Summary
Exosomal miRNAs are a type of non-coding RNA molecules present in exosomes. MiRNAs are involved in the regulation of a variety of physiological and pathological processes by transmitting information between cells through the exosome, a carrier of intercellular communication. During inflammatory responses, exosomal miRNAs can be involved in regulating the activation of inflammatory cells and the release of inflammatory mediators, thus affecting the development of inflammatory diseases. Therefore, exosomal miRNAs may be promising biomarkers for monitoring disease progression based on their functions and changes. In addition, since exosome prevents miRNAs from being degraded by RNase, drug development targeting the release of exosomal miRNA contents lays the foundation for innovative targeted therapies in the future. This review focuses on exosomal miRNAs with the aim of combing and mastering its latest developments in the current research of inflammatory diseases.
Keywords
miRNAs; inflammatory diseases; exosomal; biomarkers
Introduction
MicroRNAs (miRNAs) are a class of single-stranded non-coding RNAs approximately 21-23 nucleotides in length that regulate gene expression by binding to the 3′-untranslated region (3′-UTR) of specific mRNAs1. MiRNAs account for 1-5% of all genes in the human genom2 and regulate approximately one-third of the human genome through their multiple targets3. MiRNAs, as pivotal regulators of gene expression, intricately govern various biological cascades including differentiation, growth, development and metabolism. The complexity arising from their diminutive size, expression levels, numerous repetitive sequences within the genome and distinct modes of action poses singular hurdles in unraveling the intricate functional landscape of miRNAs4.
Extracellular vesicles (EVs) have been shown to carry many biomolecules RNA, DNA, lipids, proteins, and metabolites5, which also include miRNAs6. In recent years, numerous investigations have underscored the pivotal role of miRNAs in employing exosomes as effective vehicles for facilitating intercellular signaling and communication. Inflammatory diseases are a group of disorders in which tissue damage and inflammatory processes occur as a result of an abnormal response of the body's immune system due to a variety of causes7,8,9. Over the past three decades, epidemiological investigations have revealed a notable increase in the occurrence of inflammatory disorders10. It is noted that they are often associated with a high risk for cancer11,12.
The pivotal function of exosomal miRNAs in modulating gene expression, along with their unregulated expression patterns have been observed to be altered significantly across various types of human cancers, has garnered considerable research interest and attention, but studies on exosomal miRNAs in inflammatory diseases are less clear. The purpose of this review is to further summarize the potential influence of exosomal miRNAs on inflammatory conditions, aiming to provide deep insights.
Exosomal miRNAs biogenesis and function
In 1993, Ambros and his team made a groundbreaking discovery, unveiling the role of a gene named lin-4 in the developmental processes of the nematode Caenorhabditis elegans, and was in fact a small non-protein-coding RNA molecule. This revelation marked the inception of the miRNA field13. In the 2000, another miRNA, let-7, was discovered, which regulates the timing of development in Hidradenitis elegans14. The discovery of this miRNA is a major step forward in the development of the nematode.
The production of miRNA begins in the nucleus and ends in the cytoplasm15. Initially, miRNA synthesis is mainly transcribed through the action of RNA polymerase II16. It then undergoes the process of capping, splicing, and addition of polyadenylate tails, which results in the formation of primitive miRNAs (pri-miRNAs) containing at least one hairpin structure17,18. Within the nucleus, the primary miRNAs (pri-miRNAs) undergo cleavage by the enzyme Drosha, aided by its cofactor DGCR8, resulting in the formation of precursor miRNAs (pre-miRNAs) that range in length from 70 to 100 nucleotides19. The pre-miRNAs traverse the nuclear pore and enter the cytoplasm with the assistance of Exportin-520. Within the cytoplasm, the enzyme Dicer processes the pre-miRNAs, resulting in the formation of a double-stranded RNA duplex that includes the mature miRNA and its matching antisense strand21. Following cleavage, the deconjugating enzyme separates the double-stranded RNA, yielding a mature miRNA single strand, which subsequently associates with the RNA-induced silencing complex (RISC) that includes the Argonaute protein (Ago2)22. Upon binding, the RISC complex engages with the 3' untranslated region (3'UTR) of the target mRNA, triggering its degradation and suppressing its translation, thus regulating gene expression23,24(Figure 1).
