Skip to main content

Exosomal miRNA-mediated intercellular communications and immunomodulatory effects in tumor microenvironments

Abstract

Extracellular communication, in other words, crosstalk between cells, has a pivotal role in the survival of an organism. This communication occurs by different methods, one of which is extracellular vesicles. Exosomes, which are small lipid extracellular vesicles, have recently been discovered to have a role in signal transduction between cells inside the body. These vesicles contain important bioactive molecules including lipids, proteins, DNA, mRNA, and noncoding RNAs such as microRNAs (miRNAs). Exosomes are secreted by all cells including immune cells (macrophages, lymphocytes, granulocytes, dendritic cells, mast cells) and tumor cells. The tumor microenvironment (TME) represents a complex network that supports the growth of tumor cells. This microenvironment encompasses tumor cells themselves, the extracellular matrix, fibroblasts, endothelial cells, blood vessels, immune cells, and non-cellular components such as exosomes and cytokines. This review aims to provide insights into the latest discoveries concerning how the immune system communicates internally and with other cell types, with a specific focus on research involving exosomal miRNAs in macrophages, dendritic cells, B lymphocytes, and T lymphocytes. Additionally, we will explore the role of exosomal miRNA in the TME and the immunomodulatory effect.

Background

Exosomes

Extracellular vesicles (EVs) are divided into two basic groups: ectosomes and exosomes. Ectosomes are vesicles formed by the straight outward budding of the plasma membrane, which results in microvesicles, microparticles, and giant vesicles with diameters ranging from 50 to 1 mm. Exosomes, on the other hand, are endosomal in origin and range in diameter from 30 to 150 nm (on average 100 nm). A particular feature of exosomes in biology is their creation, which may determine their composition, as well as possibly their function, once secreted into the extracellular environment [1]. Exosomes are involved in the biology of many diseases. By transferring and presenting exosomes, exosomes can modulate immune responses and inflammation of antigen peptides, to induce the expression of inflammatory genes in recipient cells [2]. Several metabolic diseases are associated with exosomes, including metabolic disorders related to adipocytes and islet cells [3, 4]. As well as inhibiting neurotoxic oligomers, exosomes can also contribute to neurodegeneration. The development of tumors, angiogenesis, metastasis, sensitivity to chemotherapy, and immune evasion are also associated with exosomes [5, 6].

Biogenesis of exosomes

Intracellular multivesicular bodies (MVBs) containing intraluminal vesicles (ILVs) are formed after two invaginations of the plasma membrane to form exosomes. These ILVs are finally released as exosomes ranging from 30 to 150 nm in diameter by MVB combination to the plasma membrane and exocytosis. During the initial invagination of the plasma membrane, a semi-circular structure is formed that contains proteins associated with the extracellular environment and cell surface proteins. The result of this is the formation of early-sorting endosomes (ESEs) from scratch or sometimes may combine with an already existing ESE. Moreover, the trans-Golgi network, the endoplasmic reticulum plays a significant role in the formation of the ESE. Multivesicular endosomes (MVBs) are produced when ESEs mature into late-sorting endosomes (LSEs). MVBs are formed by a second invagination of the plasma membrane. This is caused by the invagination of the endosomal limiting membrane inward. This process results in the formation of MVBs containing multiple ILVs (future exosomes). The MVBs either combine with lysosomes or autophagosomes to destroy or with the plasma membrane to release the contents of ILVs as exosomes [7,8,9,10]. This biogenesis of exosomes discriminates exosomes from EVs that develop via outward budding of the plasma membrane, apoptotic bodies, or necrotic blebs of the plasma membrane. Immune cells, mesenchymal stem cells, fibroblasts, neurons, endothelial cells (ECs), and epithelium cells all secrete exosomes (Fig. 1a).

Fig. 1
figure 1

Biogenesis of Exosomes: Exosomes are formed through the invagination of the plasma membrane in two phases. a The first phase involves budding formation, which incorporates cell surface proteins, extracellular soluble proteins, and other small molecules, resulting in the formation of early sorting endosomes (ESE). ESEs then mature and convert into late-sorting endosomes (LSE). The second phase involves inward budding of the endosomal limiting membrane, which incorporates constituents from the endoplasmic reticulum (ER), trans-Golgi network (TGN), as well as small biomolecules such as nucleic acids and mitochondria. This process leads to the formation of multivesicular bodies (MVBs). Subsequently, intracellular vesicles (exosomes) are gradually released into the microenvironment after MVBs fuse with the plasma membrane. Additionally, MVBs can fuse with lysosomes or autophagosomes for the degradation of their contents, which can be recycled by the cells. b The structure of exosomes: Exosomes contain a wide range of molecules, including nucleic acids, proteins, metabolites, and lipids. Both the membrane-bound components and the soluble components of exosomes represent the cellular origin from which they were released

The structure of exosomes

After secretion of exosomes from the cell surface, the exosomes have the ability to transport their contents to the cytoplasm of recipient cells by fusing with the plasma membrane. This process, called intracellular signaling, occurs by engaging the proteins on the surface of exosomes with cell surface protein on the recipient cell [11]. Therefore, exosome contents play a crucial role in determining their effect. Firstly, the exosomes plasma membrane is a phospholipid bilayer, the lipid consists of cholesterol, phosphatidylserine, saturated fatty acids, hexosylceramides, and sphingomyelin [11]. The proteome of exosomes includes endosomal, nuclear proteins, cytosolic, and plasma [12]. Exosome-enriched proteins include those involved in membrane delivering and fusing, such as Rab GTPases and annexins, as well as those involved in exosome biogenesis (ESCRT complex, ALIX, TSG101). Moreover, exosomes enriched with integrins, MHC class II proteins, tetraspanins (CD9, CD63, CD81, and CD82), epithelial cell adhesion molecules (EpCAM), heat shock proteins (HSP70, HSP90), and members of the human epidermal receptor (HER) family [13,14,15]. The protein constituents of exosomes reflect the biogenesis pathway and cell origin. Also, exosomes contain nucleic acids such as many types of RNAs including mRNA, transfer RNAs (tRNAs), viral RNA, and noncoding RNAs such as microRNA (miRNAs) and lncRNAs which are functional and can influence the transcription process in the recipient cells [16]. Exosomes contain small fragments of single-stranded DNA [17] and large fragments of double-stranded DNA [18] (Fig. 1b).

Communication between cells via exosomes

Intercellular communication occurs due to the presence of connections such as gap junctions or through the process of cell signaling. Signaling occurs when chemical mediators or bioactive molecules are released by one cell and affect other nearby cells. For example, hormones and enzymes released by one cell affect another at a distance [19]. There is also a distinctive way for cells to communicate with distant cells via extracellular vesicles. These membrane vesicles have the ability to transfer lipids, proteins, and genetic materials and function as cargo [20]. Mathieu et al. describe exosomes as small lipid bilayer vesicles carrying different molecules called cargo, which includes proteins, glycans, lipids, metabolites, mRNA, RNA, and DNA. When specific cells secreted exosomes that can act on receptor cells at distal sites and transfer their cargo. These display many of the features of classical endocrine signaling, therefore changing receptor cell phenotypes. Communication between cells has a pivotal role in the cell’s coordination and functioning effectively. This communication between cells can be initiated by exosomes because they have the capability of signal transduction [21].

There are several mechanisms involved in the uptake of exosomes by recipient cells, including endocytosis, micropinocytosis, and phagocytosis. The mechanism of micropinocytosis involves membrane invaginations that pinch off and draw extracellular contents into the cytosol (e.g., fluid and exosomes). The process of endocytosis can be mediated by clathrins or caveolins, and cholesterol-rich microdomains in the cell membrane may facilitate this. The phagocytosis process of exosomes mostly depends on PI3k, which is carried out most efficiently by macrophages. The last way of exosome uptake is by binding directly to the recipient cell membrane and emptying its contents. These mechanisms to transfer information between cells emphasize the different functions of exosomes including immunosuppression, antigen presentation, tumor cell proliferation, autophagy, and apoptosis [22]. As most body cells secret exosomes, therefore it can be found in a wide variety of bodily fluids and can affect neighboring cells functionally. According to some studies, miRNAs make up to 50% of the total noncoding RNA cargo within EVs, depending on the cellular origin of the EV [23,24,25]. The extracellular miRNAs transfer needs to occur via exosomes as it prevents degradation by free-floating RNases. In this review, we will focus on the questions surrounding the exosomal miRNA to understand their role in the crosstalk between the immune cells and the immunomodulatory effects of exosomal miRNA inside the tumor microenvironments.

miRNA of exosomes

Exosomes, as previously described, include a vast range of molecules, including proteins, lipids, DNAs, and RNA. Noncoding RNAs, which include miRNAs, tRNA fragments, and Y-RNAs, are the largest RNA component in exosomes, and long noncoding RNAs [26]. Of these molecules, miRNA is the most bioactive molecule that attracts much interest because of its regulatory roles in gene expression. Goldie et al. discovered that the percentage of miRNA in exosomes is greater than in their parent cells [27]. miRNAs are a type of tiny, noncoding RNA with a length of 17–24 nt that intermediate post-transcriptional gene silencing by 3ʹ-binding to the untranslated region (UTR) or open reading frame (ORF) region of target mRNA [28]. Also, many biological processes involving miRNAs have been thoroughly characterized, including cell proliferation, differentiation, migration, disease initiation, and progression. miRNAs are stable in bodily fluids such as saliva, urine, breast milk, and blood. Extracellular miRNAs can be loaded into high-density lipoprotein (HDL) or bound by AGO2 protein outside of vesicles besides being packed into exosomes or microvesicles that prevent miRNA from degradation and ensure their stability [29].

Biogenesis of miRNA

Inside the nucleus, miRNA biogenesis starts when miRNA of DNA is transcribed by RNA polymerase II to primary miRNA (pri-miRNA). These pri-miRNAs undergo the transcription process as fragments of larger molecules, which are then processed in the nucleus into hairpin RNAs of 70–100 nt by, the double-stranded RNA-specific ribonuclease, Drosha. Hairpin pre-miRNAs are processed by Dicer, a double-stranded-specific ribonuclease, after being moved to the cytoplasm by exportin 5. Double-stranded miRNAs are transformed into single-stranded miRNAs after maturation, then sorted into exosomes in various ways.

There are four potential routes for miRNA sorting into exosomes that have been discovered but these mechanisms are not fully understood. The neural sphingomyelinase 2-dependent (nSMase2) pathway is the first, which enhances the production of exosomal miRNA [30]. The heterogeneous nuclear ribonucleoprotein (hnRNP) dependent pathway is the second, which regulates exosomal miRNA entrance. The miRISC-associated pathway is the third, in this pathway, the mature miRNAs are incorporated into an RNA-induced silencing complex (RISC) along with argonaute (AGO2) and glycine-tryptophan protein of 182 kDa (GW182). AGO2 primarily attaches to U or A at the end of miRNA, which is critical in facilitating the formation of the miRNA: mRNA complex and then destroying or inhibiting translation of the mRNA molecule [31]. The sequence-dependent pathway is the fourth one, which involves the 3ʹ-end of miRNA [32]. The previous studies showed that the distribution of miRNAs in the B cells and their secreted exosomes are not random. miRNAs with an adenylated 3`-end are more abundant in cells, but those with a uridylated 3`-end are more abundant in exosomes. This suggests that the 3`-end of miRNA post-transcriptional modification has an important role in directing miRNA into the exosome [33].

Immune cells communicate via exosomal miRNAs

Dendritic cells

Dendritic cells (DCs) are specialized antigen-presenting cells that play critical roles in the initiation and modulation of the innate and adaptive immune systems. DCs are leukocytes generated from bone marrow (BM) and are the most powerful form of antigen-presenting cells. Therefore, when pathogens or pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide (LPS) invade our body, the mature DCs have the ability to stimulate the adaptive immune system, including T cells. This stimulation occurs through communications between DCs and neighboring cells in different ways such as cytokine signaling, intracellular contact, and vesicle exchange [34]. DC vesicle studies are a novel field because immature and mature bone marrow-derived DCs (BMDCs) are able to exert a huge number of exosomes, which is decreased upon maturation. This trait gives a foundation for understanding the basic cell biology of how exosomes are formed, transported, and internalized [35].

Extracellular miRNA transport via exosomes and the release of exosomes containing different miRNAs by DCs is dependent on their maturation level. Extracellular miRNAs are able to transfer into many different cell types. Because previous studies showed that dye-labeled BMDC exosomes may be taken up by splenic B cells, macrophages, plasmacytoid DCs, and conventional (CD11c-high) DCs in vivo, but not by T cells, both CD4 + and CD8 + subsets. On the other hand, the internalization of immature DC-derived labeled vesicles by CD4 + T cells was achievable in vitro, showing the disparities between in vivo and in vitro experiments within the field, as well as the influence of vesicle-producing cell maturation on the receiving cell. When B cells take up exosomes of mature DC, causes the priming of naïve T cells [36]. EVs of DCs contain different types of noncoding RNAs, which change in response to immune-stimulatory or immune-suppressive. These changes include Y-RNAs, small nucleolar RNAs (snoRNAs), and miRNAs which increased inside DCs in response to any stimulus. This demonstrates how the parent cell state may be reflected in the exosomes generated. This is not always the case, as evidenced by studies that examined large populations of RNA species detected in exosomes [37]. In the field of molecular biology, exosome-mediated miRNA transfer has gained special attention as a means of regulating response between cells, including immune cells. miRNAs, for example, may be shuttled between dendritic cells (DCs) and cause target knockdown in recipient cells, in addition, to conveying at the immunological synapse between T cells and DCs. Further, mice receiving exosomal miRNA have resistance to hepatitis B and aided IFN-α antiviral responses.

