Abstract
Inflammatory bowel disease (IBD), which includes Crohn’s disease (CD) and ulcerative colitis (UC), is a complex condition without a definite cause. During IBD, immune cells such as macrophages release proinflammatory cytokines and chemokines, contributing to intestinal barrier integrity dysfunction. IBD is largely influenced by macrophages, which are classified into subtypes M1 and M2. M1 macrophages have been found to contribute to the development of IBD, whereas M2 macrophages alleviate IBD. Hence, agents that cause increased polarization of the M2 phenotype could help repair IBD. Exosomes, as ubiquitous conveyors of intercellular messages, are involved in immune responses and immune-mediated disease processes. Exosomes and their microRNA (miRNA) from healthy cells have been found to polarize macrophages to M2 to repair IBD due to their anti-inflammatory properties; however, those from inflammatory-driven cells and disease cells promote M1 macrophages to perpetuate IBD. Here, we review the biogenesis, biochemical composition, and sources of exosomes, as well as the roles of exosomes as extracellular vesicles in regulation of macrophages to repair IBD.
Keywords: Exosomes, extracellular vesicles, inflammatory bowel disease, macrophage
Introduction
IBDs are chronic inflammatory gut diseases that include Crohn’s disease (CD) and ulcerative colitis (UC) [1], resulting from the interplay of microbial, environmental, immunological, and genetic variables [2]. IBD is becoming more common in Western nations, as its frequency is rising quickly in recently industrialized nations [3]. In the IBD microenvironment, there is the secretion of several proinflammatory cytokines from immune cells, including interferon-gamma (IFN-γ), interleukin-17F (IL-17F), IL-1α, and IL-25 [4].
Macrophages play essential roles in IBD and are the main gatekeeper of intestinal immunological homeostasis [5]; nonetheless, IBD is linked to improper macrophage activation [6,7]. There is growing evidence that M1 macrophages, which are traditionally activated, predominate M2 macrophages, which are alternately activated and play a part in the development of IBD [8]. Therapeutic substances that have the ability to convert M1 macrophages that promote inflammation into M2 macrophages that inhibit it could, therefore, be helpful for IBD mitigation [8]. Consequently, controlling macrophage polarization is a viable treatment option for IBD [9].
Extracellular vesicles (EVs) are protein and phospholipid structures in which cells constantly discharge, and appear in the form of smaller (30-200 nanometers (nm)) and bigger (micron size) particulates [10]. Although these vesicles are referred to as EVs, the smaller ones are frequently dubbed exosomes [10]. Most live cells produce exosomes, which are lipid bilayer membrane vesicles measuring 30-150 nm with significant biological roles [11]. Thus, interest in exosomes’ potential to stop the onset of autoimmune disorders, including IBD, is growing [11]. Exosomes are abundant in the milieu of inflammation and are released from immune cells (macrophages, neutrophils, and dendritic cells, etc.), mesenchymal stem cells (MSCs), and platelets as intercellular messengers, which contribute to the control of inflammation through the control of gene expression and the release of anti-inflammatory substances [12]. Additionally, MSC-derived exosomes have been found to exhibit therapeutic benefits for IBD by polarizing M1 macrophages into M2 macrophages, reducing inflammatory reactions, and preserving the integrity of the intestinal barrier [13].
In this review, we summarize the biogenesis, biochemical components, sources of exosomes, and the role of exosomes in regulating macrophages to repair IBD. This review will also cover other compounds regulating macrophages and the role of EVs/nanovesicles in modulating macrophages toward IBD mitigation.
Materials and methods
PubMed and Google Scholar databases were utilized to look for journals mostly concerned with exosomes and exosome-like nanovesicles, macrophages, and IBD. Articles were retrieved up to 2023 using keywords, including “exosome, macrophage, and IBD”. After entering these keywords into PubMed and Google Scholar, a review process was conducted to ensure the relevance and eligibility of the journals selected.
Exosome and its sources
Exosomes are microscopic, extracellular vesicles that contain proteins, lipids, nucleic acids, and other bioactive materials that are involved in both normal and pathological activities within the body [14] and are present in different bodily fluids, including amniotic fluid [15], blood plasma [16], saliva [17], breast milk [18], serum [17,19,20], ascites [21], urine [15], nasal secretions [22], and cerebrospinal fluid [23]. Exosome-like vesicles (ELVs) are crucial for intercellular communication by serving as cellular transfer vehicles for biomolecules, and this distinctive quality justifies their use as bio-inspired medication delivery devices [24]. Exosomes have the ability to change the biological activity of recipient cells by transferring their cargo inside these cells [25].
Exosomes are secreted by a variety of cells, including bone marrow MSCs [26,27], umbilical cord MSCs [28-33], adipose tissue-derived MSCs [34-37], neutrophils [38], dendritic cells (DCs) [39], amniotic fluid stem cells [40], ginger [41], tumors [42], mast cells [43], plasma platelets [44], T lymphocytes [45], mantle cell lymphoma [46], macrophage (M1) [47,48], M2 macrophage [49], and bovine colostrum [50]. Exosomes are appealing in biotechnology and biomedical research. After all, they can serve as both therapeutic agents and disease biomarkers because they share components with their parent cells [51]. Therefore, it is necessary to understand the exosome’s biogenesis and biochemical makeup to determine the characteristics that cause it to operate as it does.
Biogenesis of exosome
Most mammalian cell types manufacture exosomes and extracellular vesicles encased in membranes that originate in the endosomal space before release [52]. The final content of exosomes is produced as a result of interactions between endosomes, where they first develop, and then transfer into other intracellular vesicles and organelles [53]. Exosomes originate in multivesicular compartments within eukaryotic cells and are released during the fusion of these compartments with the plasma membrane [54]. Through the process of exocytosis, cells produce exosomes, which are then absorbed by target cells and have the ability to transmit biological information across nearby or distant cells [55]. The intraluminal vesicles of multivesicular endosomes (MVEs) are released as exosomes into the extracellular milieu or segregated into lysosomes for cargo destruction [56] (Figure 1).
Figure 1.
Biogenesis of exosome. Intraluminal vesicles from MVBs release exosomes into the extracellular space. Exosomes move to the target cells via endocytosis, direct fusion, or binding to receptors. Abbreviation: MVBs, multivesicular bodies.
Exosome biogenesis, secretion, and release have generally been demonstrated to be influenced by a number of processes. Insights into the generation and operation of extracellular vesicles may, therefore, be gained from research on the endosomal sorting complex required for transport (ESCRT) machinery [57]; hence, MVB production depends heavily on the elements of the ESCRT pathway [58]. Nonetheless, it has been demonstrated that gene silencing for two ESCRT-0 elements (HRS, STAM1), one of ESCRT-I (TSG101), as well as a late-acting component (VPS4B), causes regular variations in the secretion of exosomes [59]. The ESCRT machinery is essential for the synthesis of MVEs, allowing cargo choice and intraluminal vesicle (ILV) budding [60]. Additionally, it has been discovered that heparanase modulates the syndecan-syntenin-ALIX pathway, promoting the budding of endosomal membranes and the exosomes’ biogenesis by cutting the syndecans’ heparan sulfate chains [61]. This mechanism regulates the choice of a particular exosome cargo [61]. Although research has demonstrated that the exosomal protein syntenin promotes the formation of exosomes, uncovered information indicates that the GTPase ADP ribosylation factor 6 (ARF6) and its effector phospholipase D2 (PLD2) also influence exosomes by regulating the budding of ILVs into MVBs [62]. Besides, caveolin-1 (Cav1), which controls the amount of cholesterol in the endosomal compartment/MVBs, controls exosome biogenesis and exosomal protein cargo sorting [63]. Similarly, curcumin activates ceramide synthesis, which increases intracellular ceramide-dihydroceramide concentration, leading to excessive ceramide and a tenfold increase in exosome/microvesicle secretion [64]. Moreover, the mechanistic target of rapamycin complex 1 (mTORC1) has been demonstrated to be a negative regulator of exosome release, which results in a net loss of cellular membrane and protein content [65]. However, ISGylation is a new ubiquitin-like modification that regulates the generation of exosomes [66]. ISGylation of the MVB protein TSG101 causes it to aggregate and degrade, which is enough to prevent exosome secretion [66].
Biochemical components of exosome
Exosomes’ molecular makeup reflects the fact that they originated in endosomes as ILVs [67]. Their varied components, which can represent their cell of origin, include nucleic acids, proteins, lipids, amino acids, and metabolites [53]. Moreover, exosomes contain proteins, miRNAs, and messenger RNAs (mRNAs) (exosome shuttle RNA, or esRNA), which may provide a new platform for diagnostics [68]. In general, several of these biochemical components of exosomes have been recognized as indicators for therapeutic and diagnostic approaches to some diseases. This may be because of where they came from biologically and what was in their cargo, which explains why they are helpful as biomarkers for diseases, including IBD. Generally, irrespective of where the exosome is being produced and secreted, it has been shown that exosomes may carry nucleic acids, proteins, and lipids (Figure 2). Each element is detailed below.
Figure 2.
The chemical makeup of exosomes. An exosome (zoomed) is released from a source cell, showing the various biochemical components that are being transferred to the target cell. Abbreviations: ARF, ADP ribosylation factor 6; ESCRT, endosomal sorting complex needed for transport; LAMP, lysosomal associated membrane protein; MHC, major histocompatibility complex; mRNA, messenger RNA; miRNA, microRNA.