Over a decade ago, Valadi H and colleagues introduced the notion that miRNAs and mRNAs can be exchanged between cells via vesicular transport and protein-mediated mechanisms. Their groundbreaking research unveiled the presence of these molecules within extracellular vesicles (EVs) released by various cell lines. This study confirmed that these EVs can be efficiently taken up by recipient cells, facilitating the delivery of their molecular contents into target cells25.
Exosomes typically featuring diameters within the range of 40 to 160 nanometers, which classify them as smaller than 200 nm in diameter, distinguishing them as a distinct type of small EVs (sEVs)26,27,28. Exosomes exhibit a ubiquitous presence across various bodily fluids in humans, encompassing saliva, urine, breast milk, semen, cerebrospinal fluid, as well as ascites fluid2930. Numerous investigations conducted recently have shown that miRNAs communicate with one another between cells by using exosomes as a carrier3132. In particular, exosomal miRNAs are released by donor cells through mechanisms of paracrine or distal secretion, and then taken up by recipient cell in a variety of forms such as fusion, endocytosis and receptor33. Among the intricate mechanisms governing miRNA incorporation into exosomes, the nerve sphingomyelinase 2 (nsMase2)-related pathway was the the first protein reported to be intimately linked with the secretion of miRNAs into exosomes. Its down-regulation reduces the amounts of exosomal miRNAs, whereas its overexpression increases exosomal miRNA levels34. The level of Ago2 and its phosphorylation participate in the release of certain exosomal miRNA35. Furthermore, the heterogeneous nuclear ribonucleoprotein (hnRNP) family of proteins participates in the exosomal miRNA packaging. Specifically, hnRNPA2B1 and hnRNPA1 exhibit a remarkable ability to recognize recognize specific miRNA tetranucleotide sequences, facilitating their selective loading into exosomes36. RNA sequencing of human B cells and their associated exosomes by Koppers-Lalic D et al yielded that miRNAs featuring adenylated 3' termini were predominantly retained within the cells, whereas those with uridylylated 3' ends were preferentially sorted into exosomes37.
Due to their role in immunity and gut barrier function, exosomal miRNAs can be used as biomarkers. The non-invasive nature and ease of collection of urinary diagnostics and salivary are attracting increasing attention today. For example, miR-2909 has emerged as a specific and noninvasive biomarker in urinary exosomes of prostate cancer patients38. A group of exosomal miRNAs, including let-7a, miR-21, miR-23a, miR-150, miR-223, miR-1229 and miR-1246, can be used as diagnostic biomarkers for patients with colorectal cancer39.
In addition to its key role in tumor growth and development, exosomal miRNAs has also been proved to play a crucial function in the regulation of gene expression. The breast cancer cell lines MCF-10A and MDA-MB-231 can reduce ZO-1 gene expression in endothelial cells by releasing of miR-105 via exosomes, thereby facilitating metastasis to the lung and brain, emphasizing its role in cancer progression40.
Exosomal miRNAs have also been found to have immune response modulation. Fabbri M et al. discovered that exosomal miRNAs function as ligands, capable of binding to toll-like receptors (TLRs) and triggering immune cells activation, highlighting their immunomodulatory potential41. Exosomal miRNAs are capable of reprogramme immunoreactive factors and the function of immune target cells such as T lymphocytes, dendritic cells (DCs) and natural killer (NK) cells42. We recently reported that exosomes secreted from regulatory T cells and gingival-derived mensenchymal stem cells treated inflammatory arthritis and the miRNAs play a key role in controlling inflammatory cells and disease onset and development43,44. Epstein-Barr Virus (EBV) is the first virus known to encode miRNAs (EBV-miRNAs)45. Pegtel DM et al demonstrated that mature microRNAs encoded by EBV, when produced by infected B cells, are released through exosomes and subsequently function in uninfected recipient cells46(Table 1).