Previous studies have shown that DCs communicate with each other through exosomal miR-155, miR-146a, miR-148a, and miR-451. Exosomal miRNAs taken up by receipt cells cause suppression of target genes and can modify the cellular response to endotoxin, for example, miR-155 upregulates while miR-146a downregulates the expression of inflammatory genes [38]. Furthermore, activated bystander T cells can stimulate monocyte-derived DCs to produce vesicle miR-155, resulting in further T cell activation [39]. Alexander M. et. al. found that exosomes of DC contain miR-146a, an anti-inflammatory miRNA, and miR-155, a pro-inflammatory miRNA, taken up by receipt DCs in vitro resulting in modifying their target genes SHIP1, BACH1, Traf6, and IRAK-1 [38]. Hematopoietic cells can also transfer specific miRNAs between themselves, changing their responses. Monocytes can be differentiated into immature DCs and secrete IL-12P70 after receiving mature-derived EVs. These EVs-derived immature DCs play a role in the skewing of T cells, especially for Th1 rather than Th2, this capability is lost after the maturation of EVs-derived DCs.

Exosomes mediate communication between DCs as well as between other cells and DCs. For instance, when DCs receive exosomal miRNA-155 of tumor cells, leading to antigen triggering. Also, in the plasma of patients with systemic lupus erythematosus (SLE) activation of plasma cell-like DC and production of proinflammatory cytokines and IFN-α can be induced by TLR7 endogenous ligands within exosomal delivered miRNA. Therefore, miRNAs may represent new pathogenic mediators of autoimmune reactions as well as possible therapeutic targets for type I interferon-mediated illnesses [40]. When immature and mature DCs differentiated into a tolerogenic DC population by endotoxin-treated mesenchymal stem cell (MSC)-derived exosomes, resulting in a dropping in the number of DCs surface markers and proliferation of lymphocytes and release of IL-6, and increasing IL-10, TGF-β releasing. However, the limited targeting and specificity of exosome administration severely limit its usefulness. As a result, Li et al. developed an RNA delivery system based on MSC-derived exosomes capable of targeting particular DC binding and increasing HLA-G expression, with HLA-G inhibiting DC-triggered allogeneic T cell proliferation and eliciting long-term immunological tolerance [41]. Exosomal miR-21-5p and miR-223-3p derived mesenchymal stem cells (MSC) reduce migration and prevent maturation of DCs respectively by acting on the CD83 gene [42]. Exosomes from various origins can thus control DC development, maturation, and function, and they may be significant regulators of DC-induced immunological responses (Fig. 2a) and (Table 1).

Fig. 2
figure 2

a Dendritic cells (DCs) communicate with other DCs through the transfer of exosomal miRNAs, specifically miR-451, miR-148a, miR-155, and miR-146. Monocytes can differentiate into immature DCs, which then activate T cells upon receiving these miRNAs from DCs. b Macrophages play a role in regulating various cell types within the body through the transfer of exosomal miRNAs. For instance, M1-derived exosomal miR-155 regulates mesenchymal stem cells (MSCs) and M1-derived exosomal miRNAs inhibit bone generation, while M1-derived exosomal miR-148a regulates angiogenesis in endothelial cells. On the other hand, M2-derived exosomal miRNAs, such as miR-378a, promote bone generation. Additionally, miR-148a induces wound healing in cardiomyocytes, and miR-21-5p stimulates fibrinogenesis in tendon cells

Table 1 The role of exosomal miRNAs mediating signal transduction between immune cells

Macrophages

Macrophages act against pathogens and boost wound healing throughout the body, as part of the innate immune system. They are found in both lymphoid and non-lymphoid tissues. Macrophages are categorized based on their basic function and activation. Macrophages can be classified as traditionally activated (M1) macrophages (known as pro-inflammatory), wound-healing macrophages (also known as alternatively activated or anti-inflammatory (M2) macrophages), and regulatory macrophages (Mregs). The M2 macrophages are further sub-categorized into M2a, M2b, M2c, and M2d subtypes. Therefore, M2 macrophage-derived exosomes can be used as a therapy to decrease pro-inflammatory macrophage phenotypes, that most abundant in several diseases such as obesity and atherosclerosis [43]. Ruibing Yang et al. studied the roles of exosomes derived from M2a, M2b, and M2c macrophage phenotypes in colitis triggered by dextran sulfate sodium (DSS). They found that M2b macrophage-derived exosomes significantly reduced the severity of colitis in mice induced by DDS. This is due to M2b macrophage-derived exosomes increasing the splenic number of T regulatory (Treg) cells and the level of IL-4 in mice, while the levels of major cytokines linked to colitis (IL-1, IL-6, IL-7A) were dramatically decreased [44].

Laura Bouchareychas et al. found that polarization of exosomes derived from bone marrow-derived macrophage-derived with IL-4 causes inflammation suppression by targeting tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa B (NF-κB) through miR-146b, miR-99a, and miR-378a in the recipient macrophage [45]. Mesenchymal stem cells (MSCs)-derived exosomes have the ability to improve atherosclerosis in APOE−/− mice by decreasing the macrophage infiltration in the plaque through the miR-let7/ IGF2BP1/PTEN pathway. Further, MSCs-exosomes increased M2 macrophage polarization in the plaque via the miR- let7/HMGA2/NF-κB pathway [46]. It was found that miR-155 in M1 macrophage EV reduces the MSC osteogenic differentiation, while miR-378 in M2 macrophage EV increased MSC osteoinductive gene expression. This confirms the role of M2 macrophage in the bone healing process [47]. Also, some studies showed that miR-21-5p in macrophage-derived exosomes enhanced fibrogenesis in tendons through the change in the Smad7 signaling pathway after injection into mice. In cardiomyocytes, exosomal miR-148a of M2 macrophage modulates the transcription factor thioredoxin-interacting protein (TXNIP) to allow better response to myocardial injury, which is essential to myocardial survival [48].

There are numerous pathways by which tumor-associated macrophages (TAMs) contribute to tumor production, development, metastasis, and immune escape. In tumor cells, exosomes are able to modulate immune responses. Exosomes derived from tumors can induce macrophage polarization, which may result in the activation of anti-inflammatory pro-tumorigenic M2 immune cells or anti-inflammatory antitumorigenic M1 immune cells, alter the M1/M2 ratio in the tumor microenvironment, and enhance tumor growth [49]. Adriamycin-resistant breast cancer cells produce exosomal miR-222, which targets Phosphatase and Tensin Homolog (PTEN) gene, and activates the Akt pathway, leading to macrophage polarization to M2 and tumor progression [50]. Oxidative phosphorylation increases in macrophages in response to let-7a exosomal miR released by the hypoxic tumor cells, leading to the promotion of M2 macrophages and suppression of insulin-Akt-mammalian target of rapamycin (mTOR) signaling pathway [51, 52] (Fig. 2b) and (Table 1).

The exosomal miR-23a-PTEN-AKT pathway could suppress T-cell function via an endoplasmic reticulum stress response in hepatocellular carcinoma cells (HCC) by releasing exosomal miR-23a-3p and upregulating PD-L1 expression in macrophages, according to Liu et al. [53]. Previous studies showed that exosomes have a pivotal role in the polarization of macrophages from one phenotype to another. Holder et al. demonstrated that the human placenta internalizes secreted exosomes from macrophages via the clathrin-mediated endocytosis process. This results in the modulation of proinflammatory cytokine production in the placenta, including IL-6, IL-8, and IL-10. In this way, maternal immune cells communicate with the placenta via exosomes, which may facilitate responses to maternal inflammation and infection, thereby preventing harm to the fetus [54].

T lymphocytes

T cells have a pivotal role in the adaptive immune system by protecting against infection and cancer, but they are also involved in many immunological diseases. T lymphocytes have different types; the first type is the naïve T cell that matured and is released by the thymus but has not yet encountered its corresponding antigen, so it is called in the stage between maturity and activation. Each naïve T cell has a particular T cell receptor that identifies a specific antigen [55]. The second type is CD8 + T cells (cytotoxic T cells), which have the ability to attack infected cells and kill them using granule sacs that contain digestive enzymes. The third type is CD4 + T cells (helper T cells) which activate CD8 + T cells, macrophages, and increment antibody production by B cell lymphocytes, this process is called immunological response [56]. The fourth type is Regulatory T cells (Tregs) which lower the immune responses when a highly active response is no longer required by monitoring B and T cell functions. The fifth type is Natural Killer T cells which discriminate between infected and malignant cells and target cells that lack molecular markers that identify them as bodily cells. The last one is Memory T cells which give lifelong protection against some diseases by protecting against previously encountered antigens [57].

Exosomes have an immunomodulatory effect on lymphocytes such as regulation of their proliferation, activation, chemotaxis, differentiation, and mediation of diseases. CD8 + T cells induced by tumor-derived exosomes develop a suppressor phenotype (SP), which may act synergistically with exosomal proteins and RNA, making immunosuppressive exosomes a potential therapeutic target [58]. Exosomes derived from Pancreatic cancer (PC) are taken up by T lymphocytes, resulting in a change of gene expression and T lymphocytes exhibit cytotoxic activity after exosome treatment. This is due to PC-derived exosomes that induce Endoplasmic Reticulum (ER) stress-mediated apoptosis of T lymphocytes through p38 MAPK [59, 60]. When a thyroid cell-derived exosome containing TPO, HSP60, and MHC-II has exposed to IFN-γ activated dendritic cells in vitro, causes releasing of proinflammatory cytokines IFN-g, IL-17A, and IL-22 by CD4 + T lymphocytes, while inhibiting IL-4, IL-10, and TGF-b1 expression and release, ultimately suppressing thyroiditis [61].

Exosomes have a critical role in cellular communication during immune synapsis (IS) via the transfer of miRNA. María Mittelbrunn et.al. found that transfer of miRNA-335 of T cells-derived exosome to APCs, in a unidirectional manner, during immune synapses downregulates translation of SRY-Box Transcription Factor 4 mRNA (SOX-4 mRNA), also, exosomal miR-92a level increase in Raji cells after conjugation with T lymphoblasts [62]. Lola Fernández-Messina et.al found that directional transfer of T-cell exosomal miRNA (mmu-miR-20a-5p, mmu-miR-25-3p, and mmu-miR-155-3p) to the B-cell upon immune synapse resulting in B cell function control, proliferation, survival, regulation, and antibody production, by targeting BIM and PTEN [63]. Sometimes T cell exosomes have a cytotoxic effect, for example, Claudine Guay et. al. discovered the transfer of exosomal miRNA (miR-142-3p, miR-142-5p, and miR-155) released by T cells to β cell, insulin-secreting cells, induce apoptosis in type 1 diabetes. This is due to the induction of Ccl2, Ccl7, and Cxcl10 in the pancreatic β cells [64].

Some immune cells, including T helper 1 (Th1) cells, were suppressed by transferring miRNAs of Treg cells, which inhibited the proliferation and release of cytokines by Th1 cells. In colitis and autoimmunity, they found that Treg-cell-derived Let-7d-containing exosomes target Cox-2 in Th1 cells leading to decreasing INF-γ, Therefore, lethal Th1-cell-mediated inflammation is prevented [65, 66]. Also, Tregs produce interleukin-35 + EVs that act in trans on bystander T cells, causing exhaustion and suppressing immunity. Transferring of exosomal miR-150-5p and miR-142-3P derived Tregs cells to DCs causes tolerogenic phenotype, with increased IL-10 and reduced IL-6 secretion [67]. Deng, J.-n. et. al. found that hsa-miR-7-5P derived sepsis exosomes have an antiapoptotic effect on T lymphocytes by targeting the proapoptotic gene Bad in the cGMP-PKG signaling pathway [68]. Exosomal miRNA (hsa-miR-24-3p, hsa-miR-891a, hsa-miR-106a-5p, hsa-miR-20a-5p, and hsa-miR-1908) derived-nasopharyngeal carcinoma (NPC) mediate T-cell dysfunction by targeting the down-regulation of the MAPK1 and JAK/STAT pathways [69]. Exosomal miR-125a-3p derived MSC can suppress the differentiation of effector T cells (CD4 + and CD8 +), and improve the survival of CD4 + , CD25 + , Foxp3 + Tregs [42]. Natural killer cell secrets exosomal miR-10b-5p, miR-92a-3p, and miR-155-5p, which target GATA3, and TBX21 leading to Th1 polarization and IFN-γ and IL-2 production [70] (Fig. 3a) and (Table 1).

