S
,
Wu
Y
,
Fu
M
,
Zhou
C
, et al.
Perspective from single-cell sequencing: Is inflammation in acute ischemic stroke beneficial or detrimental
?
CNS Neurosci Ther
2024
30
e14510
. doi:

3.

Wang
Z
,
Wang
Z.
The role of macrophages polarization in sepsis-induced acute lung injury
.
Front Immunol
2023
,
14
,
1209438
. doi: https://doi.org/

4.

Swenson
KE
,
Swenson
ER.
Pathophysiology of acute respiratory distress syndrome and COVID-19 lung injury
.
Crit Care Clin
2021
,
37
,
749
776
. doi: https://doi.org/

5.

He
YQ
,
Zhou
CC
,
Yu
LY
,
Wang
L
,
Deng
JL
,
Tao
YL
, et al.
Natural product derived phytochemicals in managing acute lung injury by multiple mechanisms
.
Pharmacol Res
2021
,
163
,
105224
. doi: https://doi.org/

6.

Li
R
,
Tao
C
,
Sun
J
,
Zhang
C
,
Xu
P
,
Yin
Y
, et al.
Endovascular vs medical management of acute basilar artery occlusion: a secondary analysis of a randomized clinical trial
.
JAMA Neurol
2024
,
81
1043
1050
. doi:  https://doi.org/

7.

Tong
C
,
Xu
L.
Endovascular treatment for large ischemic core stroke: challenges and opportunities
.
Int J Surg (London, England)
2024
. doi:  https://doi.org/

8.

Shekhar
S
,
Liu
Y
,
Wang
S
,
Zhang
H
,
Fang
X
,
Zhang
J
, et al.
Novel mechanistic insights and potential therapeutic impact of trpc6 in neurovascular coupling and ischemic stroke
.
Int J Mol Sci
2021
,
22
2074
. doi:  https://doi.org/

9.

Wei
YH
,
Bi
RT
,
Qiu
YM
,
Zhang
CL
,
Li
JZ
,
Li
YN
, et al.
The gastrointestinal–brain–microbiota axis: a promising therapeutic target for ischemic stroke
.
Front Immunol
2023
,
14
,
1141387
. doi: https://doi.org/

10.

Liu
Z
,
Tang
Y
,
Zhang
Z
,
Liu
Q
,
Wang
M
,
Li
W
,
Yang
GY.
Engineering neurovascular unit and blood-brain barrier for ischemic stroke modeling
.
Adv Healthcare Mater
2023
,
12
,
e2202638
. doi: https://doi.org/

11.

Chen
Z
,
Zhang
J
,
Pan
Y
,
Hao
Z
,
Li
S.
Extracellular vesicles as carriers for noncoding RNA-based regulation of macrophage/microglia polarization: an emerging candidate regulator for lung and traumatic brain injuries
.
Front Immunol
2024
,
15
,
1343364
. doi: https://doi.org/

12.

Hermann
DM
,
Xin
W
,
Bähr
M
,
Giebel
B
,
Doeppner
TR.
Emerging roles of extracellular vesicle-associated non-coding RNAs in hypoxia: Insights from cancer, myocardial infarction and ischemic stroke
.
Theranostics
2022
,
12
,
5776
5802
. doi: https://doi.org/

13.

Pan
Y
,
Liu
Y
,
Wei
W
,
Yang
X
,
Wang
Z
,
Xin
W.
Extracellular vesicles as delivery shippers for noncoding rna-based modulation of angiogenesis: insights from ischemic stroke and cancer
.
Small (Weinheim-Bergstrasse, Germany)
2023
,
19
,
e2205739
. doi: https://doi.org/

14.

Dutta
S
,
Zhu
Y
,
Han
Y
,
Almuntashiri
S
,
Wang
X
,
Zhang
D.
Long Noncoding RNA: a novel insight into the pathogenesis of acute lung injury
.
J Clin Med
2023
,
12
,
604
doi: https://doi.org/

15.

Ni
L
,
Tang
C
,
Wang
Y
,
Wan
J
,
Charles
MG
,
Zhang
Z
, et al.
Construction of a miRNA-based nomogram model to predict the prognosis of endometrial cancer
.
J Pers Med
2022
,
12
,
1154
. doi: https://doi.org/

16.

Xin
W
,
Qin
Y
,
Lei
P
,
Zhang
J
,
Yang
X
,
Wang
Z.
From cerebral ischemia towards myocardial, renal, and hepatic ischemia: exosomal miRNAs as a general concept of intercellular communication in ischemia-reperfusion injury
.
Mol Ther Nucleic Acids
2022
,
29
,
900
922
. doi: https://doi.org/

17.

Lai
CP
,
Breakefield
XO.
Role of exosomes/microvesicles in the nervous system and use in emerging therapies
.
Front Physiol
2012
,
3
,
228
. doi: https://doi.org/

18.