Proteins
The identification of exosomal marker proteins such as CD24, CD9, annexin-1, and heat shock protein (HSP) 70 has been made, and these proteins also showed the proper buoyant density and antigen orientation [68]. Meanwhile, recent investigations have discovered that MSC exosomal surface marker proteins obtained from the human umbilical cord include CD9, CD63, CD81, HSP 70, Alix, and tumor susceptibility gene 101 (TSG101) without calnexin [29,30,69-71]. Additional marker proteins that are frequently seen in the exosomes from β-cells include tetraspanin proteins (CD63, CD82, and CD81), lysosomal associated membrane protein (LAMP)-1 and LAMP-2, intercellular adhesion molecule-1 (ICAM-1), flottilin-1, G-proteins, Alix, TSG101, and Ras-associated binding (Rab) proteins [72]. Meanwhile, TSG101, CD81, and syntenin have been similarly discovered to have a relative expression in exosome subpopulations [73]. Interestingly, Zhang and colleagues have identified two exosome subpopulations. Large exosome vesicles (Exo-L) were seen with high amounts of proteins such as annexins, charged multivesicular body proteins 1A/2A/4B/5, vacuolar protein-sorting 4 homolog B, DnaJ heat shock protein family (Hsp40) member A1, and myosin IC, while small exosome vesicles (Exo-S) were enriched with flotillin 1, flotillin 2, tweety family member 3, tetraspanin 14, and ESCRT-I subunit VPS37B [74]. These different exosome subpopulations might be a result of different intracellular compartments engaging in exosome loading, according to the existence of genomic DNA (gDNA) [75].
DNA
Exosomes deliver substances with functional activity to aid in intercellular communication [76]. Although the regulatory mechanism is unknown, cancer cells produce exosomes that contain substantial quantities of DNA and can modify the expression of an oncogene in a recipient cell [76]. According to Torralba and team, T cells activate DCs by transferring exosomal DNA, demonstrating a particular function for antigen-dependent interactions in protecting DCs from pathogen infection [77]. Besides, nuclear DNA builds up in the cytoplasm when the secretion of exosomes is inhibited, resulting in the cytoplasmic DNA sensing apparatus being activated [78]. This triggers the innate immune reaction, resulting in a DNA damage response that is dependent on reactive oxygen species (ROS) and thus produces apoptosis, or a cell cycle arrest resembling senescence in normal human cells [78]. According to a report from Takahashi and his team, chromosomal DNA fragments of different lengths can be found in exosomes, demonstrating that exosome secretion keeps cells in a state of equilibrium by eliminating dangerous cytoplasmic DNA [78]. Moreover, murine colitis and active human CD both showed a substantial rise in exosomal double-strand DNA (dsDNA) levels, including mitochondrial (mt) DNA and nuclear genomic DNA (nDNA) [79]. This further corroborates the idea that exosomes may contain DNA.
RNA
Exosomes have a notable enrichment of certain protein and RNA cargoes [80], and exosomes produced by metazoan cells can deliver specific membrane proteins and short RNAs to their target cells to regulate cell migration, development, and metastasis [81]. In different eukaryotes, exosomes have been found to transport RNA cargo, such as short non-coding RNAs and mRNAs, which can change the phenotype of the recipient cell [82].
As components of exosomes, RNAs are transported to exosomes by RNA-binding proteins (RBPs). The synaptotagmin-binding cytoplasmic RNA-interacting protein (Syncrip), also referred to as the heterogeneous nuclear ribonucleoprotein (hnRNP) Q, is a highly conserved RBP that regulates the exosomal partition of a group of miRNAs [83]. Additionally, the RBP Y-box protein I (YBX1) binds to microRNA (miR)-223 and is crucial for the secretion of miRNAs in exosomes by HEK293T cells [81]. The short sequence motif found in miRNAs binds to the RBP fragile X mental retardation 1 (FMR1) and directs the loading of miRNA into exosomes via interactions with elements of the ESCRT pathway [84]. Furthermore, hnRNPA2B1 packages circRNA, specifically circNEIL3, into exosomes in gliomas and transfers it to tumor-associated macrophages that have been infiltrated [85]. Interestingly, without CAVIN1, non-caveolar caveolin-1 (CAV1) promotes hnRPNK localization to MVBs, recruiting miRNAs with the AsUGnA motif and inducing their discharge within exosomes [86].
Exosome components called RNAs are increasingly being used as diagnostic markers in diseases. Extensive research has been done recently on the non-coding RNA (ncRNA) found in exosomes about categorization, localization, and possible use as biomarkers [87]. Exosomal long non-coding RNA (lncRNA) of HDAC4 (LOC85009) reduces docetaxel (DTX) resistance by controlling autophagy-related 5 (ATG5)-induced autophagy via the ubiquitin-specific proteinase 5 (USP5)/upstream transcription factor 1 (USF1) axis, putting LOC85009 forth as a possible target to overcome DTX resistance in lung adenocarcinoma treatment [88]. Additionally, four brand new transfer (t) RNA-derived small RNAs (tsRNAs) in plasma exosomes, such as tRNA-ValTAC-3, tRNA-GlyTCC-5, tRNA-ValAAC-5, and tRNA-GluCTC-5, have been discovered in individuals with liver cancer, establishing the potential of plasma exosomal tsRNA as a new diagnostic biomarker [89].
Generally, it has been discovered that exosomes contain RNA, lncRNA, miRNA/miR, and tsRNA, some of which may serve as illness diagnostic indicators.
Lipids
Exosome production and release into the external environment depend heavily on lipids and their structural involvement in exosomal membranes [90]. It has been discovered that exosome membranes contain more sphingomyelin, lysophosphatidylcholine, and phosphatidic acid than parental cells do, and interestingly, exosomes have been shown to contain an unusual phospholipid called bis (monoacylglycero) phosphate as a particular lipid indicator for exosomes [91]. Additionally, Llorente and his team found high levels of phosphatidylserine, sphingomyelin, cholesterol, and glycosphingolipids in exosomes [92]. It is also found that glycerolipid, glycerophospholipid, sphingolipid, and glycosphingolipid are contained in the exosomes of prostate cancer cells, even though the class of lipids found in the exosomes with the highest abundance is glycerophospholipid [93]. Similarly, sphingomyelins, phosphatidylinositols, and sulfatides have been found in exosomes of lipids and plasma, but exosome composition has been observed to differ in terms of lipids from plasma in areas of triacylglycerols, diacylglycerols, phosphatidylcholines, and lysophosphatidylcholines [94]. Besides, Elmallah and colleagues found phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, and hexosylceramide (HexCer) to be increased in the exosomes of non-metastatic cells and patients with cancer relative to control cells and healthy donors [95].
Exosomes and exosome-like nanovesicles in macrophage modulation toward IBD repair
Stem/stromal cell-derived exosomes
It is documented that exosome (from human umbilical cord MSCs) therapy in IBD mice reduces macrophage infiltration into the colon tissues [96]. The exosome-treated mice’s colon tissues and spleens expressed more IL-10 but less inducible nitric oxide synthase (iNOS) and IL-7 [96]. Additionally, human umbilical cord (huc) MSC-secreted exosomes have been found to relieve colitis by inhibiting casp11/4-induced macrophage pyroptosis [31]. Mechanistically, the exosomes express miR-203a-3p.2 to inhibit casp4-induced macrophage pyroptosis in an inflammatory environment [31]. Another study revealed that hucMSC-derived exosomes suppress the release of IL-1β from mouse peritoneal macrophages even when NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasomes are activated [29]. Moreover, NLRP3, an apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), caspase-1, IL-18, and IL-1β have substantially lower relative expressions in macrophages with hucMSC-derived exosomes [29]. Furthermore, exosomal miR-216a-5p produced by hypoxia-prime adipose-derived stem cells (ASCs) has shown more therapeutic effectiveness than those under normoxia (NExos) in the treatment of experimental colitis by enhancing the M2 macrophage phenotype [97].
According to other studies, intestinal epithelial cells and macrophages can internalize layer-by-layer (LbL)-exosomes from MSC to produce anti-inflammatory and tissue-repair effects [98]. Treatment with LbL-exosome can control inflammation by decreasing M1 macrophages and increasing M2 macrophages [98]. Moreover, Xu and team revealed that tumor necrosis factor α- (TNF-α-) pretreated MSCs generated from human menstrual blood (MenSC)-derived small EVs (MenSCs-sEVTNF-α) reduces colonic inflammation, accompanied by the elevation of miR-24-3p in small EVs derived from MenSCs and the polarization of M2 macrophages in the colon [99]. Besides, Cao and colleagues found that bone marrow MSC-derived EVs (BMSC-derived EVs) encourage M2-like macrophage polarization, as evidenced by a rise in the M2 marker CD163 in DSS-induced UC [100]. Also, the systemic administration of exosomes from human bone marrow-derived mesenchymal stromal cells (MSC-Exos) significantly lessens colitis in a variety of IBD models, reduces inflammatory responses, maintains the gut barrier’s integrity, and polarizes M2b macrophages [101].
Intestinal epithelial cell (IEC)-derived exosome
The intestinal epithelium is essential for maintaining gut homeostasis because it serves as a physical barrier, a center of coordination for immune defense, and a medium for communication between immune cells and bacteria [102]. IEC exosomes ameliorate IEC damage caused by glucose-oxygen depletion in vitro in addition to the severity of intestinal damage following an intestinal ischemia/reperfusion attack in vivo [103]. Moreover, exosomes from IECs and their cargo, including miRNAs, have been found to control the expression of molecules that promote inflammation in inflammatory gut tissues during sepsis [104]. Appiah and team revealed that proinflammatory cytokines IL-17A and TNF-α messaging are significantly reduced after septic-EV injection into an inflamed gut [105]. Additionally, IEC-derived luminal EVs contain miRNAs that reduce proinflammatory reactions [105]. When an organ is infected or inflamed, macrophages first adopt the M1 phenotype and release TNF-α, IL-1β, IL-23, and IL-12 in response to the stimuli; however, tissue injury may result if the M1 phase lasts longer [106]. Consequently, M2 macrophages release large volumes of transforming growth factor beta (TGF-β) and IL-10 to reduce inflammation, support tissue repair, remodeling, and vasculogenesis, and maintain homeostasis [106]. Hence, M1 macrophages may be activated to release TNF-α; however, IEC exosomes or EVs may activate M2 macrophages to reduce TNF-α as well as damage to the gut.