Table 1. Exosomal miRNAs in Inflammatory Diseases.
recipient-cell uptake | fusion | endocytosis | receptor-ligand | macropinocytosis |
mechanism | nerve sphingomyelinase 2 (nsMase2)-related pathway | Argonaute protein (Ago2) | heterogeneous nuclear ribonucleoprotein (hnRNP) family proteins | 3’-end miRNA |
function | intercellular communication | biomarkers | gene expression regulation | immune response modulation |
Inflammatory diseases and exosomal miRNAs
Inflammation is an immune response that can be triggered by non-infectious or infectious stimuli, for example, toxins, physical injury and cellular damage47,48. Exosomes inhibit or stimulate the activation of inflammasome, and there is considerable evidence that inflammatory diseases of many etiologies result in exosomal miRNAs that differ in content from basal production of exosomes49,50. Exosomal miRNAs can be involved in the regulation of inflammatory cell activation and release of inflammatory mediators51. For example, exosomes derived from human umbilical cord mesenchymal stem cells (huc-MSCs) attenuate mechanical anomalous pain and thermal hyperalgesia in inflammatory pain via miR-146a-5p/TRAF652. Exosomes derived from huc-MSCs mediate miR-181c to attenuate burn-induced excessive inflammation53.
Other miRNAs may promote the activation of inflammatory cells and exacerbate the inflammatory response. Exosomes of osteoarthritic chondrocytes can enhance mature IL-1β production and aggravate osteoarthritic synovitis via osteoarthritis by miR-449a-5p54.
This dual role makes exosomal miRNAs playing an important role in the development and progression of inflammatory diseases. Numerous studies have demonstrated that changes in the levels of specific exosomal miRNAs are associated with a variety of inflammatory diseases including autoimmune diseases, diabetes, cardiovascular diseases, and neuroinflammatory diseases. Next, we will describe in each system separately (Table 2).
Table 2. Summary of exosomal miRNAs profiles of indicated Inflammatory Diseases
Disease |
Exosomal miRNA |
Correlation with the disease(positive/negative) |
Origin of exosome |
effective object |
Target gene |
Significance |
References |
|
autoimmune disease | synovitis in osteoarthritis | miR-449a-5p | positive | Ostearthritic chondrocytes | macrophages | IL-1β | aggravated synovitis in osteoarthritis | 54 |
Systemic lupus erythematous (SLE) | miR-129, miR-142, miR-148b | positive | positive | Circulating macrophages | illustrate the curative promise of directing interventions at miRNAs in individuals with SLE | 55, 56 |
||
Lupus nephritis (LN) | Let-7a, miR-21 | negative | urine | 57 |
||||
Rheumatoid arthritis (RA) | miR-6089 | negative | serum | MiR-6089 regulates the production of inflammatory cytokines, including IL-6, IL-29, and TNF-α, through modulation of TLR4 signaling pathways | 58 |
|||
miR-150-5p | negative | mesenchymal stem cells | MMP14 and VEGF | inhibiting synoviocyte hyperplasia and angiogenesis | 59 |
|||
miR-885-5p, miR-6894-3p, miR-1268a | Biomarkers, diagnosis and prediction | 60 |
||||||
Multiple sclerosis (MS) | miR-26a, miR-122-5p | potential targets, biomarkers and therapeutic tools | 61, 62 |
|||||
experimental autoimmune encephalomyelitis | miR-23b-3p | negative | bone mesenchymal stem cells | microglial | via suppression of microglial pyroptosis | 63 |
||
myasthenia gravis (MG) | miR-106a-5p | correlates with MG severity and expected to be an early-onset myasthenia gravis biomarker in adults | 64, 65 |
|||||
cardiovascular system | atherogenesis | miR-27b-3p | positive | obesity-induced visceral adipocytes | PPARα | promotes endothelial | 66 |
|
acute myocardial infarction, AM | miR-152-3p, let-7i-5p | negativ | hypoxia-induced | Atg12 and Faslg | produce anti-apoptotic effects | 67 |
||
miR-93-5p | adipose-derived stromal cells | inflammatory cytokine | Atg7 and Toll-like receptor 4 (TLR4) | attenuates myocardial damage | 68 |
|||
miR-125 | Bone marrow mesenchymal stem cells | cardiomyocyte | facilitates ischemic cardiac repair | 69 |
||||
metabolic inflammation | obesity | miR-690 | negative | M2 polarized bone marrow-derived macrophages | obese mice | improve glucose tolerance and insulin sensitivity, |