Rajendran
L
,
Bali
J
,
Barr
MM
,
Court
FA
,
Krämer-Albers
EM
,
Picou
F
, et al.
Emerging roles of extracellular vesicles in the nervous system
.
J Neuros
2014
,
34
,
15482
9
. doi: https://doi.org/

19.

Krämer-Albers
EM
,
Hill
AF.
Extracellular vesicles: interneural shuttles of complex messages
.
Curr Opin Neurobiol
2016
,
39
,
101
7
. doi: https://doi.org/

20.

Li
Z
,
Song
Y
,
He
T
,
Wen
R
,
Li
Y
,
Chen
T
, et al.
M2 microglial small extracellular vesicles reduce glial scar formation via the miR-124/STAT3 pathway after ischemic stroke in mice
.
Theranostics
2021
,
11
,
1232
1248
. doi: https://doi.org/

21.

Li
Y
,
Liu
Z
,
Song
Y
,
Pan
JJ
,
Jiang
Y
,
Shi
X
, et al.
M2 microglia-derived extracellular vesicles promote white matter repair and functional recovery via miR-23a-5p after cerebral ischemia in mice
.
Theranostics
2022
,
12
,
3553
3573
. doi: https://doi.org/

22.

Albano
GD
,
Longo
V
,
Montalbano
AM
,
Aloi
N
,
Barone
R
, et al.
Extracellular vesicles from PBDE-47 treated M(LPS) THP-1 macrophages modulate the expression of markers of epithelial integrity, EMT, inflammation and muco-secretion in ALI culture of airway epithelium
.
Life Sci
2023
,
322
,
121616
. doi: https://doi.org/

23.

Wolf
P.
The nature and significance of platelet products in human plasma
.
Br J Haematol
1967
,
13
,
269
88
. doi: https://doi.org/

24.

Hessvik
NP
,
Llorente
A.
Current knowledge on exosome biogenesis and release
.
CMLS
2018
,
75
,
193
208
. doi: https://doi.org/

25.

Sorop
A
,
Constantinescu
D
,
Cojocaru
F
,
Dinischiotu
A
,
Cucu
D
,
Dima
SO.
Exosomal microRNAs as biomarkers and therapeutic targets for hepatocellular carcinoma
.
Int J Mol Sci
2021
,
22
,
4997
. doi: https://doi.org/

26.

Wang
X
,
Zhou
Y
,
Gao
Q
,
Ping
D
,
Wang
Y
,
Wu
W
, et al.
The role of exosomal microRNAs and oxidative stress in neurodegenerative diseases
.
Oxid Med Cell Longevity
2020
,
2020
,
1
17
doi: https://doi.org/

27.

Di Bella
MA.
Overview and update on extracellular vesicles: considerations on exosomes and their application in modern medicine
.
Biology
2022
,
11
,
804
doi: https://doi.org/

28.

Lu
TX
,
Rothenberg
ME.
MicroRNA
.
J Allergy Clin Immunol
2018
,
141
,
1202
1207
. doi: https://doi.org/

29.

Rupaimoole
R
,
Slack
FJ.
MicroRNA therapeutics: towards a new era for the management of cancer and other diseases
.
Nature Rev Drug Discov
2017
,
16
,
203
222
. doi: https://doi.org/

30.

Catalanotto
C
,
Cogoni
C
,
Zardo
G.
MicroRNA in control of gene expression: an overview of nuclear functions
.
Int J Mol Sci
2016
,
17
,
1712
doi: https://doi.org/

31.

Peng
Y
,
Croce
CM.
The role of MicroRNAs in human cancer
.
Signal Transduct Targeted Ther
2016
,
1
,
15004
. doi: https://doi.org/

32.

Kim
KM
,
Abdelmohsen
K
,
Mustapic
M
,
Kapogiannis
D
,
Gorospe
M.
RNA in extracellular vesicles
.
Wiley Interdiscip Rev RNA
2017
,
8
. doi: https://doi.org/

33.

Zhang
J
,
Li
S
,
Li
L
,
Li
M
,
Guo
C
,
Yao
J
,
Mi
S.
Exosome and exosomal microRNA: trafficking, sorting, and function
.
GPB
2015
,
13
,
17
24
. doi: https://doi.org/

34.

Villarroya-Beltri
C
,
Gutiérrez-Vázquez
C
,
Sánchez-Cabo
F
,
Pérez-Hernández
D
,
Vázquez
J
,
Martin-Cofreces
N
, et al.
Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs
.
Nat Commun
2013
,
4
,
2980
. doi: https://doi.org/

35.

Koppers-Lalic
D
,
Hackenberg
M
,
Bijnsdorp
IV
,
Zini
N
,
Middeldorp
JM
,
Ylstra
B
, et al.
Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes
.
Cell Rep
2014
,
8
,
1649
1658
. doi: https://doi.org/

36.

Guduric-Fuchs
J
,
O’Connor,
A
,
Camp,
B
,
O’Neill,
CL
,
Medina,
RJ
,
Simpson
DA.
Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types
.
BMC Genomics
2012
,
13
,
357
. doi: https://doi.org/

37.