Macrophage-derived exosome
It’s interesting to note that M2 macrophages release exosomes that inhibit IBD. For instance, Deng and team revealed that exosomal miR-590-3p produced by M2 macrophages targets large tumor suppressor homology Ser/Thr kinase 1 (LATS1) and activates Yes-associated protein (YAP)/β-catenin-regulated transcription suppresses cytokines that promote inflammation (including IL-6, TNF-α, and IL-1β), and encourages epithelial regeneration [49]. Moreover, Yang and colleagues showed that M2b macrophage exosomes protect against colitis caused by DSS, primarily controlled by the CC chemokine 1 (CCL1)/CC-chemokine receptor (CCR) 8 pathway, presenting a fresh method for treating IBD [11]. While M2 macrophages release a variety of anti-inflammatory mediators, such as TGF-β, IL-10, CCL1, CCL17, CCL18, and CCL22, M1 macrophages discharge proinflammatory substances such as TNF-α, IL-1α, IL-1β, IL-6, C-X-C motif chemokine ligand 9 (CXCL9), and CXCL10 [107]. Hence, these may imply that M2 exosomes may polarize M2 macrophages to repair IBD while suppressing M1 macrophages.
Other immune cells-derived exosome
Exosomes derived from other immune cells, such as dendritic cells (DCs) and granulocytic myeloid-derived suppressor cells (MDSCs), may play roles in mitigating gut inflammation. It has been found that exosomes generated from immature dendritic cells (IDCs) reduce the early systemic inflammatory response in sepsis by promoting the clearance of apoptotic cells through milk fat globule EGF factor VIII (MFGE8) [108]. Additionally, the TNF-α response elicited by cecal ligation and puncture (CLP) is suppressed by exosomes derived from IDC by 46%; however, in septic rats, mature DC-derived exosomes did not affect TNF-α levels [108]. Similarly, exosomes generated from bone marrow DCs (BMDCs) (containing MFG-E8) increase the removal of apoptotic cells, lower the proinflammatory response, and increase the lifespan of experimental mice with sepsis [109]. The administration of BMDC-derived exosomes decreased TNF-α and IL-6 plasma levels while increasing survival from 44% to 81% [109]. Rats treated with exosomes had peritoneal macrophages that were 2.8 times more capable of phagocytosing apoptotic thymocytes [109]. Again, it has been discovered that the spontaneous improvement of colitis is partially reversed when arginase (Arg)-1 activity in granulocytic (G)-myeloid-derived suppressor cells (MDSC) exosome is inhibited [110]. Arg-1 activity has also been linked to the G-MDSC exosome’s capacity to inhibit the delayed-type hypersensitivity (DTH) response, reduce the growth of CD4+ T cells, and suppress the secretion of IFN-γ in vitro [110]. Besides, G-MDSC exosome-treated colitis animals showed reduced TNF-α and IFN-γ blood levels [110], implying that the G-MDSC exosome may activate M2 macrophages to reduce colitis. Surprisingly, one indicator of the M2 anti-inflammatory subgroup is Arg1 [111].
Food-derived exosomes/exosome-like nanovesicles
It has been discovered that turmeric-derived nanovesicles (TNVs) encourage the conversion of M1 phenotype macrophages to M2 macrophages, regulate the gut microbiota, and repair the damaged intestinal epithelial barrier, thus exerting anti-colitis efficacy [112]. Additionally, Kim and colleagues found that ginseng-derived exosome-like nanoparticles (GENs) have the potential to both prevent and reduce inflammatory responses by downregulating proinflammatory cytokines in proinflammatory macrophages and promoting the production of anti-inflammatory macrophages [113]. Spermine, spermidine, and putrescine are polyamines with antioxidant properties, playing a crucial role in biological processes like cell proliferation and differentiation; hence, dietary polyamines have a substantial effect on human health, especially on the development and differentiation of the immune system and intestinal maturation [114]. Interestingly, in the inflammatory colon, spermidine has been shown to downregulate M1 markers while upregulating M2 macrophage markers, as well as decrease the colon’s activation of T cells and F4/80 macrophages, mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) phosphorylation, and the production of proinflammatory cytokines [115].
Additionally, Han and his team investigated colostrum-derived exosomes (Col-exo) in DSS-induced colitis and found that Col-exo stimulates macrophages and intestinal epithelial cells and produces an environment that reduces inflammation by successfully eliminating reactive oxygen species and regulating immune cytokine expression [50]. Milk-derived extracellular vesicles (mEVs) enhanced with immunomodulatory proteins and miRNAs prevent the generation of cytokines and the polarization of macrophages toward a proinflammatory phenotype [116]. According to these results, milk and other dairy products include edible nanovesicles (EVs) that have inherent immunomodulatory properties that are good for the gastrointestinal tract [116].
Other exosomes/EVs
It has been found that Trichinella spiralis-EVs (Ts-EVs) promote the infiltration of alternatively activated (M2) macrophages into the colon and prevent M1 macrophage polarization as a result of their immunomodulatory capabilities [117]. Moreover, the murine counterpart of circRNA SCAR (steatohepatitis-associated circRNA ATP5B regulator), referred to as circRNA mSCAR, declines in septic mice’s macrophages, and this is consistent with the M1 polarization being overly intense [118]. However, it has been demonstrated that the exosome-based method of delivering circRNA mSCAR into mitochondria preferentially targets macrophage mitochondria, promoting the M2 subtype of macrophage polarization, reducing systemic inflammation, and reducing septic mouse mortality [118].
Generally, irrespective of the source from which the exosome is being produced, exosomes can activate M2 subtype macrophages, which causes an anti-inflammatory impact that promotes IBD repair (Figure 3).
Figure 3.
Exosome, exosomal miRNA, and macrophage pathways in IBD repair. Exosomes, including their miRNA, polarize macrophages to M2/activate M2 macrophages. This leads to the production of anti-inflammatory cytokines to repair IBD. Abbreviations: MΦ, inactive macrophage; miRNA, microRNA.
Role of other compounds regulating macrophages towards IBD repair
Wu and colleagues have shown heme supplementation to have a protective impact on the colon tissue microenvironment in mice with DSS-induced colitis via controlling the polarization of macrophages in a heme oxygenase-1 (HO)-dependent and HO-1-independent manner [119]. Moreover, baicalin has been demonstrated to reduce the manifestations of DSS-induced colitis by modifying the polarization of macrophages toward the M2 phenotype [120]. Additionally, lentivirus-mediated short hairpin RNA (shRNA) interference with interferon regulatory factor 5 (IRF5) expression causes a transition in the phenotype of rat peritoneal macrophages from M1 to M2 and decreases IL-1β and TNF-α expression more in depressed colitis rats than in nondepressed colitis rats [121]. Besides, Lu and colleagues found that thalidomide might boost epithelial cells’ potential for self-renewal and alter M1/M2 polarization by reducing M1 protein signatures CD86 and CC-chemokine receptor 7 (CCR7) and raising M2 protein signatures CD206 and Arg-1, decreasing the occurrence and development of carcinogenesis linked to colitis compared to mice used as negative controls [122]. Again, lupeol reduces the symptoms of experimental IBD by, at least partially, suppressing M1 and encouraging M2 macrophages [8].
Furthermore, Qingchang Wenzhong decoction (QCWZD) has been found by Lu and team to reduce colonic shortening and mucosal damage by suppressing M1 macrophage polarization and the production of accompanying cytokines, such as IL-6 and TNF-α, in vivo and in vitro [123]. Similarly, it has been discovered that M1 macrophage-induced Caco-2 cell death was reduced by toxoROP16I/III-induced M2 macrophages [124]. Importantly, the sEVs of tyrosine-protein phosphatase non-receptor type 1 (Ptpn1)-knockdown macrophages have been reported to be significantly enriched in lactadherin, and recombinant lactadherin therapy reduces intestinal inflammatory response and barrier failure by promoting macrophage M2 polarization [125]. In another study, Song and the team found that both high and low electroacupuncture increased M2 macrophage percentages and lowered M1 macrophages in DSS mice [126]. Liu and the team discovered that berberine regulates the M1 polarization of macrophages in DSS-induced colitis through the AKT serine/threonine kinase 1 (AKT1)/suppressors of cytokine signaling (SOCS) 1/NF-κB signaling pathway [127]. It dramatically lowers the proportion of M1 macrophages [127]. It has also been discovered that phosphatidylmannoside (PtdMan) reduces the levels of proinflammatory cytokines in the colons of RAW264.7 cells and DSS-colitis mice, and it also regulates M1/M2 polarization via activating peroxisome proliferator-activated receptor delta (PPARγ) [128].
The dual role of exosomes in macrophage modulation in IBD
Exosomes produced by healthy cells have favorable benefits; nevertheless, exosomes produced by pathogenic cells, such as cancer cells or infected cells, may have adverse impacts on health [129]. Thus, exosomes play a double role in the regulation of macrophages in IBD. Generally, exosomes derived from healthy stem and stromal cells, food, immune cells, and other systems or pathogens regulate macrophages toward IBD repair (Table 1); however, those derived from inflammatory-driven cells and unhealthy cells aggravate IBD and its progression.
Table 1.