70 |
|
miR-1249-3p | negative | natural killer cell-derived exosome miR-1249-3p from lean mice | mice with type 2 diabetes induced by obesity | attenuates obese insulin resistance and inflammation, enhance insulin sensitivity and relieve inflammation in adipocytes and hepatocytes | 71 |
|||
miR-155 | positive | adipose tissue macrophages in obese mice | lean mice | PPARγ | insulin resistance and glucose intolerance | 72 |
||
Type 2 diabetes | miR-320a, miR-27a | metabolic Imbalance in Type 2 Diabetic Individuals | 73 |
|||||
nonalcoholic fatty liver disease | miR-122 | positive | adipocyte-derived | Sirt1 | promotes the progression of NAFLD | 74 |
||
neurodegenerative diseases | Alzheimer's disease | miR-135a, miR-384 and miR-193b | Serum | clinical biomarkers, therapeutic targets | 75 |
|||
miR-16-5p, miR-125b-5p, miR-451a and miR-605-5p | colony stimulating factor | detected in patients with early-onset AD, | 76 |
|||||
Parkinson’s disease | miR-125, miR-210, miR-450b and miR-669b | positive | exacerbate mitochondrial impairment, immune imbalance, and inflammationand then promote contribute to the overexpression and accumulation of manganese-dependent α-synuclein, | 77, 78 |
||||
miR-1, Let-7g-3p, miR-19b, miR-19b-3p, miR-10a-5p, miR-153, miR-24, miR-331-5p, miR-409-3p, miR-505 and miR195 | potential to become biomarkers | 79, 80, 81 |
||||||
Amyotrophic lateral sclerosis | miR-155, miR-146 | Microglia | influence the neuroinflammatory | 82 |
||||
miR-27a-3p | serum | potential for clinical diagnosis | 83 |
|||||
Other Inflammatory Diseases | Inflammatory bowel disease | miR-155 | positive | intestinal epithelial cells | intestinal immune cells and inflammatory cytokines | exacerbating intestinal inflammation | 84 |
|
colitis | miR-378a-5p | negative | Human umbilical cord mesenchymal stem cell | macrophage | NLRP3 | attenuate colitis | 85 |
|
acute lung injury | miR-155 | positive | serum | macrophage | SHIP1 and SOCS1 | promotes macrophage proliferation and inflammation | 86 |
|
retinal inflammation | miR-126 | negative | mesenchymal stem cell | HMGB1 | ameliorate Hyperglycemia-Induced Retinal Inflammation | 87 |
||
periodontitis | miR-143-3p | Inflammatory Periodontal Ligament Stem Cells | macrophage | PI3K/AKT/NF-κB Signaling | Drive M1 Macrophage Polarization, potential new target for periodontitis treatment | 88 |
Exosomal miRNAs in the autoimmune diseases
There is no doubt that microRNAs play a pivotal regulatory role in the immune system89. Their transfer mechanism through exosome-mediated transfer may constitute a highly efficient pathway for fine-tuning gene expression during the immune response, enhancing the coordination of the immune response, and also significantly increasing the complexity of inter-cellular communication90. Exosomal miRNAs secreted by immune cells such as B, T cells, MSCs, macrophages, DCs and other immune cells play an important role in physiological immune responses, as well as in the development of autoimmune diseases91.
It has been shown that T cell-derived miRNAs regulate specific targets in the APC and that exosomes loaded with microRNAs are unidirectionally transferred from T cells to antigen-presenting cells92.
Systemic lupus erythematosus (SLE) is a chronic diffuse connective tissue disease caused by abnormal activation of the immune system and attack its own tissues, poses challenges in both its early therapeutic intervention and precise diagnostic process93. Exosomal miRNAs can regulate the pathogenesis of SLE mechanism, and several researches reported that in patients with SLE, the expression of miR-142, miR-148b, and miR-129 is upregulated within circulating macrophages 55,56. There is still a major morbidity and mortality associated with lupus nephritis (LN) in SLE94. Let-7a and miR-21 are reduced in urinary exosomes during LN flares57. In addition, Chen F et al found that patients diagnosed with SLE and LN display notably higher levels of exosomal miR-7974 and miR-4796-5p compared to SLE patients without LN. These elevated miRNA expressions could potentially serve as valuable biomarkers to differentiate whether SLE patients with LN and distinguish autoimmune nephritis cases more broadly95.