Mulcahy
LA
,
Ink
RC
,
Carter
DR.
Routes and mechanisms of extracellular vesicle uptake
.
J Extracell Vesicles
2014
,
3
. doi:  https://doi.org/

38.

Abels
ER
,
Breakefield
XO.
Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake
.
Cell Mol Neurobiol
2016
,
36
,
301
12
. doi: https://doi.org/

39.

Yona
S
,
Kim
KW
,
Wolf
Y
,
Mildner
A
,
Varol
D
,
Breker
M
, et al.
Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis
.
Immunity
2013
,
38
,
79
91
. doi: https://doi.org/

40.

Davies
LC
,
Jenkins
SJ
,
Allen
JE
,
Taylor
PR.
Tissue-resident macrophages
.
Nat Immunol
2013
,
14
,
986
95
. doi: https://doi.org/

41.

Byrne
AJ
,
Mathie
SA
,
Gregory
LG
,
Lloyd
CM.
Pulmonary macrophages: key players in the innate defence of the airways
.
Thorax
2015
,
70
,
1189
96
. doi: https://doi.org/

42.

Woo
YD
,
Jeong
D
,
Chung
DH.
Development and functions of alveolar macrophages
.
Mol Cells
2021
,
44
,
292
300
. doi: https://doi.org/

43.

Biswas
SK
,
Chittezhath
M
,
Shalova
IN
,
Lim
JY.
Macrophage polarization and plasticity in health and disease
.
Immunol Res
2012
,
53
,
11
24
. doi: https://doi.org/

44.

Chen
X
,
Tang
J
,
Shuai
W
,
Meng
J
,
Feng
J
,
Han
Z.
Macrophage polarization and its role in the pathogenesis of acute lung injury/acute respiratory distress syndrome
.
INREFB
2020
,
69
,
883
895
. doi: https://doi.org/

45.

Saradna
A
,
Do
DC
,
Kumar
S
,
Fu
QL
,
Gao
P.
Macrophage polarization and allergic asthma
.
Trans Res: J Lab Clin Med
2018
,
191
,
1
14
. doi: https://doi.org/

46.

Mantovani
A
,
Sica
A
,
Sozzani
S
,
Allavena
P
,
Vecchi
A
,
Locati
M.
The chemokine system in diverse forms of macrophage activation and polarization
.
Trends Immunol
2004
,
25
,
677
86
. doi: https://doi.org/

47.

Parisi
L
,
Gini
E
,
Baci
D
,
Tremolati
M
,
Fanuli
M
,
Bassani
B
, et al.
Macrophage polarization in chronic inflammatory diseases: killers or builders
?
J Immunol Res
2018
.
2018
,
1
25
. doi: https://doi.org/

48.

Deng
L
,
Jian
Z
,
Xu
T
,
Li
F
,
Deng
H
,
Zhou
Y
,
Lai
S
, et al.
Macrophage polarization: an important candidate regulator for lung diseases
.
Mole (Basel, Switzerland)
2023
,
28
.
2379
doi: https://doi.org/

49.

Novak
ML
,
Koh
TJ.
Macrophage phenotypes during tissue repair
.
J Leukoc Biol
2013
,
93
,
875
81
. doi: https://doi.org/

50.

Martinez
FO
,
Sica
A
,
Mantovani
A
,
Locati
M.
Macrophage activation and polarization
.
Front Biosci J VLib
2008
,
13
,
453
61
. doi: https://doi.org/

51.

Atri
C
,
Guerfali
FZ
,
Laouini
D.
Role of human macrophage polarization in inflammation during infectious diseases
.
Int J Mol Sci
2018
,
19
,
1801
. doi: https://doi.org/

52.

Zizzo
G
,
Hilliard
BA
,
Monestier
M
,
Cohen
PL.
Efficient clearance of early apoptotic cells by human macrophages requires M2c polarization and MerTK induction
.
J immunolo (Baltimore, Md. : 1950)
2012
,
189
,
3508
20
. doi: https://doi.org/

53.

Lu
J
,
Cao
Q
,
Zheng
D
,
Sun
Y
,
Wang
C
,
Yu
X
, et al.
Discrete functions of M2a and M2c macrophage subsets determine their relative efficacy in treating chronic kidney disease
.
Kidney Int
2013
,
84
,
745
55
. doi: https://doi.org/

54.

Duluc
D
,
Delneste
Y
,
Tan
F
,
Moles
M-P
,
Grimaud
L
,
Lenoir
J
, et al.
Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood
.
Am J Hematol
2007
,
110
,
4319
30
.

55.

Wu
H
,
Xu
JB
,
He
YL
,
Peng
JJ
,
Zhang
XH
,
Chen
CQ
, et al.
Tumor‐associated macrophages promote angiogenesis and lymphangiogenesis of gastric cancer
.
J Surg Oncol
2012
,
106
,
462
8
. doi: https://doi.org/

56.

Wang
H
,
Li
J
,
Zhang
H
,
Wang
M
,
Xiao
L
,
Wang
Y
, et al.
Regulation of microglia polarization after cerebral ischemia
.
Front Cell Neurosci
2023
,
17
,
1182621
. doi: https://doi.org/

57.