Fig. 3
figure 3

Immunomodulatory Effects of Exosomal miRNA on Lymphocytes. a Exosomal miRNA-derived T cells: miR-150-5p and miR-142-3p increase the level of IL-10 and decrease IL-6 production by dendritic cells (DCs). Treg-derived miR-let-7d regulates Th1 cells, and miRNA derived from T cells also regulates B cell functions. b Exosomal miRNA-derived B cells influence the function of DCs, especially when the B cells are infected with Epstein-Barr virus (EBV). These infected B cells secrete exosomes containing EBV-derived miRNAs

B lymphocyte

B cells play an important role in the humoral immunity component of the adaptive immune system. It has many functional roles such as producing antibodies, presenting antigens (antigen-presenting cells (APCs)), and secreting cytokines. B cells differ from T cells that have receptors on their cell membrane known as B cell receptors (BCRs). Through very specific BCRs, B cells bind to the foreign antigens and produce antibodies. Saunderson et al. found that B cells secrete exosomes upon CD40/IL-4 triggering. Exosomes-derived B cell express on their surface high levels of MHC class I, MHC class II, and CD45RA (B220), also, the BCR complex which contains surface Ig, CD19, and the tetraspanins CD9 and CD81 [71]. It is important to note that miRNAs found in exosomes are not always derived from the host genome. Pegtel et al. found that viral (EBV) miRNA secreted by EBV-infected B cells are transferred, by exosomes, to and act in noninfected recipient B cells. These EBV-miRNAs are functional by repression of their target genes including CXCL11/ITAC [72]. Liao et al. found that hepatocyte-derived exosomal miRNA (miR-122, let-7b, and miR-206) could upregulate B-cell activating factor (BAFF) expression in macrophages through Toll-Like Receptor 7 Polymorphism (TLR7) activation [73] (Fig. 3b).

Mast cells

Mast cell, also known as mastocyte or labrocyte, is a part of the immune and neuroimmune systems, it has many functional roles in allergy (contains a large amount of histamine and heparin granules), anaphylaxis, wound healing, vascular permeability in tumors of brain, and angiogenesis. Exosomes derived from MCs are capable of activating B and T lymphocytes to coordinate pro- and anti-inflammatory processes. Li et al. showed that exosomal miRNA-223-derived mast cells (MCs) increased the permeability of intestinal epithelial cells and destroyed the function of intestinal barrier proteins such as tight junction proteins 1 (TJP1, ZO-1), Occludin (OCLN), Claudin 8 (CLDN8) [74]. Inflammation of the central nervous system (CNS) is a response to external stimuli, including surgery, infection, and toxins, that reveals itself partly in the activation of microglia and the release of proinflammatory cytokines. Hu, L. et.al. showed that exosomal miR-409-3P derived-MCs have a role in neuroinflammation by activation of its target, Nr4a2, in the NF-κB pathway resulting in migration of microglial [75].

In addition, MCs-derived exosomes have a role in the migration and invasion of hepatocellular carcinoma (HCC) cells. It was found that exosomal miR-490 derived-MCs can inhibit tumor metastasis in HCC by dropping the activity of the EGFR/AKT/ERK1/2 pathway [76]. For regulation of tumor cell proliferation, it was shown that tryptase secreted from MCs binds to DNA present on the surface of melanoma cell-derived exosomes, resulting in degradation of lamin B (cause nuclear remodeling), hnRNP A2/B1, suppression of SNORA55 and RNU-2 expression, upregulation of MIR16–2 expressions, and downregulation of EGR1, all of these affect the gene expression and decreased proliferation [77, 78] (Fig. 4b) and (Table 1).

Fig. 4
figure 4

miRNA and Polymorphonuclear Cells: a Neutrophils communicate with different cells through exosomal miRNA, which affects their functions. For example, miR-142-3p targets PKCA in macrophages, leading to an increase in TNFα production. In the case of diffuse systemic sclerosis (dSSc), neutrophils secrete various exosomal miRNAs that are either downregulated or upregulated. b Mast cell-derived exosomes modulate the function of other cells. For instance, miR-409-3p derived from mast cells promotes microglial migration by targeting Nr4a2. Additionally, miR-223 derived from mast cells increases the permeability of intestinal epithelial cells by targeting TJP1, ZO-1, OCLN, and CLDN8. Lastly, miR-490 derived from mast cells inhibits metastasis in hepatocellular carcinoma by targeting the EGFR/AKT/ERK pathway. c Eosinophils release exosomes that can be taken up by other eosinophils, resulting in the production of reactive oxygen species (ROS) and nitric oxide (NO). Furthermore, miRNAs derived from eosinophils can activate dendritic cells (DCs)

Eosinophils

Eosinophils are one of the immune system components, they play a role in fighting viral infections and helminth, also, implicated in antigen presentation to cells. Furthermore, eosinophils are important mediators of allergic responses and asthma pathogenesis. Exosomes have an effective role in the pathology of asthma as well as other inflammatory diseases. The exosomes-derived eosinophils are able to stimulate eosinophils for the production of reactive oxygen species (ROS) and nitric oxide (NO) this process is called the autocrine effect [79] (Fig. 4c). Mazzeo et al. was the first group showed that eosinophils have MVBs and release exosomes which raised in asthmatic patients [80]. They described how the eosinophil exosomes damage the airways in the case of asthma disease. Bronchial smooth muscle cells (BSMC) and small airway epithelial cells (SAEC) structurally remodel the airways and regulate respiratory function and immunity. Exosomes derived- asthmatic eosinophils change the behavior of SAEC by increasing their apoptosis, decreasing their wound-healing capacity, and increasing their expression of CCL26, TNF-α, and POSTN. Furthermore, asthmatic exosomes modify the performance of BSMCs, increasing their proliferation and upregulating CCR3 and VEGFA gene expression through ERK1/2-phosphorylation [81].

The differential expression of 14 miRNAs between asthmatic and healthy eosinophils was validated by previous studies. There are multiple genes associated with inflammation are directly or indirectly regulated by miRNAs such as miR-16 downregulates (tumor necrosis factor-alpha) TNFα; the miR-17 ~ 92 cluster modifies PTEN; miR-155 alters PU.1, c-Maf, and SOCS1; and miR-146a regulates TRAF6 and IRAK1. There are other miRNAs associated with the pathology of asthma and allergies, such as miR-21, which targets IL-12p35 in steroid-resistant asthma; miR-1248, which targets IL-5; and miR-15a, which inhibits VEGFA in CD4 + cells. miR-185-5p expression is a good biomarker of asthma disease severity, as it has a role in the sensitivity of corticosteroids by suppressing the mammalian target of rapamycin complex signaling pathway (mTORC) so miR-185-5p can restore sensitivity in CEM-C1 cells which are steroid-resistant. Also, miR-185-5p has a role in cell growth by targeting CDC42 and RHOA directly and VEGFA indirectly. Finally, miR-185 is involved in calcium signaling by targeting NFAT and CaMKII in myocardial cells during cardiac hypertrophy [82]. Gao et al., showed that exosomal miR-100-5p derived from human umbilical cord mesenchymal stem cell (hUCMSC) has an important role in atherosclerosis alleviation in ApoE−/− mice by repressing cell migration, inducing apoptosis, and preventing inflammatory response in eosinophils through the FZD5/Wnt/β-catenin pathway [83].

Neutrophil

Neutrophils (also called neutrocytes, heterophils, or polymorphonuclear leukocytes) are the most common form of granulocyte, accounting for 40 to 70% of all white blood cells in humans. It is estimated that 40 to 70% of human granulocyte white blood cells are neutrophils (polymorphonuclear leukocytes). They have an essential role in the innate immune system, they are present in blood circulation as phagocytes. The first (acute) phase of inflammation occurs when neutrophils migrate into the inflammatory site, especially when bacteria infect, environmental toxins are present, and certain cancers are present. Neutrophils move to blood arteries and then into interstitial space following chemical cues such as interleukin-8 (IL-8), C5a, fMLP, Leukotriene B4, and H2O2 this process is called chemotaxis. In response to the immune complex, neutrophils release multiple molecules outside the cells, such as proteases, cytokines, reactive oxygen species, and exosomes. Neutrophils also collaborate with macrophages, dendritic cells, B and T cells, and natural killer cells (NKs) to regulate the immune response by producing IFN-γ. Neutrophil-derived EXOs (neutrophil EXOs) also play important roles in immune responses. The neutrophil EXOs are capable of modulating immune responses, increasing smooth muscle cell proliferation in the lungs, and promoting reorganization of the airways in asthmatic patients [84]. Linhares-Lacerda et al. exhibited that miR-142-3p is transported from neutrophil extracellular traps (NETosis) to macrophages where it down-regulates protein kinase C alpha (PKCα) resulting in the neutralization of the excessive production of TNFα. Also, Different methods of NETosis induce different expression levels of miRNAs [85].

Recent studies have revealed that neutrophils can also functionally polarize, determining selective activity patterns related to different diseases. Neutrophils are classified into two well-defined types, known as pro-inflammatory (immunostimulating) N1s and suppressor (immunosuppressive) N2s. Tyagi et al. showed that the SOX2/CPT1A axis promotes tumor cell metabolic switching via exosomal miR-4466 secreted from nicotine-induced N2-neutrophils [86]. Li et al. identified the profile of exosomal miRNAs-derived neutrophils in diffuse cutaneous systemic sclerosis (dSSc). This profile includes downregulated miRNAs (such as hsa-miR-299-3p, hsa-miR-1323, hsa-miR-520a-3p, hsa-miR-512-3p, and hsa-miR-4755-3p) and upregulated miRNAs (such as hsa-miR-3614-5p, hsa-miR-323a-3p, hsa-miR-95-3p, hsa-miR-1260b, hsa-miR-1248, hsa-miR-1260a, hsa-miR-1268a, and hsa-miR-218-5p). The target genes of exosomal miRNAs-derived neutrophils were enriched in Wnt and AMPK signaling pathways. The expression of hsa-miR-218-5p and hsa-miR-144-3p were significantly different between anti-scl70 (−) and anti-scl70 ( +) groups from dysregulated miRNAs [87].

Occludin (OCLN) coded transmembrane proteins play a critical regulatory role in protein tight junction formation and cell permeability barrier and are essential components of the blood–brain barrier. Li et al. found that exosomal miR-122-5p derived lipopolysaccharide (LPS)-induced neutrophils promote oxidative stress, and apoptosis and increase the permeability of brain microvascular endothelial cells (BMECs) by the down-regulation of OLCN expression [88]. Polymorphonuclear neutrophils (PMNs) play a crucial role in the pathogenesis of sepsis-related acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Jiao et al. It was found that exosomal miR-30d-5p derived PMNs induce macrophage M1 polarization and primed macrophage pyroptosis by activating NF-kB signaling by targeting SOCS-1 and SIRT1 [89]. Exosomal miR-223-derived neutrophil represses the canonical NF-κB pathway by downregulating the expression of components in the signaling transduction pathway [90]. Furthermore, exosomal miR-223 derived from neutrophils suppresses inflammasome activation and the production of inflammatory cytokines by directly targeting STAT1, STAT3, and NLRP3. In cancer cells, it also regulates genes involved in cell proliferation, survival, differentiation, immune evasion, cell adhesion, and migration [90] (Fig. 4a) and (Table 1).

Tumor microenvironment

Tumors are heterogeneous tissues of cancer cells, infiltrating and resident host cells, secreted factors, and extracellular matrix. The growth and progression of tumors are facilitated by the molecular, cellular, and physical changes that tumor cells cause within their host tissues. The tumor microenvironment (TME), known as the cancer microenvironment, is a complicated and continuously evolving entity. The TME components are heterogeneous and depend on the type of tumor, but the most common include endothelial cells, pericytes, fibroblasts, immune cells, stromal cells, blood vessels, and extracellular matrix. Truffi et al. suggested that the "tumor microenvironment is not merely a mute bystander, but rather an active supporter of cancer growth" [91]. In the early stages of tumor development, cancer cells, and TME components develop a dynamic relationship that enables the survival, infiltration, and metastatic spread of cancer cells. As a means of restoring oxygen and nutrient supply and removing metabolic waste, the TME promotes angiogenesis to overcome a hypoxic and acidic microenvironment [92]. As tumors grow, they become infiltrated with diverse immune cells, including adaptive (including T cells, B cells, and natural killer (NK) cells) and innate (macrophages, neutrophils, and dendritic cells) cells, which can both promote tumor growth and inhibit it [93]. The most prevalent mechanism of tumor formation is chronic infection-induced persistent inflammation in the case of colorectal, hepatocellular, and cervical cancers.

As mentioned before, TME is characterized by hypoxia compared to the normal internal environment, this is due to the release of matrix metalloproteinases (MMPs), hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and other stimulating factors [94]. TME remodeling creates a niche for tumor cells to interact with neighboring fibroblasts, endothelial cells, and immune cells [95]. This interaction triggers a multitude of biological processes for tumor progression, including appreciation, migration, angiogenesis, immunosuppression, and drug resistance [96]. miRNAs regulate diverse biological functions through the regulation of gene expression, including proliferation, apoptosis, differentiation, migration, invasion, and drug resistance. Changes in genetics or epigenetics can lead to abnormal expression of miRNAs in cancer cells, resulting in abnormal gene expression [97, 98]. miRNAs suppress gene expression at the protein level by interacting with complementary target mRNA at 6–7 bases. Previous studies showed that miRNAs can trigger the development of TMEs and biological alterations by acting as oncogenes. Normal fibroblasts (NFs) within the tumor microenvironment (TME) undergo transformation into cancer-associated fibroblasts (CAFs) through the actions of miR-9, miR-200 s, miR-526b, and miR-655 which enhance angiogenesis and lymphangiogenesis within the TME. Additionally, miR-340-5p and miR-561 contribute to the promotion of an immunosuppressive microenvironment. Exosomes, which transport proteins, metabolites, and nucleic acids (including miRNAs), play a crucial role in mediating communication between tumor cells and the TME. Consequently, exosomes are the primary modulators responsible for the heterogeneity of the TME and exert significant influence over tumor development.