Rheumatoid arthritis (RA) is a systemic autoimmune condition marked by persistent inflammation of synovial tissue, ultimately leading to irreparable damage to the joints96,97. Exosomes derived miR-6089 regulates LPS/TLR4, which mediates the inflammatory response in patients with RA58. Exosomes derived from MSCs containing miR-150-5p exhibit a therapeutic potential to mitigate joint destruction in rheumatoid arthritis by suppressing angiogenesis and synovial cell proliferation59. Exosomes derived from Serum, especially miR-1268a, miR-6894-3p and miR-885-5p have potential as biomarkers for prediction and early diagnosis of RA60.
Systemic sclerosis (SSc) is an uncommon autoimmune disorder affecting the central nervous system, distinguished by persistent inflammation, demyelination, fibrotic tissue formation and a diverse array of clinical manifestations[98]. Research indicates that miRNA profiles associated with exosomes may act as an prognostic indicator for monitoring treatment responsiveness in individuals diagnosed with multiple sclerosis(MS)99. MiR-26a and miR-122-5p may play a role in the pathogenesis of MS61,62. Exosomal miR-23b-3p, released by bone marrow mesenchymal stem cells (BMSCs), exhibits a pivotal role in mitigating the severity of experimental autoimmune encephalomyelitis (EAE). This therapeutic effect is mediated through the suppression of microglial pyroptosis63. The expression of exosomal miR-106a-5p varies significantly across distinct subtypes of myasthenia gravis (MG), displaying a correlation with the severity of the disease64. Serum exosomal miRNAs are expected to be an early-onset myasthenia gravis biomarker in adults65.
Exosomal miRNAs in the cardiovascular system
The most important role of exosomal miRNAs is intercellular communication. Atherosclerosis is an inflammatory vascular disease100101. Exosomal miRNAs are key mediators of intercellular communication during the development of atherosclerosis, inducing or inhibiting the atherosclerosis by driving proatherogenic inducers or vasoprotective mediators102,103,104. Tang Y et al discovered that exosomal miR-27b-3p, originating from obesity-triggered visceral adipocytes, exerts a pro-inflammatory effect on endothelial cells and accelerates the development of atherosclerosis through the inhibition of PPARα[66]. Conversely, studies have shown that exosomes derived from M2-like macrophages, which are induced by IL-4, possess the capability to regulate inflammatory conditions in mice, including atherosclerosis105.
Myocardial infarction is usually caused by excessive or prolonged inflammatory response106. In acute myocardial infarction (AMI), the elevated levels of exosomal microRNAs that induced by low oxygen environments, counteract the apoptosis induced by hypoxia, in which let-7i-5p and miR-152-3p targeting Faslg and Atg12 respectively, to produce anti-apoptotic effects67. The exosomal miR-93-5p from adipose-derived stromal cells has a protective effect against AMI-induced myocardial injury68. Bone marrow-derived mesenchymal stem cells exhibit a cardioprotective role against myocardial infarction by releasing exosomal miR-125b, thereby mitigating cardiomyocyte apoptosis and fostering cardiac rejuvenation69.
miR-146a is a well-known anti-inflammatory miRNA27. Exosomal miR-146a-5p from cardiomyocytes stimulated M1 macrophage polarization induced the onset of an inflammatory response. On the contrary, it targets TRAF 6 to exert anti-inflammatory effects107.
Exosomal miRNAs in the metabolic inflammation
Metabolic inflammation is a chronic low-grade inflammation caused by excess nutrients and energy[108]. Inflammation is the link between metabolic syndrome, type 2 diabetes and obesity109. An important pathophysiological defect in type 2 diabetes and obesity is insulin resistance101111 and exosomal miRNAs play a key role in pathogenesis. Research has demonstrated that exosomes miR-690, released by M2-polarized bone marrow-derived macrophages in obese mice, exhibit the potential to insulin sensitivity tolerance and enhance glucose in obese mice upon administration, hence, miR-690 could represent a new treatment approach as an insulin sensitizer in the context of metabolic disorders70. Exosomes originating from NK cells of lean mice, carrying miR-1249-3p, were discovered to mitigate inflammation and insulin resistance in obese mice with type 2 diabetes. Additionally, these exosomes from lean NK cells augment insulin sensitivity and alleviate inflammation in adipocytes and hepatocytes71. Conversely, exosomes emanating from obese adipose tissue macrophages (ATMs) and containing miR-155 have been caused glucose intolerance and insulin resistance72. In type 2 diabetics, exosomes miR-320 and miR-27a are dysregulated73.