Ginhoux
F
,
Greter
M
,
Leboeuf
M
,
Nandi
S
,
See
P
,
Gokhan
S
, et al.
Fate mapping analysis reveals that adult microglia derive from primitive macrophages
.
Science
2010
,
330
,
841
5
. doi: https://doi.org/

58.

Ransohoff
RM
,
Brown
MA.
Innate immunity in the central nervous system
.
J Clin Invest
2012
,
122
,
1164
71
. doi: https://doi.org/

59.

Gyoneva
S
,
Ransohoff
RM.
Inflammatory reaction after traumatic brain injury: therapeutic potential of targeting cell–cell communication by chemokines
.
Trends Pharmacol Sci
2015
,
36
,
471
80
. doi: https://doi.org/

60.

Lian
L
,
Zhang
Y
,
Liu
L
,
Yang
L
,
Cai
Y
,
Zhang
J
, et al.
Neuroinflammation in ischemic stroke: focus on MicroRNA-mediated polarization of microglia
.
Front Mol Neurosci
2020
,
13
,
612439
.

61.

Var
SR
,
Shetty
AV
,
Grande
AW
,
Low
WC
,
Cheeran
MC.
Microglia and macrophages in neuroprotection, neurogenesis, and emerging therapies for stroke
.
Cells
2021
,
10
,
3555
. doi: https://doi.org/

62.

Latta
CH
,
Sudduth
TL
,
Weekman
EM
,
Brothers
HM
,
Abner
EL
,
Popa
GJ
, et al.
Determining the role of IL-4 induced neuroinflammation in microglial activity and amyloid-β using BV2 microglial cells and APP/PS1 transgenic mice
.
J Neuroinflammation
2015
,
12
,
1
13
.

63.

Chhor
V
,
Le Charpentier
T
,
Lebon
S
,
Oré
MV
,
Celador
IL
,
Josserand
J
, et al.
Characterization of phenotype markers and neuronotoxic potential of polarised primary microglia in vitro
.
Brain Behav Immun
2013
,
32
,
70
85
.

64.

Mecha
M
,
Feliú
A
,
Carrillo-Salinas
F
,
Rueda-Zubiaurre
A
,
Ortega-Gutiérrez
S
,
de Sola
RG
, et al.
Endocannabinoids drive the acquisition of an alternative phenotype in microglia
.
Brain Behav Immun
2015
,
49
,
233
45
.

65.

Fumagalli
S
,
Perego
C
,
Pischiutta
F
,
Zanier
ER
,
De Simoni
MG.
The ischemic environment drives microglia and macrophage function
.
Front Neurol
2015
,
6
,
81
. doi: https://doi.org/

66.

Kim
E
,
Cho
S.
Microglia and monocyte-derived macrophages in stroke
.
Neurother
2016
,
13
,
702
718
. doi: https://doi.org/

67.

Colton
CA.
Heterogeneity of microglial activation in the innate immune response in the brain
.
J Neuroimmune Pharmacol
2009
,
4
,
399
418
. doi: https://doi.org/

68.

Ma
Y
,
Wang
J
,
Wang
Y
,
Yang
GY.
The biphasic function of microglia in ischemic stroke
.
Prog Neurobiol
2017
,
157
,
247
272
. doi: https://doi.org/

69.

Hu
X
,
Li
P
,
Guo
Y
,
Wang
H
,
Leak
RK
,
Chen
S
, et al.
Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia
.
Stroke
2012
,
43
,
3063
70
. doi: https://doi.org/

70.

Liu
L
,
Liu
X
,
Wang
R
,
Yan
F
,
Luo
Y
,
Chandra
A
, et al.
Mild focal hypothermia regulates the dynamic polarization of microglia after ischemic stroke in mice
.
Neurol Res
2018
,
40
,
508
515
. doi: https://doi.org/

71.

Malainou
,
C
,
Abdin
,
SM
,
Lachmann
,
N
,
Matt
U
,
Herold
,
S.
Alveolar macrophages in tissue homeostasis, inflammation, and infection: evolving concepts of therapeutic targeting
.
J Clin Invest
2023
,
133
e170501
. doi: https://doi.org/

72.

Zhou
BW
,
Liu
HM
,
Xu
F
,
Jia
XH.
The role of macrophage polarization and cellular crosstalk in the pulmonary fibrotic microenvironment: a review
.
Cell Commun Signaling
2024
,
22
,
172
. doi: https://doi.org/

73.

Pervizaj-Oruqaj
L
,
Ferrero
MR
,
Matt
U
,
Herold
S.
The guardians of pulmonary harmony: alveolar macrophages orchestrating the symphony of lung inflammation and tissue homeostasis
.
J Eur Respir Soci
2024
,
33
,
230263
. doi: https://doi.org/

74.