TME and immune cells

Over the years, there has been a growing recognition of the importance of communication between cancer cells and immune cells, leading to the inclusion of this interaction as one of the emerging hallmarks of cancer by 2011 [99]. It is now well-established that the tumor microenvironment (TME) comprises cancer cells and various immune cells, stromal cells, endothelial cells, and cancer-associated fibroblasts. Cancer cells employ diverse mechanisms to evade immune surveillance and evade destruction. Consequently, several immunotherapy approaches have been developed and applied in clinical practice over the past decades, aiming to counteract these immune evasion mechanisms. Unlike traditional chemotherapy, immunotherapy primarily leverages the immune cells within or outside the TME to selectively recognize and attack cancer cells. This theoretically grants immunotherapy higher specificity and lower side effects [100].

The immune cells within the tumor microenvironment (TME) exhibit diverse functions. For instance, tumor-antagonizing immune cells, such as effector T cells, kill target cells through granule exocytosis and FasL-mediated apoptosis induction. They also secrete IFN-γ and TNFα, which induce cytotoxicity in cancer cells. NK cells contribute to the tumor-killing response by releasing perforin and granzymes to induce apoptosis in target cells. They also secrete pro-inflammatory cytokines and chemokines (such as IFN-γ, TNF, IL-6, GM-CSF, and CCL5) to promote anti-tumor activity [101]. DCs present antigens, provide costimulatory signals for T-cell activation, and interact with NK and B cells [102]. M1-polarized macrophages generate pro-inflammatory cytokines and reactive oxygen/nitrogen species to eliminate tumor cells [103], while N1-polarized neutrophils release granules containing antimicrobial and cytotoxic compounds to destroy cancerous cells. Additionally, these immune cells secrete cytokines and chemokines to attract other cells with anti-tumor activity [104].

On the other hand, immune cells that promote tumor growth include regulatory T cells, which hinder the effective response of effector T cells against cancer cells. MDSCs (myeloid-derived suppressor cells) enhance angiogenesis by producing MMP9, prokineticin 2, and VEGF, promoting the migration of cancer cells toward endothelial cells and facilitating metastasis [105]. There is controversy surrounding the role of B cells. On one hand, they can produce cytokines that coordinate with cytotoxic T lymphocyte (CTL) activity and act as potent antigen-presenting cells (APCs). However, B cells may also possess a protumorigenic potential by producing cytokines that recruit MDSCs and enhance angiogenesis [106]. They can inhibit CTL function as well.

TME and exosomal miRNAs

As the tumor progresses, exosomal miRNAs produced by primary tumor cells are transferred to non-malignant cells in the TME to promote heterogeneity [107]. While non-malignant cells can release exosomal miRNAs as a result of changing their biological activities in the TME, non-malignant cells can also secrete miRNAs to further regulate tumor cells [108]. Most previous studies have identified cancer-associated fibroblasts (CAFs), endothelial cells, and immune cells as the stromal cell receptors of cancer-derived exosomal miRNAs. Within the TME, the impact of exosomal miRNAs is particularly evident in the activation of CAFs, which remodel the extracellular matrix (ECM) and promote tumor cell dissemination. In response to exosomal miRNAs, Endothelial cells facilitate the formation of tube networks that enhance tumor cell metabolism and survival. Furthermore, exosomal miRNAs contribute to the invasion of inflammatory cells and aid in immune evasion, thereby promoting tumor colonization and proliferation. Considering these significant effects, exosomal miRNAs play a role in facilitating tumor development within the TME [109, 110].

Enhancing growth and metastasis through angiogenesis: Tumor development is highly reliant on cancer cell metabolism. A tumor with irregular blood vessel distribution and abnormal vascular function results in local hypoxia and reduced nutrition supply. Simultaneously, the distance gradient between various vascular beds causes an imbalance in medication distribution and absorption [111]. These vascular network modifications encourage the establishment of an internal microenvironment and intratumoral heterogeneity. The extensive vascular network in TME promotes cancer cell growth and metastasis. Blood vessels in the microenvironment can be modified by exosome-bound miRNAs, which are taken up and used by vascular endothelial cells. Exosomal miRNAs-derived tumor cells have been shown to induce angiogenesis in TME.

For instance, miR-23a, originating from nasopharyngeal carcinoma (NPC), exerts a crucial role in promoting angiogenesis by selectively targeting TSGA10. This targeting facilitates the phosphorylation of p-ERK and modulates the growth, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) [112]. In glioma, both miR-21 and miR-9 induce angiogenesis by activating the VEGF/p-FLK/VEGFR2 and MYC/OCT4 pathways, respectively [113, 114]. Similarly, in TGF-β1 lung cancer, miR-619-5p and miR-142-3p contribute to angiogenesis by inhibiting RCAN1,4 and PDGFR-β, as well as p-SMAD2/3, respectively [115, 116]. This angiogenic phenomenon extends to other miRNAs derived from hepatocellular carcinoma (HCC), such as miR-210-3P, which targets SMAD4 and STAT6, as well as miR-100 derived from mesenchymal stem cells (MSCs), which induces angiogenesis in breast cancer by targeting the mTOR/HIF-1α/VEGF signaling axis [117, 118]. Additionally, miR-126a, derived from myeloid-derived suppressor cells (MDSCs), enhances angiogenesis by activating inflammatory circuits and increasing IL-13 and IL-33 levels [119]. In ovarian cancer, miR-205 promotes angiogenesis by targeting the PTEN/AKT pathway, while miR-141-3p derived from the human ovarian carcinoma cell line SKOV-3 activates the JAK-STAT3 pathway in endothelial cells to induce angiogenesis [120, 121]. Exosomal miR-205-5p from NPC promotes angiogenesis and NPC metastasis by targeting DSC2 to enhance the EGFR/ERK signaling and MMP expressions [122]. Exosomal miR-9-3p from Müller glia cells enhances retinal angiogenesis by targeting the S1P1/AKT/VEGFR2 pathway in diabetic retinopathy [123].

Furthermore, exosomal miR-25-3p, originating from colorectal cancer, exhibits the ability to downregulate KLF2 and KLF4 expression. KLF2 engages the VEGFR2/p-Erk/p-Akt pathway, influencing the tube formation ability of HUVECs, whereas KLF4 activates the ZO-1/Occludin/Claudin5 pathway, leading to modifications in the vascular microenvironment, including vascular permeability, adhesion, and the formation of a circular structure in aortic rings [124]. Additionally, miR-23a derived from lung cancer promotes angiogenesis by inhibiting prolyl hydroxylase 1 and 2 (PHD1 and PHD2), resulting in HIF-1α accumulation in endothelial cells and increased permeability of vascular trans-endothelial migration of cancer cells by inhibiting ZO-1 [125]. In contrast, miRNAs derived from human liver stem cells (HLSC), including miR-15a, miR-181b, miR-874, and miR-320c, exert an anti-tumorigenic effect by suppressing tumor angiogenesis through the downregulation of target genes such as ITGB3, FGF1, EPHB4, and PLAU [126]. Also, miR-451a derived from HCC inhibits tube formation and vascular permeability of HUVECs through targeting LP1N1 [127]. Moreover, miR-9 derived from NPC demonstrates anti-angiogenic effects by up-regulating MDK and activating the PDK/Akt signaling pathway, thereby inhibiting the formation of endothelial cells [128]. The mechanisms of inducing angiogenesis by exosomal miRNAs are summarized in (Fig. 5) and (Table 2).

Fig. 5
figure 5

Mechanisms of Exosomal miRNA Regulation of Angiogenesis in Tumor Microenvironments. Exosomal miRNAs taken up by endothelial cells in the tumor microenvironment can affect angiogenesis. For example, in nasopharyngeal carcinoma (NPC), miR-23a-derived exosomes promote angiogenesis by targeting TSGA10 and activating the phosphorylation of p-ERK. In glioma, miR-21 and miR-9 induce angiogenesis by activating the VEGF/p-FLK/VEGFR2 pathway and MYC and OCT4, respectively. In non-small lung cancer, miR-619-5p and miR-142-3p promote angiogenesis by inhibiting RCAN1,4 and PDGFR-β, and p-SMAD2/3, respectively. Similar mechanisms apply to other types of cancer. For instance, in hepatocellular carcinoma (HCC), mesenchymal stem cells (MSCs), myeloid-derived suppressor cells (MDSCs), and ovarian cancer, miRNAs such as miR-210-3p, miR-100, miR-126-a, miR-205, and miR-141-3p promote angiogenesis, while miRNAs derived from hepatic lineage stem cells (HLSC) like miR-15a, miR-181b, miR-874, and miR-320c inhibit angiogenesis. *( +): increase, (−): decrease

Table 2 The effect of exosomal miRNAs in the tumor microenvironment (TME)

Immunomodulatory effect: Immune cells such as lymphocytes, dendritic cells, and macrophages invade tumor tissues and surrounding locations frequently in the TME. Tumor cells can suppress immune cell maturation and differentiation via numerous signal transduction pathways controlled by exosomal miRNAs, so producing an immunological milieu favorable for tumor development. At the same time, tumor cells frequently secrete metabolic by-products such as lactic acid, nitric oxide, reactive oxygen species, prostaglandins, and arachidonic acid in hypoxia and low nutrient supplies in the microenvironment, leading to the formation of an inflammatory microenvironment. In the tumor microenvironment, tumor cells (outbreak, escape) from immune surveillance due to the changes in the biological activity of numerous immune cells and the generation of inflammatory mediators [129].

Tumor-associated macrophages (TAMs) are essential components of the tumor microenvironment (TME), derived from circulating monocytes recruited to the tumor site. TAMs exhibit a complex and diverse role in tumor progression, displaying both pro-tumorigenic and anti-tumorigenic functions depending on their polarization state and the specific context of the TME. Pro-tumorigenic functions of TAMs involve tumor promotion by secreting growth factors, cytokines, and chemokines that enhance tumor cell proliferation, survival, and migration. They also promote angiogenesis through the secretion of pro-angiogenic factors, facilitating the formation of new blood vessels to support tumor growth and metastasis [130]. Additionally, TAMs contribute to immune suppression by releasing immunosuppressive factors that inhibit the activity of cytotoxic T cells and natural killer (NK) cells [131]. Conversely, TAMs exert anti-tumorigenic effects through tumor surveillance, participating in the recognition and elimination of tumor cells via phagocytosis and antigen presentation, thereby contributing to immune surveillance and tumor control. TAMs are also involved in tissue remodeling and wound healing by facilitating debris clearance and promoting extracellular matrix remodeling. Moreover, TAMs can be polarized towards an anti-tumor phenotype known as M1-like TAMs, which secrete pro-inflammatory cytokines and activate immune responses against the tumor. The polarization state of TAMs is highly influenced by the TME, including signals from tumor cells, stromal cells, and soluble factors present in the microenvironment. The interplay between TAMs and other immune cells, such as T cells and natural killer cells, is crucial in determining the overall immune response against the tumor [128, 132].

From these miRNAs, miR-301a-3p derived from hypoxic pancreatic cancer cells induces M2 polarization of macrophages by targeting PTEN and activating the p-mTOR, p-AKT, and PI3Kγ signaling pathways [133]. Moreover, miR-222-3p from EOC (epithelial ovarian cancer) activates macrophages, leading to the formation of TAMs via the SOCS3/STAT3 pathway [134]. Additionally, miR-21-3p, miR-125b-5P, miR-181D-5p, and miR-940 secreted from hypoxia EOC induce M2 macrophage polarization resulting in tumor progression [135]. Furthermore, M2 polarization induced by miR-21 from human head and neck cancer increases MRC1 of M2, and miR-1246, which are derived from colon cancer increase IL-10, TGFβ, AND MMPs levels [136, 137]. These exosomal miRNAs play a pivotal role in modulating the immune microenvironment by mediating immunosuppression. However, exosomal miR-125-5p derived from melanoma enhances immunity by elevating the levels of M1 phenotype markers IL-1β, CCL1, CCL2, and CD80 through the targeting of LIPA [5]. Conversely, the internalization of exosomal miR-21 by CD14 + human monocytes suppress the expression of the M1 marker while simultaneously enhancing the expression of the M2 marker such as in the case of miR-21 from human head and neck cancer.