Nonalcoholic steatohepatitis (NASH) and its associated liver cirrhosis are two intermediate and posterior stages in the progression of nonalcoholic fatty liver disease (NAFLD), in which inflammation plays a key role112. Chen K et al found that the adipocyte-derived exosome miR-122 promotes the progression of NAFLD by targeting Sirt1[74]. In addition, myeloid-specific IL-6 signaling promotes the production of exosomes enriched in miR-223 to attenuate NAFLD-associated fibrosis113.
Exosomal miRNAs in neurodegenerative diseases
Neurodegenerative diseases (NDs) are a class of neurological system disorders arises due to irregularities within the neurogenic inflammatory, which usually involve in the dysfunctions of the nervous system leading to aberrant activation of inflammatory responses114,115. Exosomal miRNAs play a dual function in the regulation of neuroinflammation. Firstly, they facilitate intercellular communication among neurons, enabling the coordinated response to inflammatory stimuli within the central nervous system (CNS). Secondly, exosomal miRNAs exhibit the remarkable capability to traverse the blood brain barrier (BBB), effectively bridging the gap between the peripheral immune system and the CNS. By crossing the BBB, these miRNAs can transmit inflammatory signals originating from the periphery, allowing the CNS to respond appropriately to systemic challenges116.
Alzheimer's disease (AD) is one of the most prevalent form of chronic neurodegenerative disorder globally that leads to impaired cognition and memory117. The etiology of AD is intricately linked with inflammatory processes, potentially exacerbating cellular injury and contributing to neuronal cell death[118]. Exosomal miRNAs provide new insights in the screening and prevention of AD. In patients with AD, serum-derived exosomes exhibit distinct patterns of miRNA expression, particularly for miR-193b, miR-384, and miR-135a, which are differentially abundant compared to healthy individuals75. Furthermore, exosomes derived from colony stimulating factor (CSF) in individuals diagnosed with early-stage AD reveal notable variations in the levels of miR-16-5p, miR-125b-5p, miR-605-5p, and miR-451a, suggesting these miRNAs may serve as potential biomarkers for the early detection of AD76.
Parkinson's disease (PD) is the second most common ND after AD. Exosome miR-450b, miR-125, miR-669b and miR-210 exacerbate mitochondrial dysfunction, immune dysregulation, and inflammatory processes via diverse signaling cascades. This, in turn, fosters the overexpression and accumulation of manganese-dependent α-synuclein, a pivotal factor in the progression of PD77,78. Multiple studies showed that exosomal miR-1, Let-7g-3p, miR-10a-5p, miR-19b, miR-19b-3p, miR-24, miR-153, miR195, miR-331-5p, miR-409-3p and miR-505 are all aberrantly expressed in PD and have the potential to become PD biomarkers79,80,81
Amyotrophic lateral sclerosis (ALS) is a catastrophic chronic progressive ND[119], microglia release exosomes rich in miR-155 and miR-146 that influence the neuroinflammatory processes of ALS82. An investigation contrasting of miR-27a-3p expression within serum exosomes between ALS patients and healthy individuals uncovered a potential link between decreased miR-27a-3p levels and ALS progression. This discovery emphasizes the promising role of this exosomal miRNA as a diagnostic biomarker for ALS, offering a potential tool for early detection and monitoring of the disease83.
Exosomal miRNAs in other inflammatory diseases
Inflammatory bowel disease (IBD) is a recurrent and chronic inflammatory disease[120], and exosomes released from intestinal epithelial cells containing miR-155 are able to modulate the activation of intestinal immune cells and the production of inflammatory cytokines, thereby exacerbating intestinal inflammation[84]. Conversely, miR-146a with anti-inflammatory, effectively hinder the synthesis of inflammatory mediators, thereby to reduce the occurrence of inflammation121.
NLRP3 inflammasome are large intracellular multimeric protein complexes formed in the cytosol, which play a central act as in the occurrence of inflammation122,123. It has been shown that intracellular inflammatory responses and cellular pyroptosis following NLRP3 activation promote the production and release of exosomes124. These exosomes, abundant in distinct miRNA molecules, modulate the inflammatory responses of recipient cells via diverse mechanisms[87,125]. For example, exosomes derived from hucMSCs mitigate colitis by modulating macrophage pyroptosis via the miR-378a-5p/NLRP3 pathway85.
Jiang K et al observed a significant enrichment of serum exosomes in the peripheral blood of mice exhibiting adverse lung inflammation acute lung injury (ALI). These exosomes are selectively loaded with miRNAs and miR-155 being the most abundant. In vivo, these exosomes exhibited the capacity to interact with lung macrophages, contributing to lung injury. In vitro, analysis revealed that miR-155, originating from the serum exosomes, promoted inflammation and macrophage proliferation by regulating key genes such as SOCS1 and SHIP1, respectively86.
Exosomes derived from MSCs regulate miR-126 by targeting HMGB1 to ameliorate retinal inflammation induced by hyperglycemia[87]. Periodontal ligament stem cells (PDLSCs) during inflammatory conditions have been shown to enhance M1 macrophage polarization via a mechanism involving exosomal miR-143-3p. This exosomal miRNA modulates the PI3K/AKT/NF-κB signaling pathway, thereby facilitating the polarization process88.
Exosomal miRNAs detection and treatment capacity
MiRNAs are ubiquitous in all body fluid types tested. The use of specific miRNA concentrations in body fluids has the potential to be used as a biomarker to detect and monitor a variety of physiopathological conditions126. Currently, diverse techniques facilitate the detection of exosomal miRNA, including quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), ratiometric fluorescent bioprobes, and some quantitative tests based on surface-enhanced Raman scattering (SERS), ratiometric electrochemistry and localized surface plasmon resonance (LSPR), respectively127.
Exosomes, as cellular vehicles, exhibit immense promise as therapeutic tools for diverse pathologies, owing to their efficient capacity to transport small molecules between cells, facilitating targeted delivery and communication128. They act as cell-to-cell "couriers", delivering critical molecular information with pinpoint accuracy. A notable benefit of utilizing exosomal miRNAs as intercellular signaling molecules lies in their protective shield against degradation by RNase enzymes, subsequently enhancing the the efficiency of miRNA delivery to target cells129 and they can be securely preserved in vitro under 4°C conditions for a duration of 48 hours, maintaining their stability126. Meanwhile, due to the crucial role of exosomal miRNAs in the onset and progression of numerous diseases, attention has shifted towards to be placed on the targeted release of exosomal miRNA contents for drug development. Existing data demonstrate that manipulation of exosomal miRNAs in vitro may be as an efficacious means for delivering miRNAs to target organs, thereby enhancing their therapeutic efficacy130. Exosomes have proven effective in transporting siRNAs to targeted cell types within mice, showcasing their versatility in targeted delivery systems131. Furthermore, exosomes possess the ability to traverse the BBB, and various investigations have been undertaken exploring their potential as a therapeutic miRNA delivery vehicle for ND116.
Discussion
In summary, although significant progress has been made in recent years in the study of the relationship between exosomal miRNAs and diseases, there are still many uncharted areas and challenges that need to be further explored. Firstly, in-depth studies on the specific mechanisms of action and regulatory networks of exosomal miRNAs in different types of diseases are needed to reveal their key roles in the disease process. Second, novel therapeutic strategies and drugs targeting exosomal miRNAs need to be developed to provide new ideas and approaches for disease prevention and treatment. The study of exosomes and their miRNA contents not only determines the mechanism of their intercellular communication, but also opens up a brand-new pathway for the treatment of diseases. Exosomes miRNAs can be used as therapeutic targets or miRNAs can be delivered to the target cells through the exosomes in order to achieve the precise treatment of diseases, which demonstrates a great potential for application and a broad prospect for development.
References
Figure Legends(0)
Supplemental information(0)