Dordoe
C
,
Huang
W
,
Bwalya
C
,
Wang
X
,
Shen
B
,
Wang
H
, et al.
The role of microglial activation on ischemic stroke: modulation by fibroblast growth factors
.
Cytokine Growth Factor Rev
2023
,
74
,
122
133
. doi: https://doi.org/

75.

Ugidos
IF
,
Pistono
C
,
Korhonen
P
,
Gómez-Budia
M
,
Sitnikova
V
,
Klecki
P
, et al.
Sex differences in poststroke inflammation: a focus on microglia across the lifespan
.
Stroke
2022
,
53
,
1500
1509
. doi: https://doi.org/

76.

Gerganova
G
,
Riddell
A
,
Miller
AA.
CNS border-associated macrophages in the homeostatic and ischaemic brain
.
Pharmacol Ther
2022
,
240
,
108220
. doi: https://doi.org/

77.

Subedi
L
,
Gaire
BP.
Phytochemicals as regulators of microglia/macrophages activation in cerebral ischemia
.
Pharmacol Res
2021
,
165
,
105419
. doi: https://doi.org/

78.

Silvin
A
,
Qian
J
,
Ginhoux
F.
Brain macrophage development, diversity and dysregulation in health and disease
.
Cell Mole Immunol
2023
,
20
,
1277
1289
. doi: https://doi.org/

79.

Khan
F
,
Pang
L
,
Dunterman
M
,
Lesniak
MS
,
Heimberger
AB
,
Chen
P.
Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy
.
J Clin Invest
2023
,
133
e163446
. doi:  https://doi.org/

80.

Feng
Y
,
Hu
X
,
Zhang
Y
,
Wang
Y.
The role of microglia in brain metastases: mechanisms and strategies
.
Aging Dis
2024
,
15
,
169
185
. doi: https://doi.org/

81.

Ragaller
M
,
Richter
T.
Acute lung injury and acute respiratory distress syndrome
.
JEmer Trauma and Shock
2010
,
3
,
43
.
51
. doi: https://doi.org/

82.

Lew
TW
,
Kwek
TK
,
Tai
D
,
Earnest
A
,
Loo
S
,
Singh
K
, et al.
Acute respiratory distress syndrome in critically ill patients with severe acute respiratory syndrome
.
JAMA
2003
,
290
,
374
80
. doi: https://doi.org/

83.

Bellani
G
,
Laffey
JG
,
Pham
T
,
Fan
E
,
Brochard
L
,
Esteban
A
, et al.
LUNG SAFE Investigators. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries
.
JAMA
2016
,
315
,
788
800
. doi: https://doi.org/

84.

Matthay
MA
,
Zemans
RL
,
Zimmerman
GA
,
Arabi
YM
,
Beitler
JR
,
Mercat
A
, et al.
Acute respiratory distress syndrome
.
Nat Rev Dis Primers
2019
,
5
,
18
. doi: https://doi.org/

85.

Jagrosse
ML
,
Dean
DA
,
Rahman
A
,
Nilsson
BL.
RNAi therapeutic strategies for acute respiratory distress syndrome
.
Transl Res
2019
,
214
,
30
49
. doi: https://doi.org/

86.

Song
L
,
Zhou
F
,
Cheng
L
,
Hu
M
,
He
Y
,
Zhang
B
, et al.
MicroRNA-34a suppresses autophagy in alveolar type II epithelial cells in acute lung injury by inhibiting FoxO3 expression
.
Inflammation
2017
,
40
,
927
936
. doi: https://doi.org/

87.

Cao
W
,
Dai
H
,
Yang
S
,
Liu
Z
,
Yi Chen
Q.
Increased serum miR-300 level serves as a potential biomarker of lipopolysaccharide-induced lung injury by targeting IκBα
.
Die Pharmazie
2017
,
72
,
5
9
. doi: https://doi.org/

88.

Lin
L
,
Hron
JD
,
Peng
SL.
Regulation of NF-κB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a
.
Immunity
2004
,
21
,
203
13
. doi: https://doi.org/

89.

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
,
1758
1771
. doi: https://doi.org/

90.

Goodwin
AJ
,
Li
P
,
Halushka
PV
,
Cook
JA
,
Sumal
AS
,
Fan
H.
Circulating miRNA 887 is differentially expressed in ARDS and modulates endothelial function
.
Am J Physiol Lung Cell Mol Physiol
2020
,
318
,
L1261
L1269
. doi: https://doi.org/

91.

Shah
D
,
Das
P
,
Alam
MA
,
Mahajan
N
,
Romero
F
,
Shahid
M
, et al.
MicroRNA-34a promotes endothelial dysfunction and mitochondrial-mediated apoptosis in murine models of acute lung injury
.
Am J Respir Cell Mol Biol
2019
,
60
,
465
477
. doi: https://doi.org/

92.

Fang
,
Y
,
Gao
F
,
Hao
J
,
Liu
,
Z.
microRNA-1246 mediates lipopolysaccharide-induced pulmonary endothelial cell apoptosis and acute lung injury by targeting angiotensin-converting enzyme 2
.
Am J Trans Res
2017
,
9
,
1287
.