Dendritic cells (DCs) represent the most potent professional antigen-presenting cells within the human body. They play a crucial role in initiating T-cell activation and orchestrating the central immune response. The regulation of cross-presentation in dendritic cells, alongside intercellular communication mediated by tumor-derived and endogenous exosomal miRNAs, can impact the maturation of DCs and their interactions with other immune cells [35, 138]. From these exosomal miRNAs, miR-212-3p from pancreatic cancer specifically targets the MHC II TF RFXAP, resulting in a diminished expression of HLA-DR, -DP, and -DQ molecules [139]. This interference negatively impacts the function of DCs (dendritic cells). In addition, miR-203 downregulates TLR4 (Toll-like receptor 4) and downstream cytokines in DCs affecting the activation of natural killer cells [140]. The miRNAs released from regulatory T cells (Tregs) possess the ability to influence the immune response. Specifically, the transfer of exosomal miR-150-5p and miR-142-3p to DCs induces a state of cellular refractoriness. This phenomenon of internalizing leads to an upregulation of IL-10 expression and a concurrent downregulation of IL-6 expression [141]. Moreover, exosomal miR-let-7d is transferred to T helper 1 (Th1) cells, resulting in the inhibition of Th1 cell proliferation and the suppression of IFN secretion. Notably, IFN, which is secreted by Th1 cells, serves as a pivotal player in the context of anti-tumor immunity [65]. In the context of breast cancer, miR-let-7i promotes DC maturation through the reduction of TGF-β, SOCS1, and KLRK1 levels, and the increase of IL-6, IL-7, IFNγ, and IL-1b [142]. Glioblastoma secretes miR-10a, and miR-21 affects the activation and differentiation of MDSCs (myeloid-derived suppressor cells) by targeting RORA and PTEN, respectively [143]. Exosomal miR-155 derived from chronic lymphatic leukemia (CLL) induce the reprogramming of monocytes to MDSCs resulting in immune suppression [144], the immunomodulatory mechanism shown in (Fig. 6) and (Table 2).

Fig. 6
figure 6

The immunomodulatory effect of exosomal miRNA in tumor microenvironments: Exosomes released by primary tumor cells are absorbed by receptors of the immune cells, wherein the exosomal miR-212-3p derived pancreatic cancer targets MHC II TF RFXAP result in reducing the expression of HLA-DR, -DP, and -DQ molecules which interfere with DCs cells function, miR-203 downregulates TLR4 and downstream cytokines in DCs, and miR-301a-3p induced the M2 polarization of macrophages via targeting PTEN and activation of the p-mTOR, p-AKT, PI3Kγ signaling pathway. In breast cancer, miR-let-7i induces DC maturation by decreasing TGF-β, SOCS1, and KLRK1 levels and increasing IL-6, IL-7, IFNγ, and IL-1b. miR-22-3p derived EOC activates macrophages to a TAM through SOCS3/STAT3 pathway. Also, miR-21-3p, miR-125B-5P, miR-181D-5p, and miR-940 induce M2 macrophage polarization.M2 polarization induced by miR-21 derived human head and neck cancer secrets miR-21 and miR-1246 derived colon cancer. DCs took up miR-142-3p, miR-150-5p derived Treg resulting in induction of a tolerogenic phenotype in these cells, with increased IL-10 and decreased IL-6 production, while miR-let-7d derived Treg inhibit Th1 proliferation and IFNγ. Glioblastoma secretes miR-10a and miR-21, which affect the activation and differentiation of MDSCs by targeting RORA and PTEN, respectively. These exosomal miRNAs modulate the immune microenvironment by mediating immunosuppression. While exosomal miR-125-5p-derived melanoma enhances immunity by increasing the level of M1 phenotype markers IL-1β, CCL1, CCL2, and CD80 via targeting LIPA. *( +): increase, (−): decrease

Tumor progress is activated by extracellular matrix remodeling: When malignant cells interact dynamically with their surrounding stroma leading to progress and growth of the tumor, this process is complicated. Fibroblasts represent the main source of connective tissues, ECM, and major cell types in the stromal [145]. The initiation and metastasis of cancer are suppressed by several inhibitory functions released by normal fibroblasts through direct communication between cells, paracrine signaling, and ECM integrity. Tumor-derived exosomal miRNAs have the capacity to initiate a series of tumor-promoting signals, triggering the transformation of normal fibroblasts (NFs) into cancer-associated fibroblasts (CAFs). This conversion stimulates the formation of new blood vessels, disrupts the homeostatic state of the extracellular matrix (ECM), and creates an optimal environment conducive to the proliferation and dissemination of cancer cells. These orchestrated effects contribute to the extensive progression of cancer within the affected tissues, shaping a landscape ideal for malignant growth [146]. The exosomal miRNAs play an important role in remodeling the ECM by changing the function of chondrocytes, osteoblasts, and certain epithelial cells. Therefore, this process affects angiogenesis, inflammatory response, and metabolic reprogramming.

The exosomes released by primary tumor cells were internalized by normal fibroblasts (NFs) through specific receptors. Within these exosomes, miR-124 derived from ovarian cancer, miR-27B-3P and miR-214-3P derived from myeloma, and miR-27a derived from gastric cancer target specific proteins, namely SPHK1, FBXW7, and CSRP2 [147,148,149]. Exosomal miR-10b derived from colorectal cancer inhibits PIK3CA, resulting in the activation of fibroblasts and their transformation into cancer-associated fibroblasts (CAFs) [150]. This transformation is characterized by an increase in the expression of α-SMA and FAP and plays a regulatory role in the migration and invasion of CAFs. In melanoma, miR-155 derived from the tumor inhibits SOCS1 and promotes the activation of the JAK2/STAT3 signaling pathway, increasing FGF2, VEGFA, and MMP9 levels in CAFs [151]. Similarly, in hepatocellular carcinoma (HCC), miR-21 derived from the tumor reduces PTEN levels, leading to the activation of the PDK1/AKT signaling pathway and an increase in the expression of VEGF, MMP2, MMP9, βFGF, and TGF-β in CAFs, thereby promoting angiogenesis [152, 153]. Additionally, exosomal miRNAs derived from breast cancer, such as miR-9, activate NFs to transform into CAFs by upregulating MMP1, EFEMP1, and COL1A1 [154]. Moreover, miR-105 induces metabolic reprogramming of CAFs by activating the MYC signaling pathway [155]. The mechanisms of ECM remodeling by exosomal miRNAs are summarized in (Fig. 7) and (Table 2).

Fig. 7
figure 7

Remodeling of ECM mechanism by exosomal miRNA. The primary tumor cells release exosomes which were taken up by the normal fibroblasts (NFs) receptors, wherein the exosomal miR-124 derived ovarian cancer, miR-27B-3P, miR-214-3P derived myeloma, and miR-27a derived gastric cancer target the proteins (SPHK1, FBXW7, CSRP2), while miR-10b derived colorectal cancer inhibits PIK3CA leading to activation of fibroblast to cancer-associated fibroblasts (CAFs) by increasing the expression of α-SMA, and FAP and regulates CAFs migration and invasion. miR-155 derived melanoma inhibits SOCS1and promote activation of FGF2, VEGFA, MMP9 in CAFs. miR-21 derived HCC decrease PTEN leading to activation of PDK1/AKT signaling pathway and increasing expression of VEGF, MMP2, MMP9, βFGF and TGF-β in CAFs, promoting angiogenesis. Exosomal miRNA-derived breast cancer such as miR-9 activates NFs to CAFs by increasing MMP1, EFEMP1, COL1A1, and miR-105 inducing metabolic reprogramming of CAFs by activating MYC signal

Conclusion

More than four decades, the groundbreaking discovery of exosomes within immature red blood cells by Stahl and Johnstone marked a pivotal milestone [17, 156, 157]. These small vesicles, known as exosomes, encapsulate a diverse cargo that reflects their cell of origin. Fascinatingly, exosomes possess the remarkable capability to transfer their contents between cells, endowing them with significant roles in both physiological and pathological conditions. Of particular interest within the exosomal cargo are microRNAs (miRNAs), which serve as key messengers in signal transduction between immune cells, including dendritic cells, macrophages, lymphocytes, neutrophils, mast cells, and non-immune cells. These miRNA-loaded exosomes orchestrate vital processes such as the control of dendritic cell development, maturation, and function, as well as the polarization of macrophages. In the context of colitis, exosomal miRNAs derived from regulatory T cells effectively suppress Th1 cells, illustrating the profound impact of miRNAs as crucial modulators of the immune system. Furthermore, exosomes emerge as the principal mediators responsible for the heterogeneous nature of the tumor microenvironment (TME). Through the transfer of their contents between tumor cells and the TME, exosomes facilitate critical adaptations of tumor cells to the low oxygen levels of the TME, promoting angiogenesis or facilitating metastasis to more favorable environments. Remarkably, exosomal miRNAs secreted by tumor cells actively reshape the extracellular matrix and potentiate tumor cell spread by activating cancer-associated fibroblasts. Consequently, exosomal miRNAs exert an immunomodulatory influence within the dynamic landscape of the TME. In conclusion, the captivating realm of exosomes and their cargo, particularly miRNAs, holds immense potential for understanding and manipulating intricate cellular interactions. These findings enhance our comprehension of fundamental biological processes and open up exciting avenues for therapeutic interventions in various pathological contexts.

Availability of data and materials

Not applicable.

Abbreviations

EVs:

Extracellular vesicles

MVBs:

Multivesicular bodies

ILVs:

Intraluminal vesicles

ESEs:

Early-sorting endosomes

LSEs:

Late-sorting endosomes

UTR:

Untranslated region

ORF:

Open reading frame

HDL:

High-density lipoprotein

pri-miRNA:

Primary miRNA

nSMase2:

Neural sphingomyelinase 2-dependent

hnRNP:

Heterogeneous nuclear ribonucleoprotein

RISC:

RNA-induced silencing complex

GW182:

Glycine-tryptophan protein of 182 kDa

DCs:

Dendritic cells

BM:

Bone marrow

PAMPs:

Pathogen-associated molecular patterns

LPS:

Lipopolysaccharide

BMDCs:

Bone marrow-derived DCs

snoRNAs:

Small nucleolar RNAs

MSC:

Mesenchymal stem cell

Mregs:

Regulatory macrophages

DSS:

Dextran sulfate sodium

Treg:

T regulatory

TNF-α:

Tumor necrosis factor-alpha

NF-κB:

Nuclear factor kappa B

TXNIP:

Thioredoxin-interacting protein

TAMs:

Tumor-associated macrophages

PTEN gene:

Phosphatase and tensin homolog gene

SP:

Suppressor phenotype

PC:

Pancreatic cancer

ER:

Endoplasmic reticulum

SOX-4 miRNA:

SRY-box transcription factor 4 mRNA

IS:

Immune synapsis

Th1:

T helper 1

APCs:

Antigen presenting cells

BCRs:

B cell receptors

BAFF:

B-cell activating factor

TLR7:

Toll-like receptor 7 polymorphism

OCLN:

Occludin

CLDN8:

Claudin 8

CNS:

Central nervous system

HCC:

Hepatocellular carcinoma

ROS:

Oxygen species

NO:

Nitric oxide

BSMC:

Bronchial smooth muscle cells

SAEC:

Small airway epithelial cells

hUCMSC:

Human umbilical cord mesenchymal stem cell

IL:

Interleukin

NKs:

Natural killer cells

PKCα:

Protein kinase C alpha

NETosis:

Neutrophil extracellular traps

dSSc:

Diffuse cutaneous systemic sclerosis

BMECs:

Brain microvascular endothelial cells

PMNs:

Polymorphonuclear neutrophils

ALI:

Acute lung injury

ARDS:

Acute respiratory distress syndrome

TME:

Tumor microenvironment

MMPs:

Matrix metalloproteinases

HIF-1α:

Hypoxia inducible factor-1α

VEGF:

Vascular endothelial growth factor

NFs:

Normal fibroblasts

NSCLC:

Non-small cell lung cancer

MDSCs:

Myeloid-derived suppressor cells

CLL:

Chronic lymphatic leukaemia

CAFs:

Cancer associated fibroblasts

SPHK1:

Sphingosine kinase 1

TAMs:

Tumor associated macrophages

HLSCs:

Human liver stem-like cells

HUVECs:

Human umbilical vein endothelial cells

PHD1 and PHD2:

Prolyl hydroxylase 1 and 2

NKs:

Natural killer cells

EOC:

Epithelial ovarian cancer

MDSCs:

Myeloid-derived suppressor cells

α-SMA:

α-Smooth muscle actin

FAP:

Fibroblast activating protein

References

  1. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Gao K, Jin J, Huang C, Li J, Luo H, Li L, Huang Y, Jiang Y. Exosomes derived from septic mouse serum modulate immune responses via exosome-associated cytokines. Fron Immunol. 2019;10:1560.

    CAS  Google Scholar 

  3. Gao G, Li C, Zhu J, Wang Y, Huang Y, Zhao S, Sheng S, Song Y, Ji C, Li C. Glutaminase 1 regulates neuroinflammation after cerebral ischemia through enhancing microglial activation and pro-inflammatory exosome release. Fron Immunol. 2020;11:161.

    CAS  Google Scholar 

  4. Chen C, Wang J, Sun M, Li J, Wang H-MD. Toward the next-generation phyto-nanomedicines: cell-derived nanovesicles (CDNs) for natural product delivery. Biomed Pharmacother. 2022;145: 112416.