Sources of exosomes or exosome-like nanovesicles modulating macrophages toward IBD repair
Exosomes/exosome-like nanovesicles source | Condition | Mechanism | Reference |
---|---|---|---|
Stem/stromal cell | |||
Hypoxia-prime adipose-derived stem cells | DSS-induced colitis | Control the HMGB1/TLR4/NF-κB signaling pathway to cause macrophage M2 polarization. | [97] |
Human umbilical cord MSCs | DSS-induced colitis | Prevent macrophage pyroptosis brought on by casp4 in an inflammatory setting. | [31] |
Human bone marrow-derived MSCs | DSS/TNBS induced colitis | Reduce inflammatory reactions, protects intestinal barrier integrity, polarizes M2b macrophages, and stimulates the production of IL-10. Primarily affects colonic macrophages. | [101] |
Human umbilical cord MSCs | DSS-induced colitis | Enhance the expression of the IL-10 gene and decreases the infiltration of macrophages, TNF-α, IL-1β, IL-6, iNOS, and IL-7 genes. | [96] |
Human umbilical cord MSCs | DSS-induced colitis | Suppress the release of IL-1β from mouse peritoneal macrophages and considerably lowers the relative expression of NLRP3, ASC, caspase-1, IL-18, and IL-1β in macrophages. | [29] |
LbL-MSCs | UC | Increase M2 macrophages to control inflammation while decreasing M1 macrophages. | [98] |
Human menstrual blood | DSS-induced colitis | Reduce colonic inflammation and polarize M2 macrophages in the colon. | [99] |
Bone marrow MSC | DSS-induced UC | Encourage the polarization of M2-like macrophages, which is indicated by a rise in the M2 marker CD163. Upregulate TGF-β and IL-10 and downregulate TNF-α, CCL-24, TNF-α, VEGF-A, and IFN-γ levels. | [100] |
Macrophage | |||
M2 | DSS-induced colitis | MiR-590-3p, highly concentrated in M2 exosomes, suppresses the production of proinflammatory cytokines such as IL-1β, IL-6, and tumor necrosis factor-α. | [49] |
M2 | DSS-induced colitis | Inhibit IL-1β, IL-6, and IL-17A. Offer protection against colitis produced by DSS, mostly through the CC chemokine 1 (CCL1)/CCR8 axis. | [11] |
Food | |||
Turmeric | Colitis | It increase the M1 phenotype’s transition to M2 macrophages and restores the impaired gut epithelial barrier. | [112] |
Ginseng | DSS-induced colitis | Stimulate the synthesis of anti-inflammatory macrophages while downregulating proinflammatory cytokines in proinflammatory macrophages. | [113] |
Spermidine | DSS-induced colitis | Increase the markers for M2 macrophages while decreasing the markers for M1 macrophages in the inflammatory colons. | [115] |
Colostrum | DSS-induced UC | Stimulate the growth of macrophages and colonic epithelial cells, reduce inflammation by efficiently eliminating ROS, and control the release of immune cytokines. | [50] |
Milk | DSS-induced UC | Reduce the inflammatory response by blocking the activation of the NLRP3 inflammasome and the TLR4-NF-κB signaling pathway. Stop the release of cytokines and macrophage polarization toward a proinflammatory phenotype. | [116] |
Other exosomes/EVs | |||
Trichinella spiralis | DSS-induced colitis | Activate M2 macrophages in the colon and prevent M1 macrophage polarization. | [117] |
HEK293T cells | Sepsis-induced intestinal damage | Minimize systemic inflammation and mortality while promoting M2 polarization by reducing mitochondrial reactive oxygen species (mtROS). | [118] |
Other immune cells | |||
G-MDSC | DSS-induced colitis | The G-MDSC exosome (containing Arg-1) inhibits the delayed-type hypersensitivity response, reduces the growth of CD4+ T cells, and decreases IFN-γ and TNF-α levels. | [110] |
Abbreviations: ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; DSS, dextran sulfate sodium; G, granulocytic; HMGB1, high mobility group box 1; IFN-γ, interferon-gamma; IL, interleukin; ILVs, intraluminal vesicles; iNOS, inducible nitric oxide synthase; M, macrophage; MDSC, myeloid-derived suppressor cells; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3, NOD-like receptor family, pyrin domain-containing 3; ROS, reactive oxygen species; TGF-β, transforming growth factor beta; TLR4, Toll-like receptor 4; TNBS, 2,4,6-trinitrobenzenesulfonic acid solution; TNF, tumor necrosis factor; VEGF, vascular endothelial growth factor.
Exosomes and their microRNA, driven by inflammatory stimuli, immune cells, and disease cells, regulate macrophages to support IBD and its progression. For instance, exosomal miR-93-5p secreted by granulocytic myeloid-derived suppressor cells (G-MDSC) in response to IL-6 encourages the development of monocytic MDSCs (M-MDSC) into M2 macrophages and involves a signal transducer and activator of transcription 3 (STAT3) signaling route that promotes the progression from colitis to malignancy [130]. Besides, treatment of RAW264.7 macrophages with serum exosomes derived from mice induced by DSS encourages phosphorylation of p38 and extracellular signal-regulated kinase (ERK) as well as the generation of TNFα [131]. Additionally, Gong and colleagues have demonstrated that serum exosomes (from CD) may circulate into the intestine’s mucosa and greatly exacerbate colitis by controlling macrophage activation and the function of the epithelial barrier [132]. Moreover, CD14+ Mφ contributes to CD pathogenesis by causing activation-induced cell death (AICD) opposition in CD4+ T cells by releasing TNF in the exosomal membrane and activating TNFR2/NF-κB signaling [133].
Conclusion and perspective
IBD, which includes both UC and CD, is a complex illness with an unknown cause. Immune cells involved in IBD, such as macrophages, emit cytokines and chemokines that promote inflammation and compromise intestinal barrier integrity. It has been discovered that M1 macrophages exacerbate IBD, while M2 macrophages mitigate the disease. Exosomes, as ubiquitous conveyors of intercellular messages, are involved in immune mechanisms and immune-mediated disease processes. Additionally, exosomes derived from healthy stem and stromal cells, food, immune cells, parasites, and other artificial systems have been shown to regulate macrophages toward IBD repair. Moreover, other compounds have also been found to regulate macrophages toward IBD repair. However, exosomes from inflammatory-driven and disease cells promote M1 macrophages to perpetuate IBD. Therefore, in the future, more studies should be done on immune cell-derived exosomes and their modulation of macrophages to repair IBD. Also, immune cell-derived exosomes other than macrophages should be explored to determine the mechanism that regulates macrophages toward IBD repair.
Acknowledgements
This study was supported by the National Natural Science Fund of China (Grant No. 82250410378), the 2022 Jiangsu Excellent postdoctoral program (Grant No. 2022ZB634), the open project of the clinical medical research center of Gynecology and Traditional Chinese Medicine of Zhenjiang (Grant No. SS202204-KFC01), Zhenjiang key research and development plan (social development) (Grant Nos. SH2022062, SH2022091 and SH2023050), College student practice innovation training program, Orientation projects, Jiangsu Province (Grant No. 202310299119Y) and Shangqiu Medical College open project in 2023 (project No. KFKT23005).
Disclosure of conflict of interest
None.
References
- 1.Yang L, Wu G, Wu Q, Peng L, Yuan L. METTL3 overexpression aggravates LPS-induced cellular inflammation in mouse intestinal epithelial cells and DSS-induced IBD in mice. Cell Death Discov. 2022;8:62. doi: 10.1038/s41420-022-00849-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Park J, Cheon JH. Incidence and prevalence of inflammatory bowel disease across Asia. Yonsei Med J. 2021;62:99–108. doi: 10.3349/ymj.2021.62.2.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kaplan GG, Ng SC. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology. 2017;152:313–321. e312. doi: 10.1053/j.gastro.2016.10.020. [DOI] [PubMed] [Google Scholar]
- 4.Liu Z, Wang H. Probiotics alleviate inflammatory bowel disease in mice by regulating intestinal microorganisms-bile acid-NLRP3 inflammasome pathway. Acta Biochim Pol. 2021;68:687–693. doi: 10.18388/abp.2020_5597. [DOI] [PubMed] [Google Scholar]
- 5.Pan X, Zhu Q, Pan LL, Sun J. Macrophage immunometabolism in inflammatory bowel diseases: from pathogenesis to therapy. Pharmacol Ther. 2022;238:108176. doi: 10.1016/j.pharmthera.2022.108176. [DOI] [PubMed] [Google Scholar]
- 6.Castro-Dopico T, Fleming A, Dennison TW, Ferdinand JR, Harcourt K, Stewart BJ, Cader Z, Tuong ZK, Jing C, Lok LSC, Mathews RJ, Portet A, Kaser A, Clare S, Clatworthy MR. GM-CSF calibrates macrophage defense and wound healing programs during intestinal infection and inflammation. Cell Rep. 2020;32:107857. doi: 10.1016/j.celrep.2020.107857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Seyedizade SS, Afshari K, Bayat S, Rahmani F, Momtaz S, Rezaei N, Abdolghaffari AH. Current status of M1 and M2 macrophages pathway as drug targets for inflammatory bowel disease. Arch Immunol Ther Exp (Warsz) 2020;68:10. doi: 10.1007/s00005-020-00576-4. [DOI] [PubMed] [Google Scholar]