93.

Li
W
,
Qiu
X
,
Liu
J
,
Han
Y
,
Wei
D
,
Ji
G
,
Jiang
He.
miR-27a protects against acute lung injury in LPS-treated mice by inhibiting NF-κB-mediated inflammatory response
.
Int J Clin Exp Path
2018
,
11
,
2980
2989

94.

Yang
Y
,
Yang
F
,
Yu
X
,
Wang
B
,
Yang
Y
,
Zhou
X
, et al.
miR-16 inhibits NLRP3 inflammasome activation by directly targeting TLR4 in acute lung injury
.
Biomed. Pharmacother
2019
,
112
,
108664
.

95.

Yang
J
,
Chen
Y
,
Jiang
K
,
Zhao
G
,
Guo
S
,
Liu
J
, et al.
MicroRNA‐182 supplies negative feedback regulation to ameliorate lipopolysaccharide‐induced ALI in mice by targeting TLR4
.
J Cell Physiol
2020
,
235
,
5925
5937
. doi: https://doi.org/

96.

Yu
Y
,
Yu
G
,
Ding
Z
,
Li
S
,
Fang
Q.
Overexpression of miR-145-5p alleviated LPS-induced acute lung injury
.
J Biol Regul Homeost Agents
2019
,
33
,
1063
72
.

97.

Li
X
,
Wang
J
,
Wu
H
,
Guo
P
,
Wang
C
,
Wang
Y
,
Zhang
Z.
Reduced peripheral blood miR‑140 may be a biomarker for acute lung injury by targeting Toll‑like receptor 4 (TLR4)
.
Exper Ther Med
2018
,
16
,
3632
3638
. doi: https://doi.org/

98.

Liu
C
,
Xiao
K
,
Xie
L.
Advances in mesenchymal stromal cell therapy for acute lung injury/acute respiratory distress syndrome
.
Front Cell Dev Biol
2022
,
10
,
951764
. doi: https://doi.org/

99.

Zhu
M
,
Ma
X
,
Huang
J
,
Lu
FG
,
Chen
Y
,
Hu
J
, et al.
Extracellular vesicle-derived miR-1249-5p regulates influenza A virus-induced acute lung injury in RAW246.7 cells through targeting SLC4A1
.
Microbes Infect
2022
,
24
,
104998
. doi: https://doi.org/

100.

Li
C
,
Deng
C
,
Zhou
T
,
Hu
J
,
Dai
B
,
Yi
F
, et al.
MicroRNA-370 carried by M2 macrophage-derived exosomes alleviates asthma progression through inhibiting the FGF1/MAPK/STAT1 axis
.
Int J Biol Sci
2021
,
17
,
1795
1807
. doi: https://doi.org/

101.

Niu
Z
,
Wang
L
,
Qin
X
,
Ye
Z
,
Xie
B
,
Hu
Y.
Macrophage derived miR-7219-3p-containing exosomes mediate fibroblast trans-differentiation by targeting SPRY1 in silicosis
.
Toxicology
2022
,
479
,
153310
. doi: https://doi.org/

102.

Xu
Y
,
Zhang
C
,
Cai
D
,
Zhu
R
,
Cao
Y.
Exosomal miR-155-5p drives widespread macrophage M1 polarization in hypervirulent Klebsiella pneumoniae-induced acute lung injury via the MSK1/p38-MAPK axis
.
CMBL
2023
,
28
,
92
. doi: https://doi.org/

103.

Guiot
J
,
Cambier
M
,
Boeckx
A
,
Henket
M
,
Nivelles
O
,
Gester
F
, et al.
Macrophage-derived exosomes attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3p
.
Thorax
2020
,
75
,
870
881
. doi: https://doi.org/

104.

Todoran
R
,
Falcione
SR
,
Clarke
M
,
Joy
T
,
Boghozian
R
,
Jickling
GC.
microRNA as a therapeutic for ischemic stroke
.
Neurochem Int
2023
,
163
,
105487
. doi: https://doi.org/

105.

Li
SS
,
Wu
JJ
,
Xing
XX
,
Zhang
JP
,
Shan
CL
,
Hua
XY
, et al.
Focal ischemic stroke modifies microglia-derived exosomal miRNAs: potential role of mir-212-5p in neuronal protection and functional recovery
.
Biol Res
2023
,
56
,
52
. doi: https://doi.org/

106.

Song
Y
,
Li
Z
,
He
T
,
Qu
M
,
Jiang
L
,
Li
W
, et al.
M2 microglia-derived exosomes protect the mouse brain from ischemia-reperfusion injury via exosomal miR-124
.
Theranostics
2019
,
9
,
2910
2923
. doi: https://doi.org/

107.

Song
Y
,
Shi
R
,
Liu
Y
,
Cui
F
,
Han
L
,
Wang
C
, et al.
M2 Microglia extracellular vesicle miR-124 regulates neural stem cell differentiation in ischemic stroke via AAK1/NOTCH
.
Stroke
2023
,
54
,
2629
2639
. doi: https://doi.org/

108.