    CAS  PubMed  Google Scholar 

  5. Gerloff D, Lützkendorf J, Moritz RK, Wersig T, Mäder K, Müller LP, Sunderkötter C. Melanoma-derived exosomal miR-125b-5p educates tumor associated macrophages (TAMs) by targeting lysosomal acid lipase A (LIPA). Cancers. 2020;12:464.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Xiao Y, Zhong J, Zhong B, Huang J, Jiang L, Jiang Y, Yuan J, Sun J, Dai L, Yang C. Exosomes as potential sources of biomarkers in colorectal cancer. Cancer Lett. 2020;476:13–22.

    CAS  PubMed  Google Scholar 

  7. Stoorvogel W, Strous GJ, Geuze HJ, Oorschot V, Schwartzt AL. Late endosomes derive from early endosomes by maturation. Cell. 1991;65:417–27.

    CAS  PubMed  Google Scholar 

  8. Huotari J, Helenius A. Endosome maturation. EMBO J. 2011;30:3481–500.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Bebelman MP, Bun P, Huveneers S, van Niel G, Pegtel DM, Verweij FJ. Real-time imaging of multivesicular body–plasma membrane fusion to quantify exosome release from single cells. Nat Protoc. 2020;15:102–21.

    CAS  PubMed  Google Scholar 

  10. Van Niel G, d’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.

    PubMed  Google Scholar 

  11. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev. 2014;30:255–89.

    CAS  Google Scholar 

  12. Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–25.

    CAS  PubMed  Google Scholar 

  13. Runz S, Keller S, Rupp C, Stoeck A, Issa Y, Koensgen D, Mustea A, Sehouli J, Kristiansen G, Altevogt P. Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM. Gynecol Oncol. 2007;107:563–71.

    CAS  PubMed  Google Scholar 

  14. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79.

    PubMed  Google Scholar 

  15. Baran J, Baj-Krzyworzeka M, Weglarczyk K, Szatanek R, Zembala M, Barbasz J, Czupryna A, Szczepanik A, Zembala M. Circulating tumour-derived microvesicles in plasma of gastric cancer patients. Cancer Immunol Immunother. 2010;59:841–50.

    CAS  PubMed  Google Scholar 

  16. Kalluri R. The biology and function of exosomes in cancer. J Clin Investig. 2016;126:1208–15.

    PubMed  PubMed Central  Google Scholar 

  17. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97:329–39.

    CAS  PubMed  Google Scholar 

  18. Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Koch M, Zhang J, Weitz J, Chin L, Futreal A. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014;289:3869–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Tarasov VV, Svistunov AA, Chubarev VN, Dostdar SA, Sokolov AV, Brzecka A, Sukocheva O, Neganova ME, Klochkov SG, Somasundaram SG. Extracellular vesicles in cancer nanomedicine. Semin Cancer Biol. 2021;69:212–25.

  20. Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin. 2016;6:287–96.

    Google Scholar 

  21. Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21:9–17.

    CAS  PubMed  Google Scholar 

  22. Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19:1–19.

    Google Scholar 

  23. Huang X, Yuan T, Tschannen M, Sun Z, Jacob H, Du M, Liang M, Dittmar RL, Liu Y, Liang M. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genom. 2013;14:1–14.

    Google Scholar 

  24. Huang Q, Yang J, Zheng J, Hsueh C, Guo Y, Zhou L. Characterization of selective exosomal microRNA expression profile derived from laryngeal squamous cell carcinoma detected by next generation sequencing. Oncol Rep. 2018;40:2584–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Manier S, Liu C-J, Avet-Loiseau H, Park J, Shi J, Campigotto F, Salem KZ, Huynh D, Glavey SV, Rivotto B. Prognostic role of circulating exosomal miRNAs in multiple myeloma. Blood J Am Soc Hem. 2017;129:2429–36.

    CAS  Google Scholar 

  26. Driedonks TA, Nolte-’t Hoen EN. Circulating Y-RNAs in extracellular vesicles and ribonucleoprotein complexes; implications for the immune system. Front Immunol. 2019;9:3164.

    PubMed  PubMed Central  Google Scholar 

  27. Goldie BJ, Dun MD, Lin M, Smith ND, Verrills NM, Dayas CV, Cairns MJ. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Res. 2014;42:9195–208.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Henke JI, Goergen D, Zheng J, Song Y, Schüttler CG, Fehr C, Jünemann C, Niepmann M. microRNA-122 stimulates translation of hepatitis C virus RNA. EMBO J. 2008;27:3300–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Turchinovich A, Weiz L, Burwinkel B. Extracellular miRNAs: the mystery of their origin and function. Trends Biochem Sci. 2012;37:460–5.

    CAS  PubMed  Google Scholar 

  30. Kosaka N, Iguchi H, Hagiwara K, Yoshioka Y, Takeshita F, Ochiya T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J Biol Chem. 2013;288:10849–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Frank F, Sonenberg N, Nagar B. Structural basis for 5′-nucleotide base-specific recognition of guide RNA by human AGO2. Nature. 2010;465:818–22.

    CAS  PubMed  Google Scholar 

  32. Koppers-Lalic D, Hackenberg M, Bijnsdorp IV, van Eijndhoven MA, Sadek P, Sie D, Zini N, Middeldorp JM, Ylstra B, de Menezes RX. Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes. Cell Rep. 2014;8:1649–58.

    CAS  PubMed  Google Scholar 

  33. Han S, Wang Z, Liu J, Wang H-MD, Yuan Q. miR-29a-3p-dependent COL3A1 and COL5A1 expression reduction assists sulforaphane to inhibit gastric cancer progression. Biochem Pharmacol. 2021;188: 114539.

    CAS  PubMed  Google Scholar 

  34. Théry C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Molecular characterization of dendritic cell-derived exosomes: selective accumulation of the heat shock protein hsc73. J Cell Biol. 1999;147:599–610.

    PubMed  PubMed Central  Google Scholar 

  35. Montecalvo A, Larregina AT, Shufesky WJ, Beer Stolz D, Sullivan ML, Karlsson JM, Baty CJ, Gibson GA, Erdos G, Wang Z. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood J Am Soc Hem. 2012;119:756–66.

    CAS  Google Scholar 

  36. Segura E, Nicco C, Lombard B, Véron P, Raposo G, Batteux F, Amigorena S, Théry C. ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood. 2005;106:216–23.

    CAS  PubMed  Google Scholar 

  37. O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21:585–606.

    PubMed  PubMed Central  Google Scholar 

  38. Alexander M, Hu R, Runtsch MC, Kagele DA, Mosbruger TL, Tolmachova T, Seabra MC, Round JL, Ward DM, O’Connell RM. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat Commun. 2015;6:7321.

    CAS  PubMed  Google Scholar 

  39. Lindenbergh MF, Koerhuis DG, Borg EG, van’t Veld EM, Driedonks TA, Wubbolts R, Stoorvogel W, Boes M. Bystander T-cells support clonal T-cell activation by controlling the release of dendritic cell-derived immune-stimulatory extracellular vesicles. Fron Immunol. 2019;10:448.

    CAS  Google Scholar 

  40. Salvi V, Gianello V, Busatto S, Bergese P, Andreoli L, D’Oro U, Zingoni A, Tincani A, Sozzani S, Bosisio D, Exosome-delivered microRNAs promote IFN-α secretion by human plasmacytoid DCs via TLR7. JCI Insight. 2018;3(10):e98204. https://doi.org/10.1172/jci.insight.98204.

  41. Li C, Guo F, Wang X, Liu D, Wu B, Wang F, Chen W. Exosome-based targeted RNA delivery for immune tolerance induction in skin transplantation. J Biomed Mat Res Part A. 2020;108:1493–500.

    CAS  Google Scholar 

  42. Asgarpour K, Shojaei Z, Amiri F, Ai J, Mahjoubin-Tehran M, Ghasemi F, ArefNezhad R, Hamblin MR, Mirzaei H. Exosomal microRNAs derived from mesenchymal stem cells: cell-to-cell messages. Cell Commun Signal. 2020;18:1–16.

    Google Scholar 

  43. Nelson MC, O’Connell RM. MicroRNAs: at the interface of metabolic pathways and inflammatory responses by macrophages. Front Immunol. 2020;11:1797.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Bouchareychas L, Duong P, Covarrubias S, Alsop E, Phu TA, Chung A, Gomes M, Wong D, Meechoovet B, Capili A. Macrophage exosomes resolve atherosclerosis by regulating hematopoiesis and inflammation via MicroRNA cargo. Cell Rep. 2020;32: 107881.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Li J, Xue H, Li T, Chu X, Xin D, Xiong Y, Qiu W, Gao X, Qian M, Xu J. Exosomes derived from mesenchymal stem cells attenuate the progression of atherosclerosis in ApoE−/-mice via miR-let7 mediated infiltration and polarization of M2 macrophage. Biochem Biophys Res Commun. 2019;510:565–72.

    CAS  PubMed  Google Scholar 

  47. Kang M, Huang C-C, Lu Y, Shirazi S, Gajendrareddy P, Ravindran S, Cooper LF. Bone regeneration is mediated by macrophage extracellular vesicles. Bone. 2020;141: 115627.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Cui H, He Y, Chen S, Zhang D, Yu Y, Fan C. Macrophage-derived miRNA-containing exosomes induce peritendinous fibrosis after tendon injury through the miR-21-5p/Smad7 pathway. Mol Ther Nucleic Acids. 2019;14:114–30.

    CAS  PubMed  Google Scholar 

  49. Ferrante CJ, Leibovich SJ. Regulation of macrophage polarization and wound healing. Adv Wound Care. 2012;1:10–6.

    Google Scholar 

  50. Chen W-X, Wang D-D, Zhu B, Zhu Y-Z, Zheng L, Feng Z-Q, Qin X-H. Exosomal miR-222 from adriamycin-resistant MCF-7 breast cancer cells promote macrophages M2 polarization via PTEN/Akt to induce tumor progression. Aging (Albany NY). 2021;13:10415.

    CAS  PubMed  Google Scholar 

  51. Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, Yeoh KW, Kon OL, Tam JP, Sze SK. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019;38:5158–73.

    CAS  PubMed  Google Scholar 

  52. Wu K-J, Wang W, Wang H-MD, Leung C-H, Ma D-L. Interfering with S100B–effector protein interactions for cancer therapy. Drug Discov Today. 2020;25:1754–61.

    CAS  PubMed  Google Scholar 

  53. Liu J, Fan L, Yu H, Zhang J, He Y, Feng D, Wang F, Li X, Liu Q, Li Y. Endoplasmic reticulum stress causes liver cancer cells to release exosomal miR-23a-3p and up-regulate programmed death ligand 1 expression in macrophages. Hepatology. 2019;70:241–58.

    CAS  PubMed  Google Scholar 

  54. Holder B, Jones T, Sancho Shimizu V, Rice TF, Donaldson B, Bouqueau M, Forbes K, Kampmann B. Macrophage exosomes induce placental inflammatory cytokines: a novel mode of maternal–placental messaging. Traffic. 2016;17:168–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Cano R, Lopera H. Chapter 5: Introduction to T and B lymphocytes. In Anaya JM, Shoenfeld Y, Rojas-Villarraga A, Cano RLE, Lopera HDE, editors. Autoimmunity: from bench to bedside. El Rosario University Press, Bogota (Colombia). 2013.

  56. Cox MA, Kahan SM, Zajac AJ. Anti-viral CD8 T cells and the cytokines that they love. Virology. 2013;435:157–69.

    CAS  PubMed  Google Scholar 

  57. Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol. 2014;14:24–35.

    CAS  PubMed  Google Scholar 

  58. Maybruck BT, Pfannenstiel LW, Diaz-Montero M, Gastman BR. Tumor-derived exosomes induce CD8+ T cell suppressors. J Immunother Cancer. 2017;5:1–15.

    Google Scholar 

  59. Shen T, Huang Z, Shi C, Pu X, Xu X, Wu Z, Ding G, Cao L. Pancreatic cancer-derived exosomes induce apoptosis of T lymphocytes through the p38 MAPK-mediated endoplasmic reticulum stress. FASEB J. 2020;34:8442–58.

    CAS  PubMed  Google Scholar 

  60. Chou H-L, Fong Y, Lin H-H, Tsai EM, Chen JY-F, Chang W-T, Wu C-Y, David Wang H-M, Huang H-W, Chiu C-C. An acetamide derivative as a camptothecin sensitizer for human non-small-cell lung cancer cells through increased oxidative stress and JNK activation. Oxid Med Cell Longev. 2016;2016:9128102. https://doi.org/10.1155/2016/9128102.

  61. Cui X, Wang S, Zhao N, Wang S, Wang Z, Huang M, Liu Y, Qin J, Shan Z, Teng W. Thyrocyte-derived exosome-targeted dendritic cells stimulate strong CD4+ T lymphocyte responses. Mol Cell Endocrinol. 2020;506: 110756.

    CAS  PubMed  Google Scholar 

  62. Mittelbrunn M, Gutiérrez-Vázquez C, Villarroya-Beltri C, González S, Sánchez-Cabo F, González MÁ, Bernad A, Sánchez-Madrid F. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun. 2011;2:1–10.

    Google Scholar 

  63. Fernández-Messina L, Rodríguez-Galán A, de Yébenes VG, Gutiérrez-Vázquez C, Tenreiro S, Seabra MC, Ramiro AR, Sánchez-Madrid F. Transfer of extracellular vesicle-micro RNA controls germinal center reaction and antibody production. EMBO Rep. 2020;21: e48925.