- 8.Zhu Y, Li X, Chen J, Chen T, Shi Z, Lei M, Zhang Y, Bai P, Li Y, Fei X. The pentacyclic triterpene Lupeol switches M1 macrophages to M2 and ameliorates experimental inflammatory bowel disease. Int Immunopharmacol. 2016;30:74–84. doi: 10.1016/j.intimp.2015.11.031. [DOI] [PubMed] [Google Scholar]
- 9.Liu X, Ren X, Zhou L, Liu K, Deng L, Qing Q, Li J, Zhi F, Li M. Tollip orchestrates macrophage polarization to alleviate intestinal mucosal inflammation. J Crohns Colitis. 2022;16:1151–1167. doi: 10.1093/ecco-jcc/jjac019. [DOI] [PubMed] [Google Scholar]
- 10.Rayamajhi S, Nguyen TDT, Marasini R, Aryal S. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater. 2019;94:482–494. doi: 10.1016/j.actbio.2019.05.054. [DOI] [PubMed] [Google Scholar]
- 11.Yang R, Liao Y, Wang L, He P, Hu Y, Yuan D, Wu Z, Sun X. Exosomes derived from M2b macrophages attenuate DSS-induced colitis. Front Immunol. 2019;10:2346. doi: 10.3389/fimmu.2019.02346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wang C, Xu M, Fan Q, Li C, Zhou X. Therapeutic potential of exosome-based personalized delivery platform in chronic inflammatory diseases. Asian J Pharm Sci. 2023;18:100772. doi: 10.1016/j.ajps.2022.100772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Song JY, Kang HJ, Hong JS, Kim CJ, Shim JY, Lee CW, Choi J. Umbilical cord-derived mesenchymal stem cell extracts reduce colitis in mice by re-polarizing intestinal macrophages. Sci Rep. 2017;7:9412. doi: 10.1038/s41598-017-09827-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int J Nanomedicine. 2020;15:6917–6934. doi: 10.2147/IJN.S264498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Keller S, Rupp C, Stoeck A, Runz S, Fogel M, Lugert S, Hager HD, Abdel-Bakky MS, Gutwein P, Altevogt P. CD24 is a marker of exosomes secreted into urine and amniotic fluid. Kidney Int. 2007;72:1095–1102. doi: 10.1038/sj.ki.5002486. [DOI] [PubMed] [Google Scholar]
- 16.Baranyai T, Herczeg K, Onódi Z, Voszka I, Módos K, Marton N, Nagy G, Mäger I, Wood MJ, El Andaloussi S, Pálinkás Z, Kumar V, Nagy P, Kittel Á, Buzás EI, Ferdinandy P, Giricz Z. Isolation of exosomes from blood plasma: qualitative and quantitative comparison of ultracentrifugation and size exclusion chromatography methods. PLoS One. 2015;10:e0145686. doi: 10.1371/journal.pone.0145686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 2012;7:e30679. doi: 10.1371/journal.pone.0030679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Admyre C, Johansson SM, Qazi KR, Filén JJ, Lahesmaa R, Norman M, Neve EP, Scheynius A, Gabrielsson S. Exosomes with immune modulatory features are present in human breast milk. J Immunol. 2007;179:1969–1978. doi: 10.4049/jimmunol.179.3.1969. [DOI] [PubMed] [Google Scholar]
- 19.Almqvist N, Lönnqvist A, Hultkrantz S, Rask C, Telemo E. Serum-derived exosomes from antigen-fed mice prevent allergic sensitization in a model of allergic asthma. Immunology. 2008;125:21–27. doi: 10.1111/j.1365-2567.2008.02812.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shao J, Jin Y, Shao C, Fan H, Wang X, Yang G. Serum exosomal pregnancy zone protein as a promising biomarker in inflammatory bowel disease. Cell Mol Biol Lett. 2021;26:36. doi: 10.1186/s11658-021-00280-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Navabi H, Croston D, Hobot J, Clayton A, Zitvogel L, Jasani B, Bailey-Wood R, Wilson K, Tabi Z, Mason MD, Adams M. Preparation of human ovarian cancer ascites-derived exosomes for a clinical trial. Blood Cells Mol Dis. 2005;35:149–152. doi: 10.1016/j.bcmd.2005.06.008. [DOI] [PubMed] [Google Scholar]
- 22.Qiu S, Duan X, Geng X, Xie J, Gao H. Antigen-specific activities of CD8+ T cells in the nasal mucosa of patients with nasal allergy. Asian Pac J Allergy Immunol. 2012;30:107–113. [PubMed] [Google Scholar]
- 23.Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, Chalmers RT, Webb DJ, Dear JW. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012;10:5. doi: 10.1186/1479-5876-10-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Stremersch S, Vandenbroucke RE, Van Wonterghem E, Hendrix A, De Smedt SC, Raemdonck K. Comparing exosome-like vesicles with liposomes for the functional cellular delivery of small RNAs. J Control Release. 2016;232:51–61. doi: 10.1016/j.jconrel.2016.04.005. [DOI] [PubMed] [Google Scholar]
- 25.Mirzaei R, Zamani F, Hajibaba M, Rasouli-Saravani A, Noroozbeygi M, Gorgani M, Hosseini-Fard SR, Jalalifar S, Ajdarkosh H, Abedi SH, Keyvani H, Karampoor S. The pathogenic, therapeutic and diagnostic role of exosomal microRNA in the autoimmune diseases. J Neuroimmunol. 2021;358:577640. doi: 10.1016/j.jneuroim.2021.577640. [DOI] [PubMed] [Google Scholar]
- 26.Chen J, Ding C, Yang X, Zhao J. BMSCs-derived exosomal miR-126-3p inhibits the viability of NSCLC cells by targeting PTPN9. J BUON. 2021;26:1832–1841. [PubMed] [Google Scholar]
- 27.Rong X, Liu J, Yao X, Jiang T, Wang Y, Xie F. Human bone marrow mesenchymal stem cells-derived exosomes alleviate liver fibrosis through the Wnt/β-catenin pathway. Stem Cell Res Ther. 2019;10:98. doi: 10.1186/s13287-019-1204-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sun Y, Shi H, Yin S, Ji C, Zhang X, Zhang B, Wu P, Shi Y, Mao F, Yan Y, Xu W, Qian H. Human mesenchymal stem cell derived exosomes alleviate type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano. 2018;12:7613–7628. doi: 10.1021/acsnano.7b07643. [DOI] [PubMed] [Google Scholar]
- 29.Cai X, Zhang ZY, Yuan JT, Ocansey DKW, Tu Q, Zhang X, Qian H, Xu WR, Qiu W, Mao F. hucMSC-derived exosomes attenuate colitis by regulating macrophage pyroptosis via the miR-378a-5p/NLRP3 axis. Stem Cell Res Ther. 2021;12:416. doi: 10.1186/s13287-021-02492-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang J, Pei B, Yan J, Xu X, Fang AN, Ocansey DKW, Zhang X, Qian H, Xu W, Mao F. hucMSC-derived exosomes alleviate the deterioration of colitis via the miR-146a/SUMO1 axis. Mol Pharm. 2022;19:484–493. doi: 10.1021/acs.molpharmaceut.1c00450. [DOI] [PubMed] [Google Scholar]
- 31.Xu Y, Tang X, Fang A, Yan J, Kofi Wiredu Ocansey D, Zhang X, Mao F. HucMSC-Ex carrying miR-203a-3p.2 ameliorates colitis through the suppression of caspase11/4-induced macrophage pyroptosis. Int Immunopharmacol. 2022;110:108925. doi: 10.1016/j.intimp.2022.108925. [DOI] [PubMed] [Google Scholar]
- 32.Wang Y, Zhang Y, Lu B, Xi J, Ocansey DKW, Mao F, Hao D, Yan Y. hucMSC-ex alleviates IBD-associated intestinal fibrosis by inhibiting ERK phosphorylation in intestinal fibroblasts. Stem Cells Int. 2023;2023:2828981. doi: 10.1155/2023/2828981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang G, Yuan J, Cai X, Xu Z, Wang J, Ocansey DKW, Yan Y, Qian H, Zhang X, Xu W, Mao F. HucMSC-exosomes carrying miR-326 inhibit neddylation to relieve inflammatory bowel disease in mice. Clin Transl Med. 2020;10:e113. doi: 10.1002/ctm2.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cho BS, Kim JO, Ha DH, Yi YW. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem Cell Res Ther. 2018;9:187. doi: 10.1186/s13287-018-0939-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Heidari N, Abbasi-Kenarsari H, Namaki S, Baghaei K, Zali MR, Ghaffari Khaligh S, Hashemi SM. Adipose-derived mesenchymal stem cell-secreted exosome alleviates dextran sulfate sodium-induced acute colitis by Treg cell induction and inflammatory cytokine reduction. J Cell Physiol. 2021;236:5906–5920. doi: 10.1002/jcp.30275. [DOI] [PubMed] [Google Scholar]
- 36.Yu H, Yang X, Xiao X, Xu M, Yang Y, Xue C, Li X, Wang S, Zhao RC. Human adipose mesenchymal stem cell-derived exosomes protect mice from DSS-induced inflammatory bowel disease by promoting intestinal-stem-cell and epithelial regeneration. Aging Dis. 2021;12:1423–1437. doi: 10.14336/AD.2021.0601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhang W, Bai X, Zhao B, Li Y, Zhang Y, Li Z, Wang X, Luo L, Han F, Zhang J, Han S, Cai W, Su L, Tao K, Shi J, Hu D. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp Cell Res. 2018;370:333–342. doi: 10.1016/j.yexcr.2018.06.035. [DOI] [PubMed] [Google Scholar]
- 38.Zhang J, Ji C, Zhang H, Shi H, Mao F, Qian H, Xu W, Wang D, Pan J, Fang X, Santos HA, Zhang X. Engineered neutrophil-derived exosome-like vesicles for targeted cancer therapy. Sci Adv. 2022;8:eabj8207. doi: 10.1126/sciadv.abj8207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang L, Yu Z, Wan S, Wu F, Chen W, Zhang B, Lin D, Liu J, Xie H, Sun X, Wu Z. Exosomes derived from dendritic cells treated with schistosoma japonicum soluble egg antigen attenuate DSS-induced colitis. Front Pharmacol. 2017;8:651. doi: 10.3389/fphar.2017.00651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Xiao GY, Cheng CC, Chiang YS, Cheng WT, Liu IH, Wu SC. Exosomal miR-10a derived from amniotic fluid stem cells preserves ovarian follicles after chemotherapy. Sci Rep. 2016;6:23120. doi: 10.1038/srep23120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mao Y, Han M, Chen C, Wang X, Han J, Gao Y, Wang S. A biomimetic nanocomposite made of a ginger-derived exosome and an inorganic framework for high-performance delivery of oral antibodies. Nanoscale. 2021;13:20157–20169. doi: 10.1039/d1nr06015e. [DOI] [PubMed] [Google Scholar]
- 42.Moradi-Chaleshtori M, Bandehpour M, Soudi S, Mohammadi-Yeganeh S, Hashemi SM. In vitro and in vivo evaluation of anti-tumoral effect of M1 phenotype induction in macrophages by miR-130 and miR-33 containing exosomes. Cancer Immunol Immunother. 2021;70:1323–1339. doi: 10.1007/s00262-020-02762-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ekström K, Valadi H, Sjöstrand M, Malmhäll C, Bossios A, Eldh M, Lötvall J. Characterization of mRNA and microRNA in human mast cell-derived exosomes and their transfer to other mast cells and blood CD34 progenitor cells. J Extracell Vesicles. 2012:1. doi: 10.3402/jev.v1i0.18389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Saumell-Esnaola M, Delgado D, García Del Caño G, Beitia M, Sallés J, González-Burguera I, Sánchez P, López de Jesús M, Barrondo S, Sánchez M. Isolation of platelet-derived exosomes from human platelet-rich plasma: biochemical and morphological characterization. Int J Mol Sci. 2022;23:2861. doi: 10.3390/ijms23052861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Guay C, Kruit JK, Rome S, Menoud V, Mulder NL, Jurdzinski A, Mancarella F, Sebastiani G, Donda A, Gonzalez BJ, Jandus C, Bouzakri K, Pinget M, Boitard C, Romero P, Dotta F, Regazzi R. Lymphocyte-derived exosomal microRNAs promote pancreatic β cell death and may contribute to type 1 diabetes development. Cell Metab. 2019;29:348–361. e346. doi: 10.1016/j.cmet.2018.09.011. [DOI] [PubMed] [Google Scholar]
- 46.Hazan-Halevy I, Rosenblum D, Weinstein S, Bairey O, Raanani P, Peer D. Cell-specific uptake of mantle cell lymphoma-derived exosomes by malignant and non-malignant B-lymphocytes. Cancer Lett. 2015;364:59–69. doi: 10.1016/j.canlet.2015.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Li Z, Suo B, Long G, Gao Y, Song J, Zhang M, Feng B, Shang C, Wang D. Exosomal miRNA-16-5p derived from M1 macrophages enhances T cell-dependent immune response by regulating PD-L1 in gastric cancer. Front Cell Dev Biol. 2020;8:572689. doi: 10.3389/fcell.2020.572689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lu J, Liu D, Tan Y, Deng F, Li R. M1 macrophage exosomes miR-21a-5p aggravates inflammatory bowel disease through decreasing E-cadherin and subsequent ILC2 activation. J Cell Mol Med. 2021;25:3041–3050. doi: 10.1111/jcmm.16348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Deng F, Yan J, Lu J, Luo M, Xia P, Liu S, Wang X, Zhi F, Liu D. M2 macrophage-derived exosomal miR-590-3p attenuates DSS-induced mucosal damage and promotes epithelial repair via the LATS1/YAP/β-catenin signalling axis. J Crohns Colitis. 2021;15:665–677. doi: 10.1093/ecco-jcc/jjaa214. [DOI] [PubMed] [Google Scholar]
- 50.Han G, Cho H, Kim H, Jang Y, Jang H, Kim DE, Kim ES, Kim EH, Hwang KY, Kim K, Yang Y, Kim SH. Bovine colostrum derived-exosomes prevent dextran sulfate sodium-induced intestinal colitis via suppression of inflammation and oxidative stress. Biomater Sci. 2022;10:2076–2087. doi: 10.1039/d1bm01797g. [DOI] [PubMed] [Google Scholar]
- 51.Lai JJ, Chau ZL, Chen SY, Hill JJ, Korpany KV, Liang NW, Lin LH, Lin YH, Liu JK, Liu YC, Lunde R, Shen WT. Exosome processing and characterization approaches for research and technology development. Adv Sci (Weinh) 2022;9:e2103222. doi: 10.1002/advs.202103222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yue B, Yang H, Wang J, Ru W, Wu J, Huang Y, Lan X, Lei C, Chen H. Exosome biogenesis, secretion and function of exosomal miRNAs in skeletal muscle myogenesis. Cell Prolif. 2020;53:e12857. doi: 10.1111/cpr.12857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977. doi: 10.1126/science.aau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–125. doi: 10.1016/j.ceb.2014.05.004. [DOI] [PubMed] [Google Scholar]
- 55.He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8:237–255. doi: 10.7150/thno.21945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brügger B, Simons M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319:1244–1247. doi: 10.1126/science.1153124. [DOI] [PubMed] [Google Scholar]
- 57.Juan T, Fürthauer M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol. 2018;74:66–77. doi: 10.1016/j.semcdb.2017.08.022. [DOI] [PubMed] [Google Scholar]
- 58.Tamai K, Tanaka N, Nakano T, Kakazu E, Kondo Y, Inoue J, Shiina M, Fukushima K, Hoshino T, Sano K, Ueno Y, Shimosegawa T, Sugamura K. Exosome secretion of dendritic cells is regulated by Hrs, an ESCRT-0 protein. Biochem Biophys Res Commun. 2010;399:384–390. doi: 10.1016/j.bbrc.2010.07.083. [DOI] [PubMed] [Google Scholar]
- 59.Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, Manel N, Moita LF, Théry C, Raposo G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126:5553–5565. doi: 10.1242/jcs.128868. [DOI] [PubMed] [Google Scholar]
- 60.Quinney KB, Frankel EB, Shankar R, Kasberg W, Luong P, Audhya A. Growth factor stimulation promotes multivesicular endosome biogenesis by prolonging recruitment of the late-acting ESCRT machinery. Proc Natl Acad Sci U S A. 2019;116:6858–6867. doi: 10.1073/pnas.1817898116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Roucourt B, Meeussen S, Bao J, Zimmermann P, David G. Heparanase activates the syndecan-syntenin-ALIX exosome pathway. Cell Res. 2015;25:412–428. doi: 10.1038/cr.2015.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ghossoub R, Lembo F, Rubio A, Gaillard CB, Bouchet J, Vitale N, Slavík J, Machala M, Zimmermann P. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat Commun. 2014;5:3477. doi: 10.1038/ncomms4477. [DOI] [PubMed] [Google Scholar]
- 63.Albacete-Albacete L, Navarro-Lérida I, López JA, Martín-Padura I, Astudillo AM, Ferrarini A, Van-Der-Heyden M, Balsinde J, Orend G, Vázquez J, Del Pozo MÁ. ECM deposition is driven by caveolin-1-dependent regulation of exosomal biogenesis and cargo sorting. J Cell Biol. 2020;219:e202006178. doi: 10.1083/jcb.202006178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.García-Seisdedos D, Babiy B, Lerma M, Casado ME, Martínez-Botas J, Lasunción MA, Pastor Ó, Busto R. Curcumin stimulates exosome/microvesicle release in an in vitro model of intracellular lipid accumulation by increasing ceramide synthesis. Biochim Biophys Acta Mol Cell Biol Lipids. 2020;1865:158638. doi: 10.1016/j.bbalip.2020.158638. [DOI] [PubMed] [Google Scholar]
- 65.Zou W, Lai M, Zhang Y, Zheng L, Xing Z, Li T, Zou Z, Song Q, Zhao X, Xia L, Yang J, Liu A, Zhang H, Cui ZK, Jiang Y, Bai X. Exosome release is regulated by mTORC1. Adv Sci (Weinh) 2019;6:1801313. doi: 10.1002/advs.201801313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Villarroya-Beltri C, Baixauli F, Mittelbrunn M, Fernández-Delgado I, Torralba D, Moreno-Gonzalo O, Baldanta S, Enrich C, Guerra S, Sánchez-Madrid F. ISGylation controls exosome secretion by promoting lysosomal degradation of MVB proteins. Nat Commun. 2016;7:13588. doi: 10.1038/ncomms13588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.van Niel G, Porto-Carreiro I, Simoes S, Raposo G. Exosomes: a common pathway for a specialized function. J Biochem. 2006;140:13–21. doi: 10.1093/jb/mvj128. [DOI] [PubMed] [Google Scholar]
- 68.Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. J Transl Med. 2011;9:86. doi: 10.1186/1479-5876-9-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tan Y, Huang Y, Mei R, Mao F, Yang D, Liu J, Xu W, Qian H, Yan Y. HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death Dis. 2022;13:319. doi: 10.1038/s41419-022-04764-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Xu Y, Zhang L, Ocansey DKW, Wang B, Hou Y, Mei R, Yan Y, Zhang X, Zhang Z, Mao F. HucMSC-Ex alleviates inflammatory bowel disease via the lnc78583-mediated miR3202/HOXB13 pathway. J Zhejiang Univ Sci B. 2022;23:423–431. doi: 10.1631/jzus.B2100793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wei Z, Hang S, Wiredu Ocansey DK, Zhang Z, Wang B, Zhang X, Mao F. Human umbilical cord mesenchymal stem cells derived exosome shuttling miR-129-5p attenuates inflammatory bowel disease by inhibiting ferroptosis. J Nanobiotechnology. 2023;21:188. doi: 10.1186/s12951-023-01951-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Palmisano G, Jensen SS, Le Bihan MC, Lainé J, McGuire JN, Pociot F, Larsen MR. Characterization of membrane-shed microvesicles from cytokine-stimulated β-cells using proteomics strategies. Mol Cell Proteomics. 2012;11:230–243. doi: 10.1074/mcp.M111.012732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jeppesen DK, Hvam ML, Primdahl-Bengtson B, Boysen AT, Whitehead B, Dyrskjøt L, Orntoft TF, Howard KA, Ostenfeld MS. Comparative analysis of discrete exosome fractions obtained by differential centrifugation. J Extracell Vesicles. 2014;3:25011. doi: 10.3402/jev.v3.25011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, Mark MT, Molina H, Martin AB, Bojmar L, Fang J, Rampersaud S, Hoshino A, Matei I, Kenific CM, Nakajima M, Mutvei AP, Sansone P, Buehring W, Wang H, Jimenez JP, Cohen-Gould L, Paknejad N, Brendel M, Manova-Todorova K, Magalhães A, Ferreira JA, Osório H, Silva AM, Massey A, Cubillos-Ruiz JR, Galletti G, Giannakakou P, Cuervo AM, Blenis J, Schwartz R, Brady MS, Peinado H, Bromberg J, Matsui H, Reis CA, Lyden D. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20:332–343. doi: 10.1038/s41556-018-0040-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yokoi A, Villar-Prados A, Oliphint PA, Zhang J, Song X, De Hoff P, Morey R, Liu J, Roszik J, Clise-Dwyer K, Burks JK, O’Halloran TJ, Laurent LC, Sood AK. Mechanisms of nuclear content loading to exosomes. Sci Adv. 2019;5:eaax8849. doi: 10.1126/sciadv.aax8849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Vaidya M, Smith J, Field M, Sugaya K. Analysis of regulatory sequences in exosomal DNA of NANOGP8. PLoS One. 2023;18:e0280959. doi: 10.1371/journal.pone.0280959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Torralba D, Baixauli F, Villarroya-Beltri C, Fernández-Delgado I, Latorre-Pellicer A, Acín-Pérez R, Martín-Cófreces NB, Jaso-Tamame ÁL, Iborra S, Jorge I, González-Aseguinolaza G, Garaude J, Vicente-Manzanares M, Enríquez JA, Mittelbrunn M, Sánchez-Madrid F. Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat Commun. 2018;9:2658. doi: 10.1038/s41467-018-05077-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Takahashi A, Okada R, Nagao K, Kawamata Y, Hanyu A, Yoshimoto S, Takasugi M, Watanabe S, Kanemaki MT, Obuse C, Hara E. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat Commun. 2017;8:15287. doi: 10.1038/ncomms15287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhao F, Zheng T, Gong W, Wu J, Xie H, Li W, Zhang R, Liu P, Liu J, Wu X, Zhao Y, Ren J. Extracellular vesicles package dsDNA to aggravate Crohn’s disease by activating the STING pathway. Cell Death Dis. 2021;12:815. doi: 10.1038/s41419-021-04101-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Liu XM, Ma L, Schekman R. Selective sorting of microRNAs into exosomes by phase-separated YBX1 condensates. Elife. 2021;10:e71982. doi: 10.7554/eLife.71982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Shurtleff MJ, Temoche-Diaz MM, Karfilis KV, Ri S, Schekman R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. Elife. 2016;5:e19276. doi: 10.7554/eLife.19276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lambertz U, Oviedo Ovando ME, Vasconcelos EJ, Unrau PJ, Myler PJ, Reiner NE. Small RNAs derived from tRNAs and rRNAs are highly enriched in exosomes from both old and new world Leishmania providing evidence for conserved exosomal RNA Packaging. BMC Genomics. 2015;16:151. doi: 10.1186/s12864-015-1260-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hobor F, Dallmann A, Ball NJ, Cicchini C, Battistelli C, Ogrodowicz RW, Christodoulou E, Martin SR, Castello A, Tripodi M, Taylor IA, Ramos A. A cryptic RNA-binding domain mediates Syncrip recognition and exosomal partitioning of miRNA targets. Nat Commun. 2018;9:831. doi: 10.1038/s41467-018-03182-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wozniak AL, Adams A, King KE, Dunn W, Christenson LK, Hung WT, Weinman SA. The RNA binding protein FMR1 controls selective exosomal miRNA cargo loading during inflammation. J Cell Biol. 2020;219:e201912074. doi: 10.1083/jcb.201912074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pan Z, Zhao R, Li B, Qi Y, Qiu W, Guo Q, Zhang S, Zhao S, Xu H, Li M, Gao Z, Fan Y, Xu J, Wang H, Wang S, Qiu J, Wang Q, Guo X, Deng L, Zhang P, Xue H, Li G. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer. 2022;21:16. doi: 10.1186/s12943-021-01485-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Robinson H, Ruelcke JE, Lewis A, Bond CS, Fox AH, Bharti V, Wani S, Cloonan N, Lai A, Margolin D, Li L, Salomon C, Richards RS, Farrell A, Gardiner RA, Parton RG, Cristino AS, Hill MM. Caveolin-1-driven membrane remodelling regulates hnRNPK-mediated exosomal microRNA sorting in cancer. Clin Transl Med. 2021;11:e381. doi: 10.1002/ctm2.381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Suresh PS, Thankachan S, Venkatesh T. Landscape of clinically relevant exosomal tRNA-derived non-coding RNAs. Mol Biotechnol. 2023;65:300–310. doi: 10.1007/s12033-022-00546-5. [DOI] [PubMed] [Google Scholar]
- 88.Yu Z, Tang H, Chen S, Xie Y, Shi L, Xia S, Jiang M, Li J, Chen D. Exosomal LOC85009 inhibits docetaxel resistance in lung adenocarcinoma through regulating ATG5-induced autophagy. Drug Resist Updat. 2023;67:100915. doi: 10.1016/j.drup.2022.100915. [DOI] [PubMed] [Google Scholar]
- 89.Zhu L, Li J, Gong Y, Wu Q, Tan S, Sun D, Xu X, Zuo Y, Zhao Y, Wei YQ, Wei XW, Peng Y. Exosomal tRNA-derived small RNA as a promising biomarker for cancer diagnosis. Mol Cancer. 2019;18:74. doi: 10.1186/s12943-019-1000-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Skotland T, Hessvik NP, Sandvig K, Llorente A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J Lipid Res. 2019;60:9–18. doi: 10.1194/jlr.R084343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lobasso S, Tanzarella P, Mannavola F, Tucci M, Silvestris F, Felici C, Ingrosso C, Corcelli A, Lopalco P. A lipidomic approach to identify potential biomarkers in exosomes from melanoma cells with different metastatic potential. Front Physiol. 2021;12:748895. doi: 10.3389/fphys.2021.748895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Llorente A, Skotland T, Sylvänne T, Kauhanen D, Róg T, Orłowski A, Vattulainen I, Ekroos K, Sandvig K. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. Biochim Biophys Acta. 2013;1831:1302–1309. doi: 10.1016/j.bbalip.2013.04.011. [DOI] [PubMed] [Google Scholar]
- 93.Hosseini-Beheshti E, Pham S, Adomat H, Li N, Tomlinson Guns ES. Exosomes as biomarker enriched microvesicles: characterization of exosomal proteins derived from a panel of prostate cell lines with distinct AR phenotypes. Mol Cell Proteomics. 2012;11:863–885. doi: 10.1074/mcp.M111.014845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Peterka O, Jirásko R, Chocholoušková M, Kuchař L, Wolrab D, Hájek R, Vrána D, Strouhal O, Melichar B, Holčapek M. Lipidomic characterization of exosomes isolated from human plasma using various mass spectrometry techniques. Biochim Biophys Acta Mol Cell Biol Lipids. 2020;1865:158634. doi: 10.1016/j.bbalip.2020.158634. [DOI] [PubMed] [Google Scholar]
- 95.Elmallah MIY, Ortega-Deballon P, Hermite L, Pais-De-Barros JP, Gobbo J, Garrido C. Lipidomic profiling of exosomes from colorectal cancer cells and patients reveals potential biomarkers. Mol Oncol. 2022;16:2710–2718. doi: 10.1002/1878-0261.13223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mao F, Wu Y, Tang X, Kang J, Zhang B, Yan Y, Qian H, Zhang X, Xu W. Exosomes derived from human umbilical cord mesenchymal stem cells relieve inflammatory bowel disease in mice. Biomed Res Int. 2017;2017:5356760. doi: 10.1155/2017/5356760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Qian W, Huang L, Xu Y, Lu W, Wen W, Guo Z, Zhu W, Li Y. Hypoxic ASCs-derived exosomes attenuate colitis by regulating macrophage polarization via miR-216a-5p/HMGB1 axis. Inflamm Bowel Dis. 2023;29:602–619. doi: 10.1093/ibd/izac225. [DOI] [PubMed] [Google Scholar]
- 98.Deng C, Hu Y, Conceição M, Wood MJA, Zhong H, Wang Y, Shao P, Chen J, Qiu L. Oral delivery of layer-by-layer coated exosomes for colitis therapy. J Control Release. 2023;354:635–650. doi: 10.1016/j.jconrel.2023.01.017. [DOI] [PubMed] [Google Scholar]
- 99.Xu H, Fu J, Chen L, Zhou S, Fang Y, Zhang Q, Chen X, Yuan L, Li Y, Xu Z, Xiang C. TNF-α enhances the therapeutic effects of MenSC-derived small extracellular vesicles on inflammatory bowel disease through macrophage polarization by miR-24-3p. Stem Cells Int. 2023;2023:2988907. doi: 10.1155/2023/2988907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Cao L, Xu H, Wang G, Liu M, Tian D, Yuan Z. Extracellular vesicles derived from bone marrow mesenchymal stem cells attenuate dextran sodium sulfate-induced ulcerative colitis by promoting M2 macrophage polarization. Int Immunopharmacol. 2019;72:264–274. doi: 10.1016/j.intimp.2019.04.020. [DOI] [PubMed] [Google Scholar]
- 101.Liu H, Liang Z, Wang F, Zhou C, Zheng X, Hu T, He X, Wu X, Lan P. Exosomes from mesenchymal stromal cells reduce murine colonic inflammation via a macrophage-dependent mechanism. JCI Insight. 2019;4:e131273. doi: 10.1172/jci.insight.131273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Allaire JM, Crowley SM, Law HT, Chang SY, Ko HJ, Vallance BA. The intestinal epithelium: central coordinator of mucosal immunity. Trends Immunol. 2018;39:677–696. doi: 10.1016/j.it.2018.04.002. [DOI] [PubMed] [Google Scholar]
- 103.Yang J, Zheng XG, Wu YL, Wang AP, Wang CH, Chen WX, Zhong S, Yang H. Intestinal epithelial cell-derived exosomes package microRNA-23a-3p alleviate gut damage after ischemia/reperfusion via targeting MAP4K4. Biomed Pharmacother. 2022;149:112810. doi: 10.1016/j.biopha.2022.112810. [DOI] [PubMed] [Google Scholar]
- 104.Park EJ, Shimaoka M, Kiyono H. Functional flexibility of exosomes and microRNAs of intestinal epithelial cells in affecting inflammation. Front Mol Biosci. 2022;9:854487. doi: 10.3389/fmolb.2022.854487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Appiah MG, Park EJ, Darkwah S, Kawamoto E, Akama Y, Gaowa A, Kalsan M, Ahmad S, Shimaoka M. Intestinal epithelium-derived luminally released extracellular vesicles in sepsis exhibit the ability to suppress TNF-a and IL-17A expression in mucosal inflammation. Int J Mol Sci. 2020;21:8445. doi: 10.3390/ijms21228445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233:6425–6440. doi: 10.1002/jcp.26429. [DOI] [PubMed] [Google Scholar]
- 107.Arabpour M, Saghazadeh A, Rezaei N. Anti-inflammatory and M2 macrophage polarization-promoting effect of mesenchymal stem cell-derived exosomes. Int Immunopharmacol. 2021;97:107823. doi: 10.1016/j.intimp.2021.107823. [DOI] [PubMed] [Google Scholar]
- 108.Miksa M, Wu R, Dong W, Komura H, Amin D, Ji Y, Wang Z, Wang H, Ravikumar TS, Tracey KJ, Wang P. Immature dendritic cell-derived exosomes rescue septic animals via milk fat globule epidermal growth factor-factor VIII [corrected] . J Immunol. 2009;183:5983–5990. doi: 10.4049/jimmunol.0802994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Miksa M, Wu R, Dong W, Das P, Yang D, Wang P. Dendritic cell-derived exosomes containing milk fat globule epidermal growth factor-factor VIII attenuate proinflammatory responses in sepsis. Shock. 2006;25:586–593. doi: 10.1097/01.shk.0000209533.22941.d0. [DOI] [PubMed] [Google Scholar]
- 110.Wang Y, Tian J, Tang X, Rui K, Tian X, Ma J, Ma B, Xu H, Lu L, Wang S. Exosomes released by granulocytic myeloid-derived suppressor cells attenuate DSS-induced colitis in mice. Oncotarget. 2016;7:15356–15368. doi: 10.18632/oncotarget.7324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Pourcet B, Pineda-Torra I. Transcriptional regulation of macrophage arginase 1 expression and its role in atherosclerosis. Trends Cardiovasc Med. 2013;23:143–152. doi: 10.1016/j.tcm.2012.10.003. [DOI] [PubMed] [Google Scholar]
- 112.Gao C, Zhou Y, Chen Z, Li H, Xiao Y, Hao W, Zhu Y, Vong CT, Farag MA, Wang Y, Wang S. Turmeric-derived nanovesicles as novel nanobiologics for targeted therapy of ulcerative colitis. Theranostics. 2022;12:5596–5614. doi: 10.7150/thno.73650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kim J, Zhang S, Zhu Y, Wang R, Wang J. Amelioration of colitis progression by ginseng-derived exosome-like nanoparticles through suppression of inflammatory cytokines. J Ginseng Res. 2023;47:627–637. doi: 10.1016/j.jgr.2023.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Muñoz-Esparza NC, Latorre-Moratalla ML, Comas-Basté O, Toro-Funes N, Veciana-Nogués MT, Vidal-Carou MC. Polyamines in food. Front Nutr. 2019;6:108. doi: 10.3389/fnut.2019.00108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ma L, Ni L, Yang T, Mao P, Huang X, Luo Y, Jiang Z, Hu L, Zhao Y, Fu Z, Ni Y. Preventive and therapeutic spermidine treatment attenuates acute colitis in mice. J Agric Food Chem. 2021;69:1864–1876. doi: 10.1021/acs.jafc.0c07095. [DOI] [PubMed] [Google Scholar]
- 116.Tong L, Hao H, Zhang Z, Lv Y, Liang X, Liu Q, Liu T, Gong P, Zhang L, Cao F, Pastorin G, Lee CN, Chen X, Wang JW, Yi H. Milk-derived extracellular vesicles alleviate ulcerative colitis by regulating the gut immunity and reshaping the gut microbiota. Theranostics. 2021;11:8570–8586. doi: 10.7150/thno.62046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Gao X, Yang Y, Liu X, Wang Y, Yang Y, Boireau P, Liu M, Bai X. Extracellular vesicles derived from Trichinella spiralis prevent colitis by inhibiting M1 macrophage polarization. Acta Trop. 2021;213:105761. doi: 10.1016/j.actatropica.2020.105761. [DOI] [PubMed] [Google Scholar]
- 118.Fan L, Yao L, Li Z, Wan Z, Sun W, Qiu S, Zhang W, Xiao D, Song L, Yang G, Zhang Y, Wei M, Yang X. Exosome-based mitochondrial delivery of circRNA mSCAR alleviates sepsis by orchestrating macrophage activation. Adv Sci (Weinh) 2023;10:e2205692. doi: 10.1002/advs.202205692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wu Y, Wu B, Zhang Z, Lu H, Fan C, Qi Q, Gao Y, Li H, Feng C, Zuo J, Tang W. Heme protects intestinal mucosal barrier in DSS-induced colitis through regulating macrophage polarization in both HO-1-dependent and HO-1-independent way. FASEB J. 2020;34:8028–8043. doi: 10.1096/fj.202000313RR. [DOI] [PubMed] [Google Scholar]
- 120.Zhu W, Jin Z, Yu J, Liang J, Yang Q, Li F, Shi X, Zhu X, Zhang X. Baicalin ameliorates experimental inflammatory bowel disease through polarization of macrophages to an M2 phenotype. Int Immunopharmacol. 2016;35:119–126. doi: 10.1016/j.intimp.2016.03.030. [DOI] [PubMed] [Google Scholar]
- 121.Liang C, Tang Y, Gao X, Lei N, Luo Y, Chen P, Duan S, Cao Y, Yang Y, Zhang Y. Depression exacerbates dextran sulfate sodium-induced colitis via IRF5-mediated macrophage polarization. Dig Dis Sci. 2023;68:1269–1279. doi: 10.1007/s10620-022-07679-2. [DOI] [PubMed] [Google Scholar]
- 122.Lu J, Liu D, Tan Y, Li R, Wang X, Deng F. Thalidomide attenuates colitis and is associated with the suppression of M1 macrophage polarization by targeting the transcription factor IRF5. Dig Dis Sci. 2021;66:3803–3812. doi: 10.1007/s10620-021-07067-2. [DOI] [PubMed] [Google Scholar]
- 123.Lu Q, Li J, Ding P, Mao T, Shi L, Sun Z, Tan X, Jiang H, Dong J, Li Y, Yang X, Shi R. Qingchang Wenzhong Decoction alleviates DSS-induced inflammatory bowel disease by inhibiting M1 macrophage polarization in vitro and in vivo. Biomed Res Int. 2022;2022:9427076. doi: 10.1155/2022/9427076. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 124.Xu YW, Xing RX, Zhang WH, Li L, Wu Y, Hu J, Wang C, Luo QL, Shen JL, Chen X. Toxoplasma ROP16(I/III) ameliorated inflammatory bowel diseases via inducing M2 phenotype of macrophages. World J Gastroenterol. 2019;25:6634–6652. doi: 10.3748/wjg.v25.i45.6634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Han D, Lu D, Huang S, Pang J, Wu Y, Hu J, Zhang X, Pi Y, Zhang G, Wang J. Small extracellular vesicles from Ptpn1-deficient macrophages alleviate intestinal inflammation by reprogramming macrophage polarization via lactadherin enrichment. Redox Biol. 2022;58:102558. doi: 10.1016/j.redox.2022.102558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Song S, An J, Li Y, Liu S. Electroacupuncture at ST-36 ameliorates DSS-induced acute colitis via regulating macrophage polarization induced by suppressing NLRP3/IL-1β and promoting Nrf2/HO-1. Mol Immunol. 2019;106:143–152. doi: 10.1016/j.molimm.2018.12.023. [DOI] [PubMed] [Google Scholar]
- 127.Liu Y, Liu X, Hua W, Wei Q, Fang X, Zhao Z, Ge C, Liu C, Chen C, Tao Y, Zhu Y. Berberine inhibits macrophage M1 polarization via AKT1/SOCS1/NF-κB signaling pathway to protect against DSS-induced colitis. Int Immunopharmacol. 2018;57:121–131. doi: 10.1016/j.intimp.2018.01.049. [DOI] [PubMed] [Google Scholar]
- 128.Liu Y, Li Z, Xu Y, Liu Y, Xue C. Enzymatic modification of D-mannose alleviates DSS-induced colonic inflammation in mice through macrophage polarization mediated by PPARγ. Food Funct. 2022;13:11467–11475. doi: 10.1039/d1fo03897d. [DOI] [PubMed] [Google Scholar]
- 129.Record M, Subra C, Silvente-Poirot S, Poirot M. Exosomes as intercellular signalosomes and pharmacological effectors. Biochem Pharmacol. 2011;81:1171–1182. doi: 10.1016/j.bcp.2011.02.011. [DOI] [PubMed] [Google Scholar]
- 130.Wang Y, Liu H, Zhang Z, Bian D, Shao K, Wang S, Ding Y. G-MDSC-derived exosomes mediate the differentiation of M-MDSC into M2 macrophages promoting colitis-to-cancer transition. J Immunother Cancer. 2023;11:e006166. doi: 10.1136/jitc-2022-006166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wong WY, Lee MM, Chan BD, Kam RK, Zhang G, Lu AP, Tai WC. Proteomic profiling of dextran sulfate sodium induced acute ulcerative colitis mice serum exosomes and their immunomodulatory impact on macrophages. Proteomics. 2016;16:1131–1145. doi: 10.1002/pmic.201500174. [DOI] [PubMed] [Google Scholar]
- 132.Gong L, Xiao J, Yi J, Xiao J, Lu F, Liu X. Immunomodulatory effect of serum exosomes from crohn disease on macrophages via let-7b-5p/TLR4 signaling. Inflamm Bowel Dis. 2022;28:96–108. doi: 10.1093/ibd/izab132. [DOI] [PubMed] [Google Scholar]
- 133.Liu H, Liang Z, Wang F, Zheng X, Zeng Z, He X, Gao X, Zhi M, Wu X, Wu X, Lan P. Intestinal CD14+ macrophages protect CD4+ T cells from activation-induced cell death via exosomal membrane TNF in Crohn’s disease. J Crohns Colitis. 2020;14:1619–1631. doi: 10.1093/ecco-jcc/jjaa083. [DOI] [PubMed] [Google Scholar]