Wang
M
,
Yang
Y
,
Guo
Y
,
Tan
R
,
Sheng
Y
,
Chui
H
, et al.
Xiaoxuming decoction cutting formula reduces LPS-stimulated inflammation in BV-2 cells by regulating miR-9-5p in microglia exosomes
.
Front Pharmacol
2023
,
14
,
1183612
. doi: https://doi.org/

109.

Wei
M
,
Li
C
,
Yan
Z
,
Hu
Z
,
Dong
L
, et al.
Activated microglia exosomes mediated miR-383-3p promotes neuronal necroptosis through inhibiting ATF4 expression in intracerebral hemorrhage
.
Neurochem Res
2021
,
46
,
1337
1349
. doi: https://doi.org/

110.

Xie
L
,
Zhao
H
,
Wang
Y
,
Chen
Z.
Exosomal shuttled miR-424-5p from ischemic preconditioned microglia mediates cerebral endothelial cell injury through negatively regulation of FGF2/STAT3 pathway
.
Exp Neurol
2020
,
333
,
113411
. doi: https://doi.org/

111.

Zhang
D
,
Cai
G
,
Liu
K
,
Zhuang
Z
,
Jia
KS
,
Pei
S
, et al.
Microglia exosomal miRNA-137 attenuates ischemic brain injury through targeting Notch1
.
Aging (Milano)
2021
,
13
,
4079
4095
. doi: https://doi.org/

112.

Salucci
S
,
Bartoletti Stella
A
,
Battistelli
M
,
Burattini
S
,
Bavelloni
A
,
Cocco
LI
, et al.
How inflammation pathways contribute to cell death in neuro-muscular disorders
.
Biomol
2021
,
11
,
1109
.

113.

Datta
A
,
Sarmah
D
,
Mounica
L
,
Kaur
H
,
Kesharwani
R
,
Verma
G
, et al.
Cell death pathways in ischemic stroke and targeted pharmacotherapy
.
Transl Stroke Res
2020
,
11
,
1185
1202
. doi: https://doi.org/

114.

Radak
D
,
Katsiki
N
,
Resanovic
I
,
Jovanovic
A
,
Sudar-Milovanovic
E
,
Zafirovic
S
, et al.
Apoptosis and acute brain ischemia in ischemic stroke
.
Curr Vasc Pharmacol
2017
,
15
,
115
122
. doi: https://doi.org/

115.

Fan
J
,
Saft
M
,
Sadanandan
N
,
Gonzales-Portillo
B
,
Park
YJ
,
Sanberg
PR
, et al.
LncRNAs stand as potent biomarkers and therapeutic targets for stroke
.
Front Aging Neurosci
2020
,
12
,
594571
. doi: https://doi.org/

116.

Yuan
J
,
Amin
P
,
Ofengeim
D.
Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases
.
Nat Rev Neurosci
2019
,
20
,
19
33
. doi: https://doi.org/

117.

Pan
Y
,
Jiao
Q
,
Wei
W
,
Zheng
T
,
Yang
X
,
Xin
W.
Emerging role of LncRNAs in ischemic stroke-novel insights into the regulation of inflammation
.
J Inflamm Res
2021
,
14
,
4467
4483
. doi: https://doi.org/

118.

Ge
Q
,
Zhou
Y
,
Lu
J
,
Bai
Y
,
Xie
X
,
Lu
Z.
miRNA in plasma exosome is stable under different storage conditions
.
Mole (Basel, Switzerland)
2014
,
19
,
1568
75
. doi: https://doi.org/

119.

Weng
R
,
Jiang
Z
,
Gu
Y.
Noncoding RNA as diagnostic and prognostic biomarkers in cerebrovascular disease
.
Oxid Med Cell Longev
2022
, 2022, 
8149701
. doi: https://doi.org/

120.

Zhou
J
,
Chen
L
,
Chen
B
,
Huang
S
,
Zeng
C
,
Wu
H
, et al.
Increased serum exosomal miR-134 expression in the acute ischemic stroke patients
.
BMC Neurol
2018
,
18
,
198
. doi: https://doi.org/

121.

Wang
W
,
Li
D
,
Li
R
,
Zhou
X
,
Yu
D
,
Lan
X
, et al.
Diagnosis of hyperacute and acute ischaemic stroke: the potential utility of exosomal microRNA-21-5p and microRNA-30a-5p
.
Cerebrovasc Dis (Basel, Switzerland)
2018
,
45
,
204
212
. doi: https://doi.org/

122.

Chen
Y
,
Song
Y
,
Huang
J
,
Qu
M
,
Zhang
Y
,
Geng
J
, et al.
Increased circulating exosomal miRNA-223 is associated with acute ischemic stroke
.
Front Neurol
2017
,
8
,
57
. doi: https://doi.org/

123.

Ji
Q
,
Ji
Y
,
Peng
J
,
Zhou
X
,
Chen
X
,
Zhao
H
, et al.
Increased brain-specific MiR-9 and MiR-124 in the serum exosomes of acute ischemic stroke patients
.
PLoS One
2016
,
11
,
e0163645
. doi: https://doi.org/

124.