    PubMed  PubMed Central  Google Scholar 

  64. Guay C, Kruit JK, Rome S, Menoud V, Mulder NL, Jurdzinski A, Mancarella F, Sebastiani G, Donda A, Gonzalez BJ. Lymphocyte-derived exosomal microRNAs promote pancreatic β cell death and may contribute to type 1 diabetes development. Cell Metab. 2019;29(348–61): e6.

    Google Scholar 

  65. Okoye IS, Coomes SM, Pelly VS, Czieso S, Papayannopoulos V, Tolmachova T, Seabra MC, Wilson MS. MicroRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity. 2014;41:89–103.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Wang HMD, Fu L, Cheng CC, Gao R, Lin MY, Su HL, Belinda NE, Nguyen TH, Lin W-H, Lee PC. Inhibition of LPS-induced oxidative damages and potential anti-inflammatory effects of Phyllanthus emblica extract via down-regulating NF-κB, COX-2, and iNOS in RAW 264.7 cells. Antioxidants. 2019;8:270.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Tung SL, Boardman DA, Sen M, Letizia M, Peng Q, Cianci N, Dioni L, Carlin LM, Lechler R, Bollati V. Regulatory T cell-derived extracellular vesicles modify dendritic cell function. Sci Rep. 2018;8:1–12.

    Google Scholar 

  68. Deng J-N, Li Y-Q, Liu Y, Li Q, Hu Y, Xu J-Q, Sun T-Y, Xie L-X. Exosomes derived from plasma of septic patients inhibit apoptosis of T lymphocytes by down-regulating bad via hsa-miR-7-5p. Biochem Biophys Res Commun. 2019;513:958–66.

    CAS  PubMed  Google Scholar 

  69. Ye S-B, Li Z-L, Luo D-H, Huang B-J, Chen Y-S, Zhang X-S, Cui J, Zeng Y-X, Li J. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget. 2014;5:5439.

    PubMed  PubMed Central  Google Scholar 

  70. Dosil SG, Lopez-Cobo S, Rodriguez-Galan A, Fernandez-Delgado I, Ramirez-Huesca M, Milan-Rois P, Castellanos M, Somoza A, Gómez MJ, Reyburn HT. Natural killer (NK) cell-derived extracellular-vesicle shuttled microRNAs control T cell responses. Elife. 2022;11: e76319.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Saunderson SC, Schuberth PC, Dunn AC, Miller L, Hock BD, MacKay PA, Koch N, Jack RW, McLellan AD. Induction of exosome release in primary B cells stimulated via CD40 and the IL-4 receptor. J Immunol. 2008;180:8146–52.

    CAS  PubMed  Google Scholar 

  72. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, de Gruijl TD, Würdinger T, Middeldorp JM. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci. 2010;107:6328–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Liao T-L, Chen Y-M, Hsieh S-L, Tang K-T, Chen D-Y, Yang Y-Y, Liu H-J, Yang S-S. Hepatitis C virus-induced exosomal microRNAs and toll-like receptor 7 polymorphism regulate B-cell activating factor. MBio. 2021;12:e02764-e2821.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Li M, Zhao J, Cao M, Liu R, Chen G, Li S, Xie Y, Xie J, Cheng Y, Huang L. Mast cells-derived MiR-223 destroys intestinal barrier function by inhibition of CLDN8 expression in intestinal epithelial cells. Biol Res. 2020;53:1–11.

    CAS  Google Scholar 

  75. Hu L, Si L, Dai X, Dong H, Ma Z, Sun Z, Li N, Sha H, Chen Y, Qian Y. Exosomal miR-409-3p secreted from activated mast cells promotes microglial migration, activation and neuroinflammation by targeting Nr4a2 to activate the NF-κB pathway. J Neuroinflamm. 2021;18:1–13.

    Google Scholar 

  76. Xiong L, Zhen S, Yu Q, Gong Z. HCV-E2 inhibits hepatocellular carcinoma metastasis by stimulating mast cells to secrete exosomal shuttle microRNAs. Oncol Lett. 2017;14:2141–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Rabelo Melo F, Santosh Martin S, Sommerhoff CP, Pejler G. Exosome-mediated uptake of mast cell tryptase into the nucleus of melanoma cells: a novel axis for regulating tumor cell proliferation and gene expression. Cell Death Dis. 2019;10:1–16.

    CAS  Google Scholar 

  78. Cheng Y-C, Chang Y-A, Chen Y-J, Sung H-M, Bogeski I, Su H-L, Hsu Y-L, Hsu H-MD. The roles of extracellular vesicles in malignant melanoma. Cells. 2021;10:2740.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Chou H-Y, Liu L-H, Chen C-Y, Lin I-F, Ali D, Lee AY-L, Wang H-MD. Bifunctional mechanisms of autophagy and apoptosis regulations in melanoma from Bacillus subtilis natto fermentation extract. Food Chem Toxicol. 2021;150: 112020.

    CAS  PubMed  Google Scholar 

  80. Mazzeo C, Cañas JA, Zafra MP, Marco AR, Fernández-Nieto M, Sanz V, Mittelbrunn M, Izquierdo M, Baixaulli F, Sastre J. Exosome secretion by eosinophils: a possible role in asthma pathogenesis. J Allergy Clin Immunol. 2015;135:1603–13.

    CAS  PubMed  Google Scholar 

  81. Cañas J, Sastre B, Rodrigo-Muñoz J, Fernández-Nieto M, Barranco P, Quirce S, Sastre J, Del Pozo V. Eosinophil-derived exosomes contribute to asthma remodelling by activating structural lung cells. Clin Exp Allergy. 2018;48:1173–85.

    PubMed  Google Scholar 

  82. Rodrigo-Muñoz JM, Cañas JA, Sastre B, Rego N, Greif G, Rial M, Mínguez P, Mahíllo-Fernández I, Fernández-Nieto M, Mora I. Asthma diagnosis using integrated analysis of eosinophil microRNAs. Allergy. 2019;74:507–17.

    PubMed  Google Scholar 

  83. Gao H, Yu Z, Li Y, Wang X. miR-100-5p in human umbilical cord mesenchymal stem cell-derived exosomes mediates eosinophilic inflammation to alleviate atherosclerosis via the FZD5/Wnt/β-catenin pathway. Acta Biochim Biophys Sin. 2021;53:1166–76.

    CAS  PubMed  Google Scholar 

  84. Shao S, Fang H, Zhang J, Jiang M, Xue K, Ma J, Zhang J, Lei J, Zhang Y, Li B. Neutrophil exosomes enhance the skin autoinflammation in generalized pustular psoriasis via activating keratinocytes. FASEB J. 2019;33:6813–28.

    CAS  PubMed  Google Scholar 

  85. Linhares-Lacerda L, Temerozo JR, Ribeiro-Alves M, Azevedo EP, Mojoli A, Nascimento MT, Silva-Oliveira G, Savino W, Foguel D, Bou-Habib DC. Neutrophil extracellular trap-enriched supernatants carry microRNAs able to modulate TNF-α production by macrophages. Sci Rep. 2020;10:1–13.

    Google Scholar 

  86. Tyagi A, Wu S-Y, Sharma S, Wu K, Zhao D, Deshpande R, Singh R, Li W, Topaloglu U, Ruiz J. Exosomal miR-4466 from nicotine-activated neutrophils promotes tumor cell stemness and metabolism in lung cancer metastasis. Oncogene. 2022;41:3079–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Li L, Zuo X, Liu D, Luo H, Zhu H. The profiles of miRNAs and lncRNAs in peripheral blood neutrophils exosomes of diffuse cutaneous systemic sclerosis. J Dermatol Sci. 2020;98:88–97.

    CAS  PubMed  Google Scholar 

  88. Li Q, Nong A, Huang Z, He K, Jia Y, Huang Y. Exosomes containing miR-122-5p secreted by LPS-induced neutrophils regulate the apoptosis and permeability of brain microvascular endothelial cells by targeting OCLN. Am J Transl Res. 2021;13:4167.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Jiao Y, Zhang T, Zhang C, Ji H, Tong X, Xia R, Wang W, Ma Z, Shi X. Exosomal miR-30d-5p of neutrophils induces M1 macrophage polarization and primes macrophage pyroptosis in sepsis-related acute lung injury. Crit Care. 2021;25:1–15.

    Google Scholar 

  90. Jeffries J, Zhou W, Hsu AY, Deng Q. miRNA-223 at the crossroads of inflammation and cancer. Cancer Lett. 2019;451:136–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Truffi M, Sorrentino L, Corsi F. Fibroblasts in the tumor microenvironment. Tumor Microenviron. 2020;1234:15–29. https://doi.org/10.1007/978-3-030-37184-5_2.

  92. Lan Y-H, Lu Y-S, Wu J-Y, Lee H-T, Srinophakun P, Canko GN, Chiu C-C, Wang H-MD. Cordyceps militaris reduces oxidative stress and regulates immune T cells to inhibit metastatic melanoma invasion. Antioxidants. 2022;11:1502.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. 2020;30:R921–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Hsu S-K, Cheng K-C, Mgbeahuruike MO, Lin Y-H, Wu C-Y, Wang H-MD, Yen C-H, Chiu C-C, Sheu S-J. New insight into the effects of metformin on diabetic retinopathy, aging and cancer: nonapoptotic cell death, immunosuppression, and effects beyond the AMPK pathway. Int J Mol Sci. 2021;22:9453.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Zeltz C, Primac I, Erusappan P, Alam J, Noel A, Gullberg D. Cancer-associated fibroblasts in desmoplastic tumors: emerging role of integrins. Semin Cancer Biol. 2020;62:166–81.

  96. Lei X, Lei Y, Li J-K, Du W-X, Li R-G, Yang J, Li J, Li F, Tan H-B. Immune cells within the tumor microenvironment: biological functions and roles in cancer immunotherapy. Cancer Lett. 2020;470:126–33.

    CAS  PubMed  Google Scholar 

  97. Fattore L, Ruggiero CF, Pisanu ME, Liguoro D, Cerri A, Costantini S, Capone F, Acunzo M, Romano G, Nigita G. Reprogramming miRNAs global expression orchestrates development of drug resistance in BRAF mutated melanoma. Cell Death Differ. 2019;26:1267–82.

    CAS  PubMed  Google Scholar 

  98. Wu KJ, Ho SH, Wu C, Wang HMD, Ma DL, Leung CH. Simultaneous blocking of the pan-RAF and S100B pathways as a synergistic therapeutic strategy against malignant melanoma. J Cell Mol Med. 2021;25:1972–81.

    CAS  PubMed  Google Scholar 

  99. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.

    CAS  PubMed  Google Scholar 

  100. Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, McNamara KL, Granja JM, Sarin KY, Brown RA. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25:1251–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, Takeda K, Van Dommelen SL, Degli-Esposti MA, Hayakawa Y. Activation of NK cell cytotoxicity. Mol Immunol. 2005;42:501–10.

    CAS  PubMed  Google Scholar 

  102. Batista FD, Harwood NE. The who, how and where of antigen presentation to B cells. Nat Rev Immunol. 2009;9:15–27.

    CAS  PubMed  Google Scholar 

  103. Narayanan S, Kawaguchi T, Peng X, Qi Q, Liu S, Yan L, Takabe K. Tumor infiltrating lymphocytes and macrophages improve survival in microsatellite unstable colorectal cancer. Sci Rep. 2019;9:1–10.

    CAS  Google Scholar 

  104. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431–46.

    CAS  PubMed  Google Scholar 

  105. Zhou J, Nefedova Y, Lei A, Gabrilovich D. Neutrophils and PMN-MDSC: Their biological role and interaction with stromal cells. Semin Immunol. 2018;35:19–28.

  106. Tsou P, Katayama H, Ostrin EJ, Hanash SM. The emerging role of B cells in tumor immunityB cells, cancer immunity, and autoantibodies. Cancer Res. 2016;76:5597–601.

    CAS  PubMed  Google Scholar 

  107. Li I, Nabet BY. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol Cancer. 2019;18:1–10.

    PubMed  PubMed Central  Google Scholar 

  108. Patel H, Nilendu P, Jahagirdar D, Pal JK, Sharma NK. Modulating secreted components of tumor microenvironment: a masterstroke in tumor therapeutics. Cancer Biol Ther. 2018;19:3–12.

    CAS  PubMed  Google Scholar 

  109. Zhan C, Yang X, Yin X, Hou J. Exosomes and other extracellular vesicles in oral and salivary gland cancers. Oral Dis. 2020;26:865–75.

    PubMed  Google Scholar 

  110. Tan S, Xia L, Yi P, Han Y, Tang L, Pan Q, Tian Y, Rao S, Oyang L, Liang J. Exosomal miRNAs in tumor microenvironment. J Exp Clin Cancer Res. 2020;39:1–15.

    Google Scholar 

  111. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A, O’Connor STF, Chin AR. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25:501–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Bao L, You B, Shi S, Shan Y, Zhang Q, Yue H, Zhang J, Zhang W, Shi Y, Liu Y. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene. 2018;37:2873–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Sun X, Ma X, Wang J, Zhao Y, Wang Y, Bihl JC, Chen Y, Jiang C. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget. 2017;8:36137.

    PubMed  PubMed Central  Google Scholar 

  114. Chen X, Yang F, Zhang T, Wang W, Xi W, Li Y, Zhang D, Huo Y, Zhang J, Yang A. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. J Exp Clin Cancer Res. 2019;38:1–16.

    Google Scholar 

  115. Kim DH, Park S, Kim H, Choi YJ, Kim SY, Sung KJ, Sung YH, Choi C-M, Yun M, Yi Y-S. Tumor-derived exosomal miR-619–5p promotes tumor angiogenesis and metastasis through the inhibition of RCAN1. 4. Cancer Lett. 2020;475:2–13.

    CAS  PubMed  Google Scholar 

  116. Lawson J, Dickman C, Towle R, Jabalee J, Javer A, Garnis C. Extracellular vesicle secretion of miR-142-3p from lung adenocarcinoma cells induces tumor promoting changes in the stroma through cell-cell communication. Mol Carcinog. 2019;58:376–87.

    CAS  PubMed  Google Scholar 

  117. Lin X-J, Fang J-H, Yang X-J, Zhang C, Yuan Y, Zheng L, Zhuang S-M. Hepatocellular carcinoma cell-secreted exosomal microRNA-210 promotes angiogenesis in vitro and in vivo. Mol Ther Nucleic Acids. 2018;11:243–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Pakravan K, Babashah S, Sadeghizadeh M, Mowla SJ, Mossahebi-Mohammadi M, Ataei F, Dana N, Javan M. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol. 2017;40:457–70.

    CAS  Google Scholar 

  119. Deng Z, Rong Y, Teng Y, Zhuang X, Samykutty A, Mu J, Zhang L, Cao P, Yan J, Miller D. Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene. 2017;36:639–51.

    CAS  PubMed  Google Scholar 

  120. He L, Zhu W, Chen Q, Yuan Y, Wang Y, Wang J, Wu X. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics. 2019;9:8206.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Masoumi-Dehghi S, Babashah S, Sadeghizadeh M. microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK/STAT3 and NF-κB signaling pathways. J Cell Commun Signal. 2020;14:233–44.

    PubMed  PubMed Central  Google Scholar 

  122. Yang W, Tan S, Yang L, Chen X, Yang R, Oyang L, Lin J, Xia L, Wu N, Han Y. Exosomal miR-205-5p enhances angiogenesis and nasopharyngeal carcinoma metastasis by targeting desmocollin-2. Mol Ther Oncolytics. 2022;24:612–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Liu Y, Yang Q, Fu H, Wang J, Yuan S, Li X, Xie P, Hu Z, Liu Q. Müller glia-derived exosomal miR-9-3p promotes angiogenesis by restricting sphingosine-1-phosphate receptor S1P1 in diabetic retinopathy. Mol Ther Nucleic Acids. 2022;27:491–504.

    CAS  PubMed  Google Scholar 

  124. Zeng Z, Li Y, Pan Y, Lan X, Song F, Sun J, Zhou K, Liu X, Ren X, Wang F. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018;9:5395.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Hsu Y, Hung J, Chang W, Lin Y, Pan Y, Tsai P, Wu C, Kuo P. Hypoxic lung cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene. 2017;36:4929–42.

    CAS  PubMed  Google Scholar 

  126. Lopatina T, Grange C, Fonsato V, Tapparo M, Brossa A, Fallo S, Pitino A, Herrera-Sanchez MB, Kholia S, Camussi G. Extracellular vesicles from human liver stem cells inhibit tumor angiogenesis. Int J Cancer. 2019;144:322–33.

    CAS  PubMed  Google Scholar 

  127. Zhao S, Li J, Zhang G, Wang Q, Wu C, Zhang Q, Wang H, Sun P, Xiang R, Yang S. Exosomal miR-451a functions as a tumor suppressor in hepatocellular carcinoma by targeting LPIN1. Cell Physiol Biochem. 2019;53:19–35.

    CAS  PubMed  Google Scholar 

  128. Lu J, Liu Q-H, Wang F, Tan J-J, Deng Y-Q, Peng X-H, Liu X, Zhang B, Xu X, Li X-P. Exosomal miR-9 inhibits angiogenesis by targeting MDK and regulating PDK/AKT pathway in nasopharyngeal carcinoma. J Exp Clin Cancer Res. 2018;37:1–12.

    Google Scholar 

  129. Vignard V, Labbé M, Marec N, André-Grégoire G, Jouand N, Fonteneau J-F, Labarrière N, Fradin D. MicroRNAs in tumor exosomes drive immune escape in melanomaMicroRNA in tumor immune escape. Cancer Immunol Res. 2020;8:255–67.

    CAS  PubMed  Google Scholar 

  130. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers. 2014;6:1670–90.

    PubMed  PubMed Central  Google Scholar 

  131. Allavena P, Sica A, Solinas G, Porta C, Mantovani A. The inflammatory micro-environment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol. 2008;66:1–9.

    PubMed  Google Scholar 

  132. Hao N-B, Lü M-H, Fan Y-H, Cao Y-L, Zhang Z-R, Yang S-M, Macrophages in tumor microenvironments and the progression of tumors. Clin Dev Immunol. 2012;2012:948098. https://doi.org/10.1155/2012/948098.

  133. Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, Liu B, Su L, Qiu Z. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer MetastasisTumor-promoting effects of hypoxic exosomal miR-301a. Cancer Res. 2018;78:4586–98.

    CAS  PubMed  Google Scholar 

  134. Ying X, Wu Q, Wu X, Zhu Q, Wang X, Jiang L, Chen X, Wang X. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget. 2016;7:43076.

    PubMed  PubMed Central  Google Scholar 

  135. Chen X, Zhou J, Li X, Wang X, Lin Y, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018;435:80–91.

    CAS  PubMed  Google Scholar 

  136. Cooks T, Pateras IS, Jenkins LM, Patel KM, Robles AI, Morris J, Forshew T, Appella E, Gorgoulis VG, Harris CC. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun. 2018;9:771.

    PubMed  PubMed Central  Google Scholar 

  137. Hsieh C-H, Tai S-K, Yang M-H. Snail-overexpressing cancer cells promote M2-like polarization of tumor-associated macrophages by delivering MiR-21-abundant exosomes. Neoplasia. 2018;20:775–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Kikete S, Chu X, Wang L, Bian Y. Endogenous and tumour-derived microRNAs regulate cross-presentation in dendritic cells and consequently cytotoxic T cell function. Cytotechnology. 2016;68:2223–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Ding G, Zhou L, Shen T, Cao L. IFN-γ induces the upregulation of RFXAP via inhibition of miR-212-3p in pancreatic cancer cells: a novel mechanism for IFN-γ response. Oncol Lett. 2018;15:3760–5.

    PubMed  PubMed Central  Google Scholar 

  140. Zhou M, Chen J, Zhou L, Chen W, Ding G, Cao L. Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell Immunol. 2014;292:65–9.

    CAS  PubMed  Google Scholar 

  141. Tung SL, Boardman DA, Sen M, Letizia M, Peng Q, Cianci N, Dioni L, Carlin LM, Lechler R, Bollati V. Regulatory T cell-derived extracellular vesicles modify dendritic cell function. Sci Rep. 2018;8:6065.

    PubMed  PubMed Central  Google Scholar 

  142. Taghikhani A, Hassan ZM, Ebrahimi M, Moazzeni SM. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J Cell Physiol. 2019;234:9417–27.

    CAS  PubMed  Google Scholar 

  143. Guo X, Qiu W, Liu Q, Qian M, Wang S, Zhang Z, Gao X, Chen Z, Xue H, Li G. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten pathways. Oncogene. 2018;37:4239–59.

    CAS  PubMed  Google Scholar 

  144. Bruns H, Böttcher M, Qorraj M, Fabri M, Jitschin S, Dindorf J, Busch L, Jitschin R, Mackensen A, Mougiakakos D. CLL-cell-mediated MDSC induction by exosomal miR-155 transfer is disrupted by vitamin D. Leukemia. 2017;31:985–8.

    CAS  PubMed  Google Scholar 

  145. Sun Y, Wang R, Qiao M, Xu Y, Guan W, Wang L. Cancer associated fibroblasts tailored tumor microenvironment of therapy resistance in gastrointestinal cancers. J Cell Physiol. 2018;233:6359–69.

    CAS  PubMed  Google Scholar 

  146. Alkasalias T, Moyano-Galceran L, Arsenian-Henriksson M, Lehti K. Fibroblasts in the tumor microenvironment: shield or spear? Int J Mol Sci. 2018;19:1532.

    PubMed  PubMed Central  Google Scholar 

  147. Zhang Y, Cai H, Chen S, Sun D, Zhang D, He Y. Exosomal transfer of miR-124 inhibits normal fibroblasts to cancer-associated fibroblasts transition by targeting sphingosine kinase 1 in ovarian cancer. J Cell Biochem. 2019;120:13187–201.

    CAS  PubMed  Google Scholar 

  148. Frassanito MA, Desantis V, Di Marzo L, Craparotta I, Beltrame L, Marchini S, Annese T, Visino F, Arciuli M, Saltarella I. Bone marrow fibroblasts overexpress miR-27b and miR-214 in step with multiple myeloma progression, dependent on tumour cell-derived exosomes. J Pathol. 2019;247:241–53.

    CAS  PubMed  Google Scholar 

  149. Wang J, Guan X, Zhang Y, Ge S, Zhang L, Li H, Wang X, Liu R, Ning T, Deng T. Exosomal miR-27a derived from gastric cancer cells regulates the transformation of fibroblasts into cancer-associated fibroblasts. Cell Physiol Biochem. 2018;49:869–83.

    CAS  PubMed  Google Scholar 

  150. Dai G, Yao X, Zhang Y, Gu J, Geng Y, Xue F, Zhang J. Colorectal cancer cell–derived exosomes containing miR-10b regulate fibroblast cells via the PI3K/Akt pathway. Bull Cancer. 2018;105:336–49.

    PubMed  Google Scholar 

  151. Zhou X, Yan T, Huang C, Xu Z, Wang L, Jiang E, Wang H, Chen Y, Liu K, Shao Z. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J Exp Clin Cancer Res. 2018;37:1–15.

    Google Scholar 

  152. Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J, Shi X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J Exp Clin Cancer Res. 2018;37:1–18.

    Google Scholar 

  153. Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J, Shi X. Correction: Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J Exp Clin Cancer Res. 2022;41:359.

    PubMed  PubMed Central  Google Scholar 

  154. Baroni S, Romero-Cordoba S, Plantamura I, Dugo M, D’ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone M. Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 2016;7:e2312–412.

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Yan W, Wu X, Zhou W, Fong MY, Cao M, Liu J, Liu X, Chen C-H, Fadare O, Pizzo DP. Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat Cell Biol. 2018;20:597–609.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Johnstone RM, Adam M, Hammond J, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262:9412–20.

    CAS  PubMed  Google Scholar 

  157. Harding C, Heuser J, Stahl P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur J Cell Biol. 1984;35:256–63.

    CAS  PubMed  Google Scholar 

  158. Zhong S, Li W, Chen Z, Xu J, Zhao J. MiR-222 and miR-29a contribute to the drug-resistance of breast cancer cells. Gene. 2013;531:8–14.

    CAS  PubMed  Google Scholar 

  159. Trivedi M, Talekar M, Shah P, Ouyang Q, Amiji M. Modification of tumor cell exosome content by transfection with wt-p53 and microRNA-125b expressing plasmid DNA and its effect on macrophage polarization. Oncogenesis. 2016;5:e250–350.

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, Liu B, Su L, Qiu Z. Correction: hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis. Cancer Res. 2020;80:922–1022.

    PubMed  Google Scholar 

  161. Yoshii S, Hayashi Y, Iijima H, Inoue T, Kimura K, Sakatani A, Nagai K, Fujinaga T, Hiyama S, Kodama T. Exosomal micro RNA s derived from colon cancer cells promote tumor progression by suppressing fibroblast TP 53 expression. Cancer Sci. 2019;110:2396–407.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Fang T, Lv H, Lv G, Li T, Wang C, Han Q, Yu L, Su B, Guo L, Huang S. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9:191.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

All figures were created with BioRender.com.

Funding

This research was supported by the Ministry of Science and Technology, Taiwan for the grants (MOST 110-2221-E-005-010, MOST 111-2221-E-005-009, and MOST 111-2221-E-005-026-MY3).

Author information

Authors and Affiliations

Authors

Contributions

HMN: Methodology, Conceptualization, Writing—Original Draft, Revised draft, Visualization, and created graphic illustrations. C-CC: Methodology, Conceptualization, Visualization. C-HL: Methodology, Revised draft, Visualization. MMMA: Revised draft, Conceptualization, Revision, Visualization, and created graphic illustrations. H-MDW: Supervision; Project administration, Funding acquisition, and Final Revisions. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Hui-Min David Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nail, H.M., Chiu, CC., Leung, CH. et al. Exosomal miRNA-mediated intercellular communications and immunomodulatory effects in tumor microenvironments. J Biomed Sci 30, 69 (2023). https://doi.org/10.1186/s12929-023-00964-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-023-00964-w

Keywords