Li
D
,
Liu
J
,
Wang
W
,
Li
R
,
Yu
D
,
Lan
X
,
Li
JP.
Plasma exosomal miR-422a and miR-125b-2-3p serve as biomarkers for ischemic stroke
.
Curr Neurovasc Res
2017
,
14
,
330
337
. doi: https://doi.org/

125.

Parzibut
G
,
Henket
M
,
Moermans
C
,
Struman
I
,
Louis
E
,
Malaise
M
,
Louis
R
, M et al.
A blood exosomal miRNA signature in acute respiratory distress syndrome
.
Front Mole Biosci
2021
,
8
,
640042
. doi: https://doi.org/

126.

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
,
1
11
. doi: https://doi.org/

127.

Mittelbrunn
M
,
Sánchez-Madrid
F.
Intercellular communication: diverse structures for exchange of genetic information
.
Nat Rev Mol Cell Biol
2012
,
13
,
328
35
. doi: https://doi.org/

128.

Cai
G
,
Cai
G
,
Zhou
H
,
Zhuang
Z
,
Liu
K
,
Pei
S
, et al.
Mesenchymal stem cell-derived exosome miR-542-3p suppresses inflammation and prevents cerebral infarction
.
Stem Cell Res Ther
2021
,
12
,
2
. doi: https://doi.org/

129.

Lan
B
,
Dong
X
,
Yang
Q
,
Luo
Y
,
Wen
H
,
Chen
Z
,
Chen
H.
Exosomal microRNAs: an emerging important regulator in acute lung injury
.
ACS Omega
2023
,
8
,
35523
35537
. doi: https://doi.org/

130.

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 Longevity
2021
,
2021
.

131.

Kaur
S
,
Singh
SP
,
Elkahloun
AG
,
Wu
W
,
Abu-Asab
MS
,
Roberts
DD.
CD47-dependent immunomodulatory and angiogenic activities of extracellular vesicles produced by T cells
.
Matrix Biol
2014
,
37
,
49
59
. doi: https://doi.org/

132.

Kibria
G
,
Ramos
EK
,
Lee
KE
,
Bedoyan
S
,
Huang
S
,
Samaeekia
R
, et al.
A rapid, automated surface protein profiling of single circulating exosomes in human blood
.
Sci Rep
2016
,
6
,
36502
. doi: https://doi.org/

133.

Brown
EJ
,
Frazier
WA.
Integrin-associated protein (CD47) and its ligands
.
Trends Cell Biol
2001
,
11
,
130
5
. doi: https://doi.org/

134.

Jaiswal
S
,
Jamieson
CH
,
Pang
WW
,
Park
CY
,
Chao
MP
,
Majeti
R
, et al.
CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis
.
Cell
2009
,
138
,
271
85
. doi: https://doi.org/

135.

Giovannelli
L
,
Bari
E
,
Jommi
C
,
Tartara
F
,
Armocida
D
,
Garbossa
D
, et al.
Mesenchymal stem cell secretome and extracellular vesicles for neurodegenerative diseases: risk-benefit profile and next steps for the market access
.
Bioact Mater
2023
,
29
,
16
35
. doi: https://doi.org/

136.

Seyedaghamiri
F
,
Salimi
L
,
Ghaznavi
D
,
Sokullu
E
,
Rahbarghazi
R.
Exosomes-based therapy of stroke, an emerging approach toward recovery
.
CCS
2022
,
20
,
110
. doi: https://doi.org/

137.

Mahjoubin-Tehran
M
,
Rezaei
S
,
Jesmani
A
,
Birang
N
,
Morshedi
K
,
Khanbabaei
H
, et al.
New epigenetic players in stroke pathogenesis: from non-coding RNAs to exosomal non-coding RNAs
.
Biomed Pharmacother
2021
,
140
,
111753
. doi: https://doi.org/

138.

Wang
C
,
Börger
V
,
Sardari
M
,
Murke
F
,
Skuljec
J
,
Pul
R
, et al.
Mesenchymal stromal cell–derived small extracellular vesicles induce ischemic neuroprotection by modulating leukocytes and specifically neutrophils
.
Stroke
2020
,
51
,
1825
1834
. doi: https://doi.org/

139.

Lener
T
,
Gimona
M
,
Aigner
L
,
Börger
V
,
Buzas
E
,
Camussi
G
, et al.
Applying extracellular vesicles based therapeutics in clinical trials—an ISEV position paper
.
JEV
2015
,
4
,
30087
. doi: https://doi.org/

140.

Kilchert
C
,
Wittmann
S
,
Vasiljeva
L.
The regulation and functions of the nuclear RNA exosome complex
.
Nat Rev Mol Cell Biol
2016
,
17
,
227
39
. doi: https://doi.org/

Author notes

Xianbin Wang and Ting Wang contribute equally to this work and are considered co-first authors.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected]