Skip to main content
iScience logoLink to iScience
. 2023 Mar 4;26(4):106333. doi: 10.1016/j.isci.2023.106333

Role of exosomal ncRNAs released by M2 macrophages in tumor progression of gastrointestinal cancers

Abdo Meyiah 1,4, Murad Alahdal 1,4, Eyad Elkord 1,2,3,
PMCID: PMC10031147  PMID: 36968082

Summary

Macrophages (MΦs) type 2 (M2) play crucial roles in the pathogenesis of gastrointestinal cancers (GIC) by enhancing tumor progression, invasion, and metastasis. Polarized M2 has been linked to the increase of GIC tumorigenesis and drug resistance. Several studies reported that M2-derived exosomal non-coding RNAs (Exos-ncRNAs) play pivotal roles in the modulation of the GIC tumor microenvironment (TME) and mostly promote drug resistance and immunosuppression. The impact of M2-Exos-ncRNAs is attributed to altered signaling pathways, enhancement of immunoregulatory mechanisms, and post-transcriptional modulation. Recent studies described novel targets in M2-TAMs-derived Exos-ncRNAs and potential promising clinical outcomes such as inhibiting tumor formation, invasion, and metastasis. Highlighting current knowledge of M2-Exos-ncRNAs involved in GIC pathogenesis and immunomodulation would thus be a significant contribution to improving clinical outcomes. In this review, we summarize recent updates on the role of M2-TAMs-Exos-ncRNAs in GIC pathogenesis, immunosuppression, and drug resistance. A deep understanding of M2-TAMs-derived Exos-ncRNAs could help to identify potential biomarkers and therapeutic targets.

Subject areas: Immunology, Cancer

Graphical abstract

graphic file with name fx1.jpg


Immunology; Cancer

Introduction

Gastrointestinal cancers (GIC) are a group of malignant cancers counting for 26% of the global incidence of cancers and 35% of life-threatening cancers.1,2 They consist of five major cancers: esophageal cancer (EC), gastric cancer (GC), liver cancer (LC) or hepatocellular carcinoma (HCC), pancreatic cancer (PaC) or pancreatic ductal adenocarcinoma (PDAC), and colorectal cancer (CRC). Usually, late diagnosis, poor prognosis, and low overall survival are mostly dominant in GIC patients. Despite advances in surgical procedures that improved outcomes, GIC malignancies continue to have high recurrence rates.3,4,5 Thus, several studies focused on the tumor microenvironment (TME) in terms of tumor cell biology, immune cell infiltration, and the interaction between the tumor and the immune system. Macrophages (MΦs), as one of the critical myeloid immune populations in the TME,6 were investigated enormously in terms of subtypes, polarization, interaction with tumor cells, and immunomodulatory effects.7 The population of MΦs infiltrating tumors (TAMs) usually present two polarized subtypes; classical M1 and alternative M2.8 Adding to that non-activated phenotype is named M0.9 TAMs are well known as tumor enhancers by modulating immune responses in the TME; however, two clinical studies on multivariate analyses of CRC patients (46 cases in Sweden and 160 cases in China) revealed that increased CD68TFHotspot MΦs were associated with good survival and inhibition of tumor growth.10,11 Importantly, both studies did not clearly distinguish the phenotype of these reported TAMs, which could be M1-TAMs as other literature linked the anti-tumor effects to M1-TAMs. It was reported that M1-TAMs show inflammatory boosting activity through activating T cell responses and tumor cell killing by releasing reactive oxygen/nitrogen species (ROS/RNS) besides the pro-inflammatory cytokines including tumor necrosis factor alpha (TNF-α), IL-6, and IL-1β.12 In contrast, the M2-TAM phenotype was considered mostly as “bad guys” in the TME. It exerts pro-tumorigenic effects through different pathways, such as the suppression of anti-tumor immune responses, which promote tumor growth, inducing tumor proliferation, angiogenesis, metastasis, invasion, and mediating chemotherapy resistance.13,14 Several studies demonstrated the crucial role of M2-TAMs in the pathogenesis of GIC.15 The link between the increasing number of M2-TAMs in GIC and poor prognosis has been extensively studied.16,17,18 Furthermore, M2-polarized TAMs not only produce tumor-supporting chemokines and cytokines but also release highly effective exosomes (Exos), which in turn contribute to the promotion of tumor invasion and metastasis, in addition to the modulation of epithelial-mesenchymal transition (EMT) into cancer stem cells.19,20

As known, Exos are a subtype of extracellular vesicles (EVs) with a diameter of ∼40–150 nm in size.21 It is secreted by various cell types (stromal cells, stem cells, immune cells, tumor cells, and epithelial cells) during physiological and pathophysiological conditions to facilitate cell functions.22 Various techniques have been established to investigate the biochemical and physicochemical properties of Exos despite the lack of standardized methods.23 It is possible to characterize Exos by either their physical properties, including size, shape, surface charge, and density, which can affect their cell adhesion and also uptake into neighboring cells,24 or by their biological properties, including their cargo (such as RNA species and proteins) and membrane antigens. Techniques that can be used for measuring density, size, shape, and surface charge in a suspension of vesicles (Exos), including dynamic light scattering (DLS), electron microscopy (EM), nanoparticle tracking analysis (NTA), and tunable resistive pulse sensing (TRPS).25 In general, ncRNAs in Exos are more stable than free RNAs in the extracellular fluid, due to the protective surrounding lipid bilayer.26 Exos-ncRNAs released by MΦs have various physical and chemical properties, depending on the type of RNAs (such as miRNA, lncRNA, and circRNA). Physically, they have spherical to cup-shaped with a small size, while their chemical properties depend on the source cells and the species of ncRNA. Furthermore, their stability depends on the specific ncRNA species and also the condition they are exposed to. These are just some of the general physical and chemical properties of Exos-ncRNAs released by MΦs.

Exos in the TME may promote cellular phenotypic and genetic changes, leading to enhance drug resistance, heterogeneity, and plasticity by releasing specific ncRNAs targeting molecular pathways, which regulate apoptosis and immune suppression.27 Tumor drug resistance could be attributed to the significant development of tumor heterogeneity due to the transferred Exos-ncRNAs.28 Furthermore, another study linked high levels of Exos-miR32-5p in the TME of HCC to multidrug resistance due to induction of tumor cell heterogeneity.29 A recent study compared the results of high-throughput sequencing of CRC cell line (SW620) with normal colon epithelial cells (NCM460) revealed that Exos enriched with miR-2277-3p and miR-26b-3p were responsible for heterogeneous paracrine effects in CRC.30 Thus, Exos-ncRNAs play a significant role in promoting a switch in tumor heterogeneity and plasticity.31 They induce drug resistance by creating specific signals inducing tumor heterogeneity, which enhance tumor growth and metastasis.32 Additionally, Exos-ncRNAs can promote epigenetic oncogenesis by regulating DNA methylation and histone modifications.33,34

Recently, the focus on targeting non-coding RNAs (ncRNAs) cargo in the Exos released by M2-TAMs is increasing due to promising outcomes. The silencing of miR-21-5p in M2-Exos significantly abolished the pro-metastatic effects of these M2-Exos in renal cell carcinoma (RCC).35 A recent study has shown that inhibiting lncRNA-CRNDE in M2-Exos enhances sensitivity to cisplatin (CDDP) in GC.36 Another study showed that the knock-down of miR-223 M2-Exos reduced resistance to doxorubicin in GC.37 Moreover, inhibition of miR-501-3p in M2-Exos inhibited tumor formation and metastasis in PDAC. This review intends to emphasize the importance of M2-TAMs-derived Exos-ncRNAs in the pathogenesis of GIC and highlights the potential opportunities in targeting M2-TAMs-Exos-ncRNAs in immunotherapies of GIC.

Interaction between GIC and M2-TAMs-Exos-ncRNAs in the TME

Previous studies suggested that TAMs accumulate in the tissue of GICs due to cancer-released mediators such as CCL-2 and CSF1, which recruit MΦs to infiltrate tumor tissues.17,38,39 Several pieces of evidence highlighted the role of Exos released by cancer cells in the intracellular communication between TAMs and tumor cells, thus promoting M2 polarization,40,41 as shown in Figure 1. Tumor cell-derived Exos transfer many molecules into TAMs such as oncoproteins, miRNAs, lncRNAs, circRNAs, and lipids that actively mediate polarization into M2 and thus enhancing tumor progression.42 A recent report highlighted the important role of Exos-ncRNAs in the polarization and function of MΦs.43 As presented in Figure 1, Exos released by M2-TAMs were linked to tumor cell proliferation, invasion, and metastasis.44 As reported, Exos-miR-934 regulates the activation of PI3K/AKT signaling, which induces M2-TAM polarization in CRC by inhibiting the expression of phosphatase and tensin homologs (PTEN).45 Polarized M2-TAMs induced CRC liver metastasis and promoted pre-metastatic niche formation by secreting CXCL13, which activates a CXCL13/CXCR5/NF-κB/p65/miR-934 positive feedback loop in CRC cells.45 Furthermore, Exos-miR-452-5p released by HCC induced MΦs polarization into M2 phenotype by targeting TIMP3, which promoted HCC progression. This was confirmed by the inhibition of Exos-miR-452-5p, which reduced M2 polarization and tumor progression.46 A recent study reported on the role of Exos-miR-19b-3p released by lung adenocarcinoma in the polarization of MΦs into M2 by targeting PTPRD, which induced dephosphorylation of STAT3.47 Polarized M2 by miR-19b-3p released Exos-linc00273, which promoted tumor metastasis. The same Exos-miR-19b was found to be elevated in the plasma of PDAC patients,48 but its relationship with M2 polarization in PDAC remains undisclosed. Exos-circ0048117 in esophageal squamous cells (ESC) plays a significant role in the induction of TAM polarization into M2 by sponge miR-140, resulting in increased TLR4 expression.49 M2-TAMs promote tumor cell invasion and migration by secreting TGF-β, Arg1, and IL-10.49 Moreover, Exos-miR-301a-3p derived from PDAC showed a significant promotion of M2 polarization by inducing PTEN/PI3Kγ activation, and these M2-TAMs enhanced tumor growth and metastasis.50 On the other hand, Exos-ncRNAs released by M2-TAMs showed a strong association with tumor progression, invasion, and metastasis. M2-Exos-miR-21-5p promoted tumor metastasis in RCC.35 In the avian embryo chorioallantoic membrane in vivo tumor model as well as in vitro, tumor metastasis was prevented by miR-21-5p knockdown in M2-TMAs-Exos.35 Furthermore, M2-Exos-miR-487a significantly promoted tumor proliferation and progression in vitro and in vivo in GC.51 Adding to that, Exos-miR-365 secreted by M2-TMAs was reported as a significant promoter of PDAC progression by downregulating the expression of BTG2, leading to the promotion of FAK/AKT signaling.52 In addition, high levels of Exos-miR-501-3p released by M2-TMAs in the TME of PDAC promoted tumor formation and metastasis by targeting the TGFBR3 gene, which increased the expression of TGF-β.53 Interestingly, inhibition of M2-Exos-miR-501-3p significantly reduced PDAC formation and metastasis. Another study reported that Exos released by M2-TAMs with the increase of EC invasion and metastasis by transferring lncRNA (AFAP1-AS1) that target miR-26a, leading to the activation of transcription factor 2 (ATF2) axis. Downregulation of miR-26a and upregulation of ATF2 promoted the increase of EC invasion and metastasis.54 X-chromosome-linked inhibitor of apoptosis protein (XIAP) has been detected in different types of tumors, which can play an important role in the inhibition of active apoptosis execution phase (caspase 3 and 7), and also apoptosis initiation phase (caspase 9) via subtypes of baculovirus IAP repeat (BIR2 and BIR3) domains of XIAP, resulting in blocking cell apoptosis, and promoting tumor progression.55,56 Additionally, miR-122-5p has been recognized as a key biological regulator, which can play a key role in modulating cell apoptosis and tumor progression.57 Therefore, an increased level of miR-122-5p could be related to enhanced cell apoptosis and suppression of tumor growth.58 A recent study reported that low expression of lncRNA SBF2-AS1 in M2-TAMs-Exos leads to up-regulation of miR-122-5p and accelerates the growth of PDAC through enhancing the XIAP gene.56,59 Thus, high levels of XIAP and low expressions of miR-122-5p play crucial roles in preventing apoptosis of PDAC cells.56,59 Altogether, the interaction between GIC and M2-TAMs is mostly mediated via Exos-ncRNAs besides other mediators such as cytokines. The focus on the Exos-ncRNAs communication is probably the most appropriate way to provide an efficient contribution to the clinical treatment of GIC.

Figure 1.

Figure 1

Role of tumor cell-derived Exos-ncRNAs and M2-TAMs-Exos-ncRNAs in the polarization of TAMs in the TME of GICs and modulation of TAM function

This schematic model shows the link between Exos-ncRNAs released by tumor cells and the polarization of TAMs into M2. The figure presents the role of M2-Exos-ncRNAs in the GIC tumor progression.

Role of M2-Exos-ncRNAs in the pathogenesis of different GIC types

M2-TAM accumulation in the TME of GICs is typically associated with poor prognoses and resistance to cancer therapy. Many clinical and experimental data revealed that M2-TAMs promote tumor heterogeneity, progression, and metastasis via Exos-ncRNAs,60 as presented in Figures 2 and 3. In the past few years, there has been an increased emphasis on the role and function of Exos-ncRNAs released by M2-TAMs, which can contribute to tumor progression. M2-TAMs-Exos-ncRNAs play crucial roles in modulating genomic imprinting, cellular differentiation, cell cycle, tumor progression, and a variety of biological activities. M2-TAMs-Exos-ncRNAs control transcription, translation, epigenetic modifications, and function of proteins.61 Different recent studies reported the functional roles of Exos-ncRNAs released by M2-TAMs in the development and progression of GIC, as summarized in Table 1. Although limited, we provide some evidence to declare the role of M2-TAMs-Exos-ncRNAs in different GICs, as described in the following.

Figure 2.

Figure 2

Role of Exos-ncRNAs in the TME of GICs. Exosomal ncRNAs from M2-TAMs are released into the TME and distant organs, delivering ncRNA, which can promote tumor progression

This schematic model presents the potential impact of Exos-ncRNAs released by M2-TAMs on tumor proliferation, angiogenesis, invasion, metastasis, drug resistance, and tumor immune escape.

Figure 3.

Figure 3

Potential biological functions of Exos-ncRNAs in the TME of GICs

This schematic diagram presents the impact of Exos-ncRNAs on cellular and molecular mechanisms involving GIC pathogenesis. Exos in GICs promote tumor progression and metastasis by transferring ncRNAs that enhance the expression of oncogenes and target tumor suppressor genes. They also induce onco-epigenetic modifications that induce heterogeneity and drug resistance in GIC by upregulating methylation of tumor suppressor genes and enhancing transcription of tumor proliferation genes. Furthermore, Exos-ncRNAs enhance proliferation and differentiation of immunosuppressive Tregs, and at the same time inhibit innate and adaptive anti-tumor immune responses. Exos can also transfer ncRNAs that induce CD8+ T cell exhaustion by inducing the upregulation of immune checkpoints such as PD-1 and TIM-3.

Table 1.

Summary of key studies on TAM-Exos-ncRNAs driving GIC progression

ncRNA Expression Tumor type M1/M2 Target Gene Mechanism Cell type Reference
miR-125a and b. Low HCC TAMs Significant decrease in CD90 expression Promotes HCC cell proliferation and stem cell properties. Human HCC cell lines Wang et al.62
miR-326 High HCC M1-TAMs Significant reduction in NF-κB and CD206 expression Suppresses proliferation, migration, and invasion as well as advances apoptosis of HCC through down-regulating NF-κB expression. Human HCC cell lines Bai et al.63
miR-660-5p High HCC M2-TAM Low levels of KLF3 Augments the development of liver cancer. Human cell line (HepG2). Tian et al.64
miR-27a-3p High HCC M2-TAM Downregulation of thioredoxin-interacting protein (TXNIP). Promotes stemness, proliferation, migration, invasion, and drug resistance of HCC cells. Human HCC cells Li et al.65
miR-92a-2-5p High HCC M2-TAMs Decreased level of androgen receptor (AR) Enhances the HCC invasion via regulating the AR/PHLPP/p-AKT/β-catenin signaling pathway. Human liver cancer cells Liu et al.66
lncMMPA High HCC M2-TAMs Decreased level of MiR-548s and increased level of ALDH1A3 Regulates glucose metabolism and cell proliferation in HCC cells. Human HCC cell lines. Xu et al.67
miR-223 and miR-142 High HCC MΦs Decreased levels of stathmin-1 (STMN1) and insulin-like growth factor-1 receptor
(IGF-1R)
Inhibits the cellular proliferation of HCC. Human HCC cell lines Aucher et al.68
miR-365 High PDAC M2-TAMs Significant decrease in B-cell translocation gene 2 (BTG2) and increase in the FAK/AKT. Promote cell proliferation, migration, and invasion by activating the FAK/AKT pathway. Human PDAC cell lines (PANC-1, BxPC-3, MIA Paca-2, and Capan-2). Li et al.52
miR-365 High PDAC M2-TAMs Increase levels of NTP/CDA Enhance gemcitabine resistance in PDAC. Animal PDAC cell line (PDAC K989 cell) Binenbaum et al.69
miR-221-5p
and miR-155-5p
High PDAC M2-TAMs low levels of E2F2 Promote tumor growth and angiogenesis. Mouse PDAC cell line Yang et al.70
miR-501-3p High PDAC M2-TAMs Inhibits TGFBR3 Enhances the invasion and migration of PDAC via the activating TGF-β pathway. Human PDAC cell lines Yin et al.53
lncRNA SBF2-AS1 Low PDAC M2-TAMs Increases miR-122-5p and restricts X-linked inhibitor of apoptosis protein (XIAP). Inhibits the tumorigenic ability of PC cells. Human PaC cell lines Yin et al.59
miR-16-5p High GC M1-TAMs Decrease PD-L1 T cell immune responses are activated by miR-16-5p through regulating PD-L1. Human GC Cell Line (AGS and NCI-N87). Li et al.71
miR-223 High GC TAMs Inhibit F box/FBXW7 Enhances doxorubicin resistance. Human GC cell line Gao et al.37
miR-21 High GC M2-TAMs Downregulation of PTEN Promotes chemotherapy (cisplatin) resistance and inhibits cell apoptosis. Mouse GC MFC cell and human GC MGC-803 cell lines. Zheng et al.72
miR-487a High GC M2-TAMs Downregulation of T cell intracellular antigen-1 (TIA1) Enhances tumor progression of GC cells. HS-746 T cells line (human gastric) Yang et al.51
miR-183-5p High CRC M2-TAMs Inhibits thioesterase superfamily member 4 (THEM4) Promotes CRC cell proliferation, invasion, and metastasis by facilitating the AKT/NF-κB -signaling pathway. Human CC cell lines. Zhang et al.73
miR-186-5p High CC M2-TAMs Inhibit DLC1 Promotes the proliferation and motility of CC cells. Human CC cell lines Guo et al.74
MiR-193b-3p High PaC M2- MΦs Low level of TRIM62 Promotes proliferation, invasion, migration, and glutamine uptake of PaC cells in vivo and in vitro. SW1990 cell line (Human pancreatic adenocarcinoma cell line) Zhang et al.75

Hepatocellular cancer

M2-TAMs modulate various factors in the TME of HCC to facilitate cancer progression. ncRNAs derived from M2-TAMs-Exos have been reported to upregulate immunosuppressive responses, supporting HCC initiation and progression via transferring miR-92a-2-5p.66 This study clarified that miR-92a-2-5p can enhance HCC invasion by decreasing the expression of androgen receptors (AR) by altering PHLPP/p-AKT/β-catenin signaling.66 It is believed that sex hormones, including androgen, interact with immune cells such as natural killer cells, monocytes, macrophages, and lymphocytes through their sex hormone receptors, which regulate chemokine and cytokine production that affect immune cell proliferation, differentiation, and maturation.76 Further studies demonstrated that overexpression of M2-TAMs-Exos-miR-27a-3p plays an important role in enhancing HCC cell stemness, invasion, and migration by targeting thioredoxin interacting protein (TXNIP).65 Additionally, M2-TAMs-Exos-miR-660-5p was reported to promote HCC by targeting kruppel-like factor 3 (KLF3).77 It has been shown that lncMMPA is expressed more in TAMs than in HCC cells.78 Interestingly, increased levels of lncMMPA play key roles in the polarization of M2-MΦs.67 Functionally, M2-TAMs-Exos-lncMMPA regulates glucose metabolism in HCC, which promotes tumor cell proliferation.67 Based on these studies, M2-TAMs-Exos-ncRNAs are important factors in the development of liver cancer through suppressing immune cells and accelerating cancer cell proliferation, which was linked with precancerous lesions and inhibition of tumor suppressor genes. Thus, they may have the potential to be used in the diagnosis, therapeutics, as well as management of liver cancer.

Gastric and esophageal cancers

The role of M2-TAMs-Exos-ncRNAs in the progression of GC has been well established. They play diverse roles in the initiation and progression of tumors. A recent study showed that Exos-miR-487a derived from M2-TAMs modulated gene expression of TIA1 in vivo and induced cell proliferation and progression of GC cells.51 Further to that, M2-TAMs-Exos-miR-21 inhibited GC cell apoptosis by reducing the expression of PTEN, leading to activating PI3K/AKT pathway.72 Another study reported that miR-21 acts as a tumor promoter by targeting the programmed cell death 4 (PDCD4) gene and preventing GC cell apoptosis by inhibiting the expression of PDCD4.79 Evidence supports that miR-21 plays a key role in regulating tumor suppressor genes and apoptosis-associated genes through enhancing cell survival and conferring resistance to therapies. PTEN and PDCD4 are known as tumor suppressor genes, thus loss of these genes leads to the activation of anti-apoptotic signaling, which is involved in cancer progression and chemoresistance. Other studies revealed that M2-TAMs-Exos-miR-588 contributed to enhancing GC cell proliferation and inhibiting apoptosis by targeting cylindromatosis (CYLD).80 In EC, M2-TAMs-lncRNA-AFAP1-AS1 reduced the level of miR-26a and increased the expression of transcription factor 2 (ATF2), which induces cancer cell migration, invasion, and metastasis.54 Altogether, M2-TAMs-Exos-ncRNAs in GC mainly induce drug resistance, besides eliciting immunosuppression and tumor cell proliferation, and attempts to target them have potential to inhibit tumor growth and promote anti-GC responses.

Pancreatic ductal adenocarcinoma (PDAC)

M2-TAMs-Exos-ncRNAs were linked to the aggressive growth of PDAC. M2-TAMs-Exos-miR-501-3p promoted PDAC by inhibiting transforming growth factor β-receptor-3 (TGFBR3), which triggers tumor cell migration and metastasis.53 Interestingly, TGF-β showed a role in the enhancement of TAMs-Exos-miR-501-3p, which participates in the initiation of PDAC growth by inhibiting the inflammatory immune response and eliciting immunosuppression in the TME. A recent study showed that M2-TAMs-Exos-miR-365 promoted PDAC progression by inhibiting the expression of B-cell translocation gene 2 (BTG2), leading to activating the FAK/AKT signaling pathway.52 Another study reported that M2-TAMs-Exos-miR-155-5p and miR-221-5p contributed to the promotion of tumor growth and angiogenesis of PDAC by targeting E2F2.70 E2F2 is a member of the E2F family of transcription factors, that play an important role in the regulation of the cell cycle progression,81 and loss of E2F2 expression accelerates tumor growth and metastasis.82 Moreover, M2-TAM-Exos-miR-21-5p was highly expressed in PaC and promoted tumor progression through enhancing PaC stem cell activity and differentiation via down-regulation of KLF3.83 MiR-21 is considered as an oncogenic effect in different tumors, and its overexpression can promote solid tumor development.84 Thus, miR-21 inhibition may reduce fibrosis and suppress populations of immunosuppressive cells simultaneously, thereby slowing premalignant progression and shrinking the tumors.85 Overall, M2-TAMs-Exos-ncRNAs are essential for tumor growth, development, and in drug resistance. Thus, new findings and insights on M2-TAMs-Exos-ncRNAs could improve clinical responses in PDAC.

Colorectal cancer (CRC)

Studies on the M2-TAMs-Exos-ncRNAs in CRC are few. Although limited, many findings highlighted the key role of TAMs in the TME of CRC. An in vivo study revealed that M2-TAMs-Exos-miR-186-5p promoted CRC development and progression through targeting DLC1.74 Furthermore, M2-TAMs-Exos-microRNA-155-5p was linked to the growth of CRC due to enhancing immune escape by downregulating the expression of ZC3H12B.86 ncRNAs also play a key role in regulating the expression of proteins, which are required for tumor development. For instance, in CRC, primary cancer cells release miR-21-containing Exos, which can be transferred into the liver via circulation and engulfed by MΦs.87 A recent study reported that M2-TAMs-Exos-miR-155-5p and M2-TAMs-Exos-miR-21-5p promoted CRC invasion and migration by targeting the transcription activator BRG1.44 Furthermore, miR-183-5p was highly expressed in M2-TAMs-Exos.73 This miR-183-5p is transferred into CRC cells through Exo and promotes tumor cell proliferation and metastasis by activating AKT/NF-κB pathway through targeting thioesterase superfamily member 4 (THEM4).73 Overall, M2-TAMs-Exos-ncRNAs are a cluster of tumorigenic enhancers in CRC, suggesting the need for further studies exploring their potentials as molecular targets.

Immunosuppressive impact of M2-Exos-ncRNAs in the TME of GIC

The immunosuppressive role of M2 in the TME has been recognized by several studies, which is due to different factors including mitochondrial metabolism, release of anti-inflammatory agents such as CCL3, CCL4, arginase 1, TGF-β, and IL-10, and Exos carrying immunomodulatory ncRNA,15,88,89,90 as explained in Figures 2 and 3. Importantly, there is little literature describing the impact of released Exos-ncRNAs that mediate the immunosuppressive function of M2-TAMs in GIC. In CRC, Exos-miR-203 drives M2-TAMs tumoral functions.91 Exos-miR-203 was reported as a regulator of DC immune responses by upregulating the expression of TLR4.92 Thus, M2-TAM-miR-203 may not only regulate tumor cell progression but also induces immunosuppression through regulating DC functions in the TME.93 In addition, levels of miR-29a-3p were downregulated in some GIC types like laryngocarcinoma, GC, and HCC,94,95,96 while expression of miR-21-5p was upregulated.97 However, immune modulatory effects of these miRNAs have not been elucidated in GIC. However, in epithelial ovarian cancer, microarray analysis of M2-Exos-ncRNAs revealed that M2-Exos are enriched with miR-29a-3p and miR-21-5p, which increase the ratio of Treg to Th17 by suppressing STAT3 signaling, leading to enhance tumor progression.98 Furthermore, transferring miR-146a from HCC cells to TAM induces M2 phenotype that suppresses T cell activation by inducing exhaustion through upregulating the expression of PD-1.99 Further studies reported that expression of Exos-miR-130b-3p serves as an inhibitor of extracellular cold-inducible RNA-binding protein (eCIRP), which can be released during sepsis .100 Adding to that, a recent microarray study linked the expression of miR-223, miR-124, miR-34a, miR-132, miR-125a-5p, let-7c, and miR-146a to M2 polarization and immunosuppression function.101 Most of these miRNAs are associated with GIC progressions such as miR-125a-5p and miR-124 in GC,102,103 and miR-132 in PDAC.104 Moreover, inhibition of miR-223 showed significant association with increased pro-inflammatory cytokines such as IL-1β and IL-6.105 In colon cancer, M2-TAM-Exos enriched with miR-183-5p were suggested to promote regulatory effects that support tumor growth by targeting THEM4 and activated Akt and NF-κB pathways.73 Further studies showed that M2-TAMs-Exos carrying miR-21 enhance the immune escape of tumor cells by modulating paternally expressed gene 3 (PEG3).106 However, studies revealing mechanisms of M2-Exos-ncRNAs in promoting immunosuppression in GIC are very limited and the available studies are not fully clear. Therefore, a better understanding of M2-Exos-ncRNA immunosuppressive functions is needed to develop new therapeutic targets using modulation of the immunosuppressive role of M2-TAMs in the TME of GIC.

Role of M2-Exos-ncRNAs in drug resistance of GIC

M2-TAMs-Exos induce chemotherapy resistance by modulating the expression of special genes such as mitogen-activated protein kinase 8 interacting protein 3 (MAPK8IP3), or by increasing the level of competitive endogenous RNAs (ceRNAs) such as MSTRG.292666.16 that sponge miR-6386-5p and thus induce MAPK8IP3 overexpression, leading to chemotherapy resistance.107 Recent studies reported that M2-Exos-ncRNAs can increase tumor cell chemotherapy resistance. It has been shown that M2-TAM-miR-223 increased resistance to doxorubicin and oxaliplatin by inhibiting F box and WD repeat domain-containing-7 (FBXW7) pathway in GC.37 Further to that, M2-TAMs-Exos-miR-21 promoted cisplatin resistance in GC by inhibiting apoptosis by activating PTEN/PI3K/AKT pathway.72 In addition, M2-TAMs-miR-588 promoted cisplatin resistance via targeting cylindromatosis (CYLD).80 lncRNAs have been demonstrated to be associated with chemotherapy sensitivity through modulating autophagy.108 Decrease in lncRNA-CRNDE expression in M2-Exos was associated with chemotherapy resistance in GC, whereas its increase revealed the opposite effects. A recent study has shown that the high level of Exos-lncRNA-CRNDE enhanced cell proliferation and tumor progression by targeting the expression of PTEN, and inhibiting CRNDE in M2-Exos enhanced the sensitivity to cisplatin in GC.36 Another study showed that the knock-down of M2-Exos-miR-223 reduced resistance to doxorubicin in GC.37 In an in vitro study, M2-TAM-miR-365 increased PDAC cell resistance to gemcitabine by increasing the triphospho-nucleotide pool and inducing the expression of cytidine deaminase (CDA), which is capable of inactivating intracellular gemcitabine and decreasing PDAC cell sensitivity to this therapy.69 Thus, further investigations of M2-TAMs-Exos-ncRNAs could identify potential predictive biomarkers for the response to anti-tumor drugs, as well as discover promising targets for improving sensitivity to anti-cancer drugs.

Role of Exos-ncRNAs in T cell recruitment and function in the TME of GICs

Exos-ncRNAs play important roles in regulating the recruitment and function of T cells into the TME of GICs. In recent studies, the low expression of Exos-miR-140 in GICs was significantly linked to tumor progression, metastasis, and immunosuppression.109,110 Interestingly, upregulation of PD-L1 and decreased CD8+ T cells in the TME of GC might be promoted by reduced expression of miR-140.111 Overexpression of miR-140 inhibited PD-L1 and Tregs in GC mice model, while it increased the recruitment of CD8+ T cells, leading to inhibition of GC growth.111 Another study investigating the relationship between Exos-miR-16-5p and anti-tumor immune responses in GC revealed that Exos-miR-16-5p targets PD-L1 and triggers T cell immune responses, which inhibit tumor formation.71 The investigation of Exos-miR-206 function in GC and HCC revealed that miR-206 has a key suppressor role in the TME of these tumors.112,113 Interestingly, overexpression of miR-206 promoted the recruitment of CD8+ T cells to the TME by inducing M1 polarization through increasing CCL2.114 In nasopharyngeal carcinoma (NPC), there was a significant upregulation of miR-24-3p in the serum that inhibited proliferation of Th1 and Th17 but induced differentiation of Tregs by inhibiting fibroblast growth factor 11 (FGF11) gene in T cells.115 A study investigated YAP1 transcription by lncRP11-323N12.5 in GC and T cells by binding to c-MYC in the YAP1 promoter region concluded that Exos-lncRP11-323N12.5 upregulates YAP1, which enhances Treg differentiation and immunosuppression in GC.116 Furthermore, upregulation of miR-448 in CRC suppresses apoptosis of CD8+ T cells by inhibiting the expression of IDO1.117 Overall, Exos-ncRNAs orchestrate several tumorigenesis and immunomodulation activities in the TME of GICs, as summarized in Table 2.

Table 2.

Biological functions of exosomal ncRNAs in GIC

Exosomal ncRNAs Biological and Mechanism function Target cells or molecules Source of exosome Cancer type Reference
miR-223 Enhances chemo-resistance to doxorubicin GC cells MΦs GC Gao et al.37
circSHKBP Promotes tumor progression (via proliferation, angiogenesis, migration, and invasion) GC cells GC cells GC Xie et al.118
lncRNA-PCGEM1 Promotes invasion and migration of other GC cells GC cells (normoxic cells) GC cells (hypoxic cells) GC Piao et al.119
miR-519a-3p Promotes liver metastasis HCC GC cells GC Qiu et al.120
miR-934 Promotes liver metastasis HCC CRC cells CRC Zhao et al.45
CircRNA-100338 Promotes HCC metastasis by enhancing angiogenesis and invasiveness HCC HCC cells HCC Huang et al.121
miR-106b-3p Promotes invasiveness, EMT, and metastasis Deleted in liver cancer-1 (DLC-1) CRC cells CRC Liu et al.122
miR-431-5p Promotes tumor progression CRC cells Human umbilical cord mesenchymal stem cells (hUCMSCs) CRC Qu et al.123
LncR-RPPH1 Promotes metastasis TUBB3 and TAM-M2 polarization CRC cells CRC Liang et al.124
miR-30b-5p Promotes angiogenesis Glypican-1 (GJA1) Hypoxic PaC cells PDAC Chen et al.125
miR-21 Promotes tumor progression Hepatic stellate cells HCC cells HCC Zhou et al.126
miR-19a Involved in pancreatic cancer pathogenesis, which is associated with diabetes Adenylyl cyclase-1 ADCY1 and exchange protein directly activated by Camp-2 EPAC2 PaC cells PaC Pang et al.127
miR-21-5p Promotes liver metastasis through inducing liver M1- MΦs polarization HCC CRC cells CRC Shao et al.87
miR-320a Enhances tumor progression by promoting M2-MΦs polarization. Regulates PTEN/PI3Kγ Cancer-associated fibroblast PaC Zhao et al.128
miR-34 Suppresses growth and invasion of gastric cancer Gastric cancer Cancer-associated fibroblast GC Shi et al.129
miR-139 Suppresses gastric cancer progression Matrix metalloproteinase-11 (MMP11) Cancer-associated fibroblast GC Xu et al.130
miR-4454 Enhances tumor growth and vascularization of HepG2 cells, as well as accelerating cycle arrest, apoptosis, and oxidative stress of HepG2 cells Vps4A and Rab27A HCC HCC Lin et al.131
miR-4800-3P Promotes HCC progression by regulating the Hippo pathway via targeting STK25 Targeting STK25 HCC cells HCC Lin et al.132
lncR-H19 Promotes HCC progression after propofol treatment by upregulating the microRNA-520a-3p/LIMK1 axis Targeting the microRNA-520a-3p/LIMK1 HCC cells HCC Yu et al.133
CircUHRF1 Contributes to immunosuppression through inhibiting NK cell function and may also cause resistance to anti-PD-1 immunotherapy NK cells HCC cells HCC Zhang et al.134
Circ-002136 Promotes progression of HCC Targeting miR-19a-3p/RAB1A pathway. HCC cells HCC Yuan et al.135
miR-203 Promotes tumor metastasis by promoting differentiation of monocytes to M2-MΦs Monocytes CRC cells CRC Takano et al.91
miR-203 Contributes to dysfunction of DCs through regulating TLR4 Targeting TLR4 in DCs PaC cells PaC Zhou et al.92
lncRNA ENST00000560647 Promotes tumor progression and immune escape DCs PaC cells PaC Chen et al.136
miR-301a-3p Promotes pancreatic cancer via polarizing MΦs MΦs Hypoxic PaC cells PaC Wang et al.50
miR-107 Induces expansion of MDSCs and ARG1 expression by inhibiting the DICER1 and PTEN expression MDSC GC cells GC Ren et al.137
miR-10a-5p Promotes tumor progression via inducing PD1+ MΦs population Monocyte’s differentiation into MΦ GC cells GC Zhu et al.138

Conclusions and future directions

Several studies reported the elevation of TAM levels in GIC, which was usually linked to tumor progression and lower survival rates. TAMs are essential in GIC growth, invasion, immune regulation, angiogenesis, metastasis, and progression. TAM polarization into M2 is linked to poor prognoses and drug resistance in GIC. Polarization of M2-TAMs is mainly mediated by tumor cells via released Exos and other immunomodulators that induce the secretion of M-CSF. The polarized TAMs then promote tumor progression and induce most of the resident TAMs into M2 phenotype, leading to immune suppression, drug resistance, and tumor progression. As TAMs have the capability to switch polarization into different functional phenotypes, inducing M1 phenotype could modulate the released Exos with anti-tumor functional ncRNAs, which can activate immune responses, inhibit tumor growth, and improve response to chemotherapeutic interventions. However, further explorations of the differences between Exos-ncRNAs released by M2-MΦs and M1-MΦs are still needed.

Little information is available about the molecular mechanisms mediated by M2-Exos-ncRNAs in the TME of GIC, which are supposed to orchestrate immunomodulation and tumor progression. Recently, the importance of Exos-ncRNA as targets for modulating the TME and immune responses in GIC was demonstrated by several studies. M2-TAMs release Exos-ncRNAs, which are linked to the underlying mechanisms of tumorigenesis, including tumor angiogenesis, invasion, immunosuppression, tumor cell proliferation, and drug resistance. The role of Exos-ncRNAs in molecular mechanisms supporting GIC progression has been the subject of numerous recent studies. M2-TAMs-Exos-ncRNAs demonstrated functional pathological effects in GIC by regulating tumor cell stemness, invasion, and metastasis. Further to that, recent studies reported a link between M2-TAMs-Exos-ncRNAs and modulation of tumor cells and immune cells, resulting in the promotion of immunosuppression and tumorigenesis. Moreover, M2-Exos-ncRNAs can induce tumor immune evasion and enhance resistance to chemotherapy through alteration of gene expression in cancer and immune cells, which can be used as diagnostic biomarkers and/or therapeutic targets in GICs. Further elucidation of the immunosuppressive roles of M2-Exos-ncRNAs is ultimately needed to provide a clear understanding and new therapeutic targets, relying on modulation of the immunosuppressive role of M2-TAMs in the TME of GIC. Furthermore, it is possible to reverse Exos-ncRNA effects with antagonists; therefore, developing effective and clinically valuable antagonists in GIC has important potential.

The focus on M2-TAMs-Exos-ncRNAs is very important to explore critical mechanisms in GICs. We propose that studying molecular mechanisms underlying M2-TAMs-Exos-ncRNAs functions in the TME of GIC would uncover novel targets that could lead to promising clinical interventions. Exos from M2-MΦs carry many signaling molecules, such as ncRNAs, which regulate target cell biology and cellular functions through unknown mechanisms. Thus, it may be possible to investigate how target cells take up Exos-ncRNAs, how they modulate gene expression, and how they affect cell behavior; these could serve as potential diagnostic and/or prognostic biomarkers, and to respond to anti-cancer treatments, in addition to discovering novel targets to improve anti-tumor drug sensitivity. Overall, further investigation of Exos-ncRNAs released by M2-TAMs is a promising area of research with many potential future directions.

Acknowledgments

Funding: This work was supported by the Natural and Medical Sciences Research Center, the University of Nizwa, and the University of Salford.

Author contributions

Conceptualization: A.M., M.A., and E.E.; Data curation: A.M. and M.A.; Drawing Figures: A.M. and M.A.; Writing original draft: A.M. and M.A.; Funding acquisition: E.E.; Supervision: E.E.; Validation: A.M., M.A., and E.E.; Revision: E.E.; All authors reviewed the whole work and approved the final version of the manuscript.

Declaration of interests

The authors declare that there are no financial or nonfinancial conflicts of interest related to this work.

References

  • 1.Arnold M., Abnet C.C., Neale R.E., Vignat J., Giovannucci E.L., McGlynn K.A., Bray F. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 2020;159:335–349.e15. doi: 10.1053/j.gastro.2020.02.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sung H., Siegel R.L., Rosenberg P.S., Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health. 2019;4:e137–e147. doi: 10.1016/S2468-2667(18)30267-6. [DOI] [PubMed] [Google Scholar]
  • 3.Kanda M., Mizuno A., Fujii T., Shimoyama Y., Yamada S., Tanaka C., Kobayashi D., Koike M., Iwata N., Niwa Y., et al. Tumor infiltrative pattern predicts sites of recurrence after curative gastrectomy for stages 2 and 3 gastric cancer. Ann. Surg Oncol. 2016;23:1934–1940. doi: 10.1245/s10434-016-5102-x. [DOI] [PubMed] [Google Scholar]
  • 4.Kolbeinsson H., Hoppe A., Bayat A., Kogelschatz B., Mbanugo C., Chung M., Wolf A., Assifi M.M., Wright G.P. Recurrence patterns and postrecurrence survival after curative intent resection for pancreatic ductal adenocarcinoma. Surgery. 2021;169:649–654. doi: 10.1016/j.surg.2020.06.042. [DOI] [PubMed] [Google Scholar]
  • 5.Vakiani E., Shah R.H., Berger M.F., Makohon-Moore A.P., Reiter J.G., Ostrovnaya I., Attiyeh M.A., Cercek A., Shia J., Iacobuzio-Donahue C.A., et al. Local recurrences at the anastomotic area are clonally related to the primary tumor in sporadic colorectal carcinoma. Oncotarget. 2017;8:42487–42494. doi: 10.18632/oncotarget.17200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Vitale I., Manic G., Coussens L.M., Kroemer G., Galluzzi L. Macrophages and metabolism in the tumor microenvironment. Cell Metab. 2019;30:36–50. doi: 10.1016/j.cmet.2019.06.001. [DOI] [PubMed] [Google Scholar]
  • 7.Murray P.J., Wynn T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011;11:723–737. doi: 10.1038/nri3073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ostuni R., Kratochvill F., Murray P.J., Natoli G. Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol. 2015;36:229–239. doi: 10.1016/j.it.2015.02.004. [DOI] [PubMed] [Google Scholar]
  • 9.Orekhov A.N., Orekhova V.A., Nikiforov N.G., Myasoedova V.A., Grechko A.V., Romanenko E.B., Zhang D., Chistiakov D.A. Monocyte differentiation and macrophage polarization. Vessel Plus. 2019;3:10. doi: 10.20517/2574-1209.2019.04. [DOI] [Google Scholar]
  • 10.Forssell J., Oberg A., Henriksson M.L., Stenling R., Jung A., Palmqvist R. High macrophage infiltration along the tumor front correlates with improved survival in colon cancer. Clin. Cancer Res. 2007;13:1472–1479. doi: 10.1158/1078-0432.Ccr-06-2073. [DOI] [PubMed] [Google Scholar]
  • 11.Zhou Q., Peng R.-Q., Wu X.-J., Xia Q., Hou J.-H., Ding Y., Zhou Q.-M., Zhang X., Pang Z.-Z., Wan D.-S., et al. The density of macrophages in the invasive front is inversely correlated to liver metastasis in colon cancer. J. Transl. Med. 2010;8:13. doi: 10.1186/1479-5876-8-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jeannin P., Paolini L., Adam C., Delneste Y. The roles of CSFs on the functional polarization of tumor-associated macrophages. FEBS J. 2018;285:680–699. doi: 10.1111/febs.14343. [DOI] [PubMed] [Google Scholar]
  • 13.Wan S., Kuo N., Kryczek I., Zou W., Welling T.H. Myeloid cells in hepatocellular carcinoma. Hepatology. 2015;62:1304–1312. doi: 10.1002/hep.27867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mantovani A., Allavena P. The interaction of anticancer therapies with tumor-associated macrophages. J. Exp. Med. 2015;212:435–445. doi: 10.1084/jem.20150295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.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. doi: 10.1155/2012/948098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Shu Y., Cheng P. Targeting tumor-associated macrophages for cancer immunotherapy. Biochim. Biophys. Acta. Rev. Cancer. 2020;1874:188434. doi: 10.1016/j.bbcan.2020.188434. [DOI] [PubMed] [Google Scholar]
  • 17.Franklin R.A., Li M.O. Ontogeny of tumor-associated macrophages and its implication in cancer regulation. Trends Cancer. 2016;2:20–34. doi: 10.1016/j.trecan.2015.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Taniyama D., Taniyama K., Kuraoka K., Zaitsu J., Saito A., Nakatsuka H., Sakamoto N., Sentani K., Oue N., Yasui W. Long-term follow-up study of gastric adenoma; tumor-associated macrophages are associated to carcinoma development in gastric adenoma. Gastric Cancer. 2017;20:929–939. doi: 10.1007/s10120-017-0713-x. [DOI] [PubMed] [Google Scholar]
  • 19.Fu X.T., Dai Z., Song K., Zhang Z.J., Zhou Z.J., Zhou S.L., Zhao Y.M., Xiao Y.S., Sun Q.M., Ding Z.B., Fan J. Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway. Int. J. Oncol. 2015;46:587–596. doi: 10.3892/ijo.2014.2761. [DOI] [PubMed] [Google Scholar]
  • 20.Lin Y., Xu J., Lan H. Tumor-associated macrophages in tumor metastasis: biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019;12:76. doi: 10.1186/s13045-019-0760-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kalluri R., LeBleu V.S. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977. doi: 10.1126/science.aau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Colombo M., Raposo G., Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014;30:255–289. doi: 10.1146/annurev-cellbio-101512-122326. [DOI] [PubMed] [Google Scholar]
  • 23.Liu W.Z., Ma Z.J., Kang X.W. Current status and outlook of advances in exosome isolation. Anal. Bioanal. Chem. 2022;414:7123–7141. doi: 10.1007/s00216-022-04253-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ferguson S.W., Megna J.S., Nguyen J. In: Diagnostic and Therapeutic Applications of Exosomes in Cancer. Amiji M., Ramesh R., editors. Academic Press; 2018. 3 - Composition, physicochemical and biological properties of exosomes secreted from cancer cells; pp. 27–57. [DOI] [Google Scholar]
  • 25.Xu R., Greening D.W., Zhu H.J., Takahashi N., Simpson R.J. Extracellular vesicle isolation and characterization: toward clinical application. J. Clin. Invest. 2016;126:1152–1162. doi: 10.1172/jci81129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Keller S., Ridinger J., Rupp A.K., Janssen J.W.G., Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 2011;9:86. doi: 10.1186/1479-5876-9-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Santos P., Almeida F. Role of exosomal miRNAs and the tumor microenvironment in drug resistance. Cells. 2020;9:1450. doi: 10.3390/cells9061450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang Z., Yin J., Lu C., Wei Y., Zeng A., You Y. Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J. Exp. Clin. Cancer Res. 2019;38:166. doi: 10.1186/s13046-019-1139-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fu X., Liu M., Qu S., Ma J., Zhang Y., Shi T., Wen H., Yang Y., Wang S., Wang J., et al. Exosomal microRNA-32-5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J. Exp. Clin. Cancer Res. 2018;37:52. doi: 10.1186/s13046-018-0677-7. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 30.Gao Q., Lei F., Zeng Q., Gao Z., Niu P., Junnan, Ning, Li J., Zhang J. Functional passenger-strand miRNAs in exosomes derived from human colon cancer cells and their heterogeneous paracrine effects. Int. J. Biol. Sci. 2020;16:1044–1058. doi: 10.7150/ijbs.40787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tellez-Gabriel M., Heymann D. Exosomal lncRNAs: the newest promising liquid biopsy. Cancer Drug Resist. 2019;2:1002–1017. doi: 10.20517/cdr.2019.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sharma A. Chemoresistance in cancer cells: exosomes as potential regulators of therapeutic tumor heterogeneity. Nanomedicine (Lond) 2017;12:2137–2148. doi: 10.2217/nnm-2017-0184. [DOI] [PubMed] [Google Scholar]
  • 33.Ronnegren A.L. Extracellular vesicles as potential mediators of epigenetic reprogramming. For. Immunopathol. Dis. Therap. 2015;6:133–141. doi: 10.1615/ForumImmunDisTher.2016016526. [DOI] [Google Scholar]
  • 34.Qian Z., Shen Q., Yang X., Qiu Y., Zhang W. The role of extracellular vesicles: an epigenetic view of the cancer microenvironment. BioMed Res. Int. 2015;2015:649161. doi: 10.1155/2015/649161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhang Z., Hu J., Ishihara M., Sharrow A.C., Flora K., He Y., Wu L. The miRNA-21-5p payload in exosomes from M2 macrophages drives tumor cell aggression via PTEN/Akt signaling in renal cell carcinoma. Int. J. Mol. Sci. 2022;23:3005. doi: 10.3390/ijms23063005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Xin L., Zhou L.Q., Liu C., Zeng F., Yuan Y.W., Zhou Q., Li S.H., Wu Y., Wang J.L., Wu D.Z., Lu H. Transfer of LncRNA CRNDE in TAM-derived exosomes is linked with cisplatin resistance in gastric cancer. EMBO Rep. 2021;22:e52124. doi: 10.15252/embr.202052124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gao H., Ma J., Cheng Y., Zheng P. Exosomal transfer of macrophage-derived miR-223 confers doxorubicin resistance in gastric cancer. OncoTargets Ther. 2020;13:12169–12179. doi: 10.2147/ott.S283542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Laviron M., Boissonnas A. Ontogeny of tumor-associated macrophages. Front. Immunol. 2019;10:1799. doi: 10.3389/fimmu.2019.01799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhou K., Cheng T., Zhan J., Peng X., Zhang Y., Wen J., Chen X., Ying M. Targeting tumor-associated macrophages in the tumor microenvironment. Oncol. Lett. 2020;20:234. doi: 10.3892/ol.2020.12097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chen Q., Li Y., Gao W., Chen L., Xu W., Zhu X. Exosome-mediated crosstalk between tumor and tumor-associated macrophages. Front. Mol. Biosci. 2021;8:764222. doi: 10.3389/fmolb.2021.764222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jiang Z., Zhang Y., Zhang Y., Jia Z., Zhang Z., Yang J. Cancer derived exosomes induce macrophages immunosuppressive polarization to promote bladder cancer progression. Cell Commun. Signal. 2021;19:93. doi: 10.1186/s12964-021-00768-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Han C., Zhang C., Wang H., Zhao L. Exosome-mediated communication between tumor cells and tumor-associated macrophages: implications for tumor microenvironment. OncoImmunology. 2021;10:1887552. doi: 10.1080/2162402x.2021.1887552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li Y., Tan J., Miao Y., Zhang Q. MicroRNA in extracellular vesicles regulates inflammation through macrophages under hypoxia. Cell Death Discov. 2021;7:285. doi: 10.1038/s41420-021-00670-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lan J., Sun L., Xu F., Liu L., Hu F., Song D., Hou Z., Wu W., Luo X., Wang J., et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79:146–158. doi: 10.1158/0008-5472.Can-18-0014. [DOI] [PubMed] [Google Scholar]
  • 45.Zhao S., Mi Y., Guan B., Zheng B., Wei P., Gu Y., Zhang Z., Cai S., Xu Y., Li X., et al. Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J. Hematol. Oncol. 2020;13:156. doi: 10.1186/s13045-020-00991-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zongqiang H., Jiapeng C., Yingpeng Z., Chuntao Y., Yiting W., Jiashun Z., Li L. Exosomal miR-452-5p induce M2 macrophage polarization to accelerate hepatocellular carcinoma progression by targeting TIMP3. J. Immunol. Res. 2022;2022:1032106. doi: 10.1155/2022/1032106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen J., Zhang K., Zhi Y., Wu Y., Chen B., Bai J., Wang X. Tumor-derived exosomal miR-19b-3p facilitates M2 macrophage polarization and exosomal LINC00273 secretion to promote lung adenocarcinoma metastasis via Hippo pathway. Clin. Transl. Med. 2021;11:e478. doi: 10.1002/ctm2.478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang L., Wu J., Ye N., Li F., Zhan H., Chen S., Xu J. Plasma-derived exosome MiR-19b acts as a diagnostic marker for pancreatic cancer. Front. Oncol. 2021;11:739111. doi: 10.3389/fonc.2021.739111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lu Q., Wang X., Zhu J., Fei X., Chen H., Li C. Hypoxic tumor-derived exosomal Circ0048117 facilitates M2 macrophage polarization acting as miR-140 sponge in esophageal squamous cell carcinoma. OncoTargets Ther. 2020;13:11883–11897. doi: 10.2147/ott.S284192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.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 metastasis. Cancer Res. 2018;78:4586–4598. doi: 10.1158/0008-5472.Can-17-3841. [DOI] [PubMed] [Google Scholar]
  • 51.Yang X., Cai S., Shu Y., Deng X., Zhang Y., He N., Wan L., Chen X., Qu Y., Yu S. Exosomal miR-487a derived from m2 macrophage promotes the progression of gastric cancer. Cell Cycle. 2021;20:434–444. doi: 10.1080/15384101.2021.1878326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Li X., Xu H., Yi J., Dong C., Zhang H., Wang Z., Miao L., Zhou W. miR-365 secreted from M2 Macrophage-derived extracellular vesicles promotes pancreatic ductal adenocarcinoma progression through the BTG2/FAK/AKT axis. J. Cell Mol. Med. 2021;25:4671–4683. doi: 10.1111/jcmm.16405. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 53.Yin Z., Ma T., Huang B., Lin L., Zhou Y., Yan J., Zou Y., Chen S. Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. J. Exp. Clin. Cancer Res. 2019;38:310. doi: 10.1186/s13046-019-1313-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mi X., Xu R., Hong S., Xu T., Zhang W., Liu M. M2 macrophage-derived exosomal lncRNA AFAP1-AS1 and MicroRNA-26a affect cell migration and metastasis in esophageal cancer. Mol. Ther. Nucleic Acids. 2020;22:779–790. doi: 10.1016/j.omtn.2020.09.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Vasudevan D., Ryoo H.D. Regulation of cell death by IAPs and their antagonists. Curr. Top. Dev. Biol. 2015;114:185–208. doi: 10.1016/bs.ctdb.2015.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tu H., Costa M. XIAP's profile in human cancer. Biomolecules. 2020;10:1493. doi: 10.3390/biom10111493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pei Z.J., Zhang Z.G., Hu A.X., Yang F., Gai Y. miR-122-5p inhibits tumor cell proliferation and induces apoptosis by targeting MYC in gastric cancer cells. Pharmazie. 2017;72:344–347. doi: 10.1691/ph.2017.6404. [DOI] [PubMed] [Google Scholar]
  • 58.Xu Z., Liu G., Zhang M., Zhang Z., Jia Y., Peng L., Zhu Y., Hu J., Huang R., Sun X. miR-122-5p inhibits the proliferation, invasion and growth of bile duct carcinoma cells by targeting ALDOA. Cell. Physiol. Biochem. 2018;48:2596–2606. doi: 10.1159/000492702. [DOI] [PubMed] [Google Scholar]
  • 59.Yin Z., Zhou Y., Ma T., Chen S., Shi N., Zou Y., Hou B., Zhang C. Down-regulated lncRNA SBF2-AS1 in M2 macrophage-derived exosomes elevates miR-122-5p to restrict XIAP, thereby limiting pancreatic cancer development. J. Cell Mol. Med. 2020;24:5028–5038. doi: 10.1111/jcmm.15125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Maia J., Caja S., Strano Moraes M.C., Couto N., Costa-Silva B. Exosome-Based cell-cell communication in the tumor microenvironment. Front. Cell Dev. Biol. 2018;6:18. doi: 10.3389/fcell.2018.00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Xu Z., Chen Y., Ma L., Chen Y., Liu J., Guo Y., Yu T., Zhang L., Zhu L., Shu Y. Role of exosomal non-coding RNAs from tumor cells and tumor-associated macrophages in the tumor microenvironment. Mol. Ther. 2022;30:3133–3154. doi: 10.1016/j.ymthe.2022.01.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang Y., Wang B., Xiao S., Li Y., Chen Q. miR-125a/b inhibits tumor-associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90. J. Cell. Biochem. 2019;120:3046–3055. doi: 10.1002/jcb.27436. [DOI] [PubMed] [Google Scholar]
  • 63.Bai Z.-z., Li H.-y., Li C.-h., Sheng C.-l., Zhao X.-n. M1 macrophage-derived exosomal MicroRNA-326 suppresses hepatocellular carcinoma cell progression via mediating NF-κB signaling pathway. Nanoscale Res. Lett. 2020;15:221. doi: 10.1186/s11671-020-03432-8. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 64.Tian B., Zhou L., Wang J., Yang P. miR-660-5p-loaded M2 macrophages-derived exosomes augment hepatocellular carcinoma development through regulating KLF3. Int. Immunopharmacol. 2021;101:108157. doi: 10.1016/j.intimp.2021.108157. [DOI] [PubMed] [Google Scholar]
  • 65.Li W., Xin X., Li X., Geng J., Sun Y. Exosomes secreted by M2 macrophages promote cancer stemness of hepatocellular carcinoma via the miR-27a-3p/TXNIP pathways. Int. Immunopharmacol. 2021;101:107585. doi: 10.1016/j.intimp.2021.107585. [DOI] [PubMed] [Google Scholar]
  • 66.Liu G., Ouyang X., Sun Y., Xiao Y., You B., Gao Y., Yeh S., Li Y., Chang C. The miR-92a-2-5p in exosomes from macrophages increases liver cancer cells invasion via altering the AR/PHLPP/p-AKT/β-catenin signaling. Cell Death Differ. 2020;27:3258–3272. doi: 10.1038/s41418-020-0575-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Xu M., Zhou C., Weng J., Chen Z., Zhou Q., Gao J., Shi G., Ke A., Ren N., Sun H., Shen Y. Tumor associated macrophages-derived exosomes facilitate hepatocellular carcinoma malignance by transferring lncMMPA to tumor cells and activating glycolysis pathway. J. Exp. Clin. Cancer Res. 2022;41:253. doi: 10.1186/s13046-022-02458-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Aucher A., Rudnicka D., Davis D.M. MicroRNAs transfer from human macrophages to hepato-carcinoma cells and inhibit proliferation. J. Immunol. 2013;191:6250–6260. doi: 10.4049/jimmunol.1301728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Binenbaum Y., Fridman E., Yaari Z., Milman N., Schroeder A., Ben David G., Shlomi T., Gil Z. Transfer of miRNA in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Cancer Res. 2018;78:5287–5299. doi: 10.1158/0008-5472.Can-18-0124. [DOI] [PubMed] [Google Scholar]
  • 70.Yang Y., Guo Z., Chen W., Wang X., Cao M., Han X., Zhang K., Teng B., Cao J., Wu W., et al. M2 macrophage-derived exosomes promote angiogenesis and growth of pancreatic ductal adenocarcinoma by targeting E2F2. Mol. Ther. 2021;29:1226–1238. doi: 10.1016/j.ymthe.2020.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li Z., Suo B., Long G., Gao Y., Song J., Zhang M., Feng B., Shang C., Wang D. Exosomal miRNA-16-5p derived from M1 macrophages enhances T cell-dependent immune response by regulating PD-L1 in gastric cancer. Front. Cell Dev. Biol. 2020;8:572689. doi: 10.3389/fcell.2020.572689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zheng P., Chen L., Yuan X., Luo Q., Liu Y., Xie G., Ma Y., Shen L. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J. Exp. Clin. Cancer Res. 2017;36:53. doi: 10.1186/s13046-017-0528-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhang S., Li D., Zhao M., Yang F., Sang C., Yan C., Wang Z., Li Y. Exosomal miR-183-5p shuttled by M2 polarized tumor-associated macrophage promotes the development of colon cancer via targeting THEM4 mediated PI3K/AKT and NF-κB pathways. Front. Oncol. 2021;11:672684. doi: 10.3389/fonc.2021.672684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Guo J., Wang X., Guo Q., Zhu S., Li P., Zhang S., Min L. M2 macrophage derived extracellular vesicle-mediated transfer of MiR-186-5p promotes colon cancer progression by targeting DLC1. Int. J. Biol. Sci. 2022;18:1663–1676. doi: 10.7150/ijbs.69405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang K., Li Y.J., Peng L.J., Gao H.F., Liu L.M., Chen H. M2 macrophage-derived exosomal miR-193b-3p promotes progression and glutamine uptake of pancreatic cancer by targeting TRIM62. Biol. Direct. 2023;18:1. doi: 10.1186/s13062-023-00356-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bhatia A., Sekhon H.K., Kaur G. Sex hormones and immune dimorphism. Sci. World J. 2014;2014:159150. doi: 10.1155/2014/159150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zhao X.Q., Zhou H. Letter to the editor regarding "miR-660-5p-loaded M2 macrophages-derived exosomes augment hepatocellular carcinoma development through regulating KLF3". Int. Immunopharmacol. 2022;106:108554. doi: 10.1016/j.intimp.2022.108554. [DOI] [PubMed] [Google Scholar]
  • 78.Ma L., Hernandez M.O., Zhao Y., Mehta M., Tran B., Kelly M., Rae Z., Hernandez J.M., Davis J.L., Martin S.P., et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer Cell. 2019;36:418–430.e6. doi: 10.1016/j.ccell.2019.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wang J.J., Wang Z.Y., Chen R., Xiong J., Yao Y.L., Wu J.H., Li G.X. Macrophage-secreted exosomes delivering miRNA-21 inhibitor can regulate BGC-823 cell proliferation. Asian Pac. J. Cancer Prev. 2015;16:4203–4209. doi: 10.7314/apjcp.2015.16.10.4203. [DOI] [PubMed] [Google Scholar]
  • 80.Cui H.Y., Rong J.S., Chen J., Guo J., Zhu J.Q., Ruan M., Zuo R.R., Zhang S.S., Qi J.M., Zhang B.H. Exosomal microRNA-588 from M2 polarized macrophages contributes to cisplatin resistance of gastric cancer cells. World J. Gastroenterol. 2021;27:6079–6092. doi: 10.3748/wjg.v27.i36.6079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Infante A., Laresgoiti U., Fernández-Rueda J., Fullaondo A., Galán J., Díaz-Uriarte R., Malumbres M., Field S.J., Zubiaga A.M. E2F2 represses cell cycle regulators to maintain quiescence. Cell Cycle. 2008;7:3915–3927. doi: 10.4161/cc.7.24.7379. [DOI] [PubMed] [Google Scholar]
  • 82.Yuwanita I., Barnes D., Monterey M.D., O'Reilly S., Andrechek E.R. Increased metastasis with loss of E2F2 in Myc-driven tumors. Oncotarget. 2015;6:38210–38224. doi: 10.18632/oncotarget.5690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Chang J., Li H., Zhu Z., Mei P., Hu W., Xiong X., Tao J. microRNA-21-5p from M2 macrophage-derived extracellular vesicles promotes the differentiation and activity of pancreatic cancer stem cells by mediating KLF3. Cell Biol. Toxicol. 2022;38:577–590. doi: 10.1007/s10565-021-09597-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Rhim J., Baek W., Seo Y., Kim J.H. From molecular mechanisms to therapeutics: understanding MicroRNA-21 in cancer. Cells. 2022;11:2791. doi: 10.3390/cells11182791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chu N.J., Anders R.A., Fertig E.J., Cao M., Hopkins A.C., Keenan B.P., Popovic A., Armstrong T.D., Jaffee E.M., Zimmerman J.W. Inhibition of miR-21 regulates mutant KRAS effector pathways and intercepts pancreatic ductal adenocarcinoma development. Cancer Prev. Res. 2020;13:569–582. doi: 10.1158/1940-6207.Capr-20-0053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ma Y.S., Wu T.M., Ling C.C., Yu F., Zhang J., Cao P.S., Gu L.P., Wang H.M., Xu H., Li L., et al. M2 macrophage-derived exosomal microRNA-155-5p promotes the immune escape of colon cancer by downregulating ZC3H12B. Mol. Ther. Oncolytics. 2021;20:484–498. doi: 10.1016/j.omto.2021.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Shao Y., Chen T., Zheng X., Yang S., Xu K., Chen X., Xu F., Wang L., Shen Y., Wang T., et al. Colorectal cancer-derived small extracellular vesicles establish an inflammatory premetastatic niche in liver metastasis. Carcinogenesis. 2018;39:1368–1379. doi: 10.1093/carcin/bgy115. [DOI] [PubMed] [Google Scholar]
  • 88.Oyarce C., Vizcaino-Castro A., Chen S., Boerma A., Daemen T. Re-polarization of immunosuppressive macrophages to tumor-cytotoxic macrophages by repurposed metabolic drugs. OncoImmunology. 2021;10:1898753. doi: 10.1080/2162402x.2021.1898753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kumari N., Choi S.H. Tumor-associated macrophages in cancer: recent advancements in cancer nanoimmunotherapies. J. Exp. Clin. Cancer Res. 2022;41:68. doi: 10.1186/s13046-022-02272-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ahmad A. Epigenetic regulation of immunosuppressive tumor-associated macrophages through dysregulated microRNAs. Semin. Cell Dev. Biol. 2022;124:26–33. doi: 10.1016/j.semcdb.2021.09.001. [DOI] [PubMed] [Google Scholar]
  • 91.Takano Y., Masuda T., Iinuma H., Yamaguchi R., Sato K., Tobo T., Hirata H., Kuroda Y., Nambara S., Hayashi N., et al. Circulating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget. 2017;8:78598–78613. doi: 10.18632/oncotarget.20009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.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;292:65–69. doi: 10.1016/j.cellimm.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 93.Wu X.-q., Dai Y., Yang Y., Huang C., Meng X.-m., Wu B.-m., Li J. Emerging role of microRNAs in regulating macrophage activation and polarization in immune response and inflammation. Immunology. 2016;148:237–248. doi: 10.1111/imm.12608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Su J., Lu E., Lu L., Zhang C. MiR-29a-3p suppresses cell proliferation in laryngocarcinoma by targeting prominin 1. FEBS Open Bio. 2017;7:645–651. doi: 10.1002/2211-5463.12199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang X., Liu S., Cao L., Zhang T., Yue D., Wang L., Ping Y., He Q., Zhang C., Wang M., et al. miR-29a-3p suppresses cell proliferation and migration by downregulating IGF1R in hepatocellular carcinoma. Oncotarget. 2017;8:86592–86603. doi: 10.18632/oncotarget.21246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bai F., Jiu M., You Y., Feng Y., Xin R., Liu X., Mo L., Nie Y. miR-29a-3p represses proliferation and metastasis of gastric cancer cells via attenuating HAS3 levels. Mol. Med. Rep. 2018;17:8145–8152. doi: 10.3892/mmr.2018.8896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Jiang Y., Zhang M., Guo T., Yang C., Zhang C., Hao J. MicroRNA-21-5p promotes proliferation of gastric cancer cells through targeting SMAD7. OncoTargets Ther. 2018;11:4901–4911. doi: 10.2147/ott.S163771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhou J., Li X., Wu X., Zhang T., Zhu Q., Wang X., Wang H., Wang K., Lin Y., Wang X. Exosomes released from tumor-associated macrophages transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol. Res. 2018;6:1578–1592. doi: 10.1158/2326-6066.Cir-17-0479. [DOI] [PubMed] [Google Scholar]
  • 99.Yin C., Han Q., Xu D., Zheng B., Zhao X., Zhang J. SALL4-mediated upregulation of exosomal miR-146a-5p drives T-cell exhaustion by M2 tumor-associated macrophages in HCC. OncoImmunology. 2019;8:e1601479. doi: 10.1080/2162402X.2019.1601479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Gurien S.D., Aziz M., Jin H., Wang H., He M., Al-Abed Y., Nicastro J.M., Coppa G.F., Wang P. Extracellular microRNA 130b-3p inhibits eCIRP-induced inflammation. EMBO Rep. 2020;21:e48075. doi: 10.15252/embr.201948075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Essandoh K., Li Y., Huo J., Fan G.C. MiRNA-mediated macrophage polarization and its potential role in the regulation of inflammatory response. Shock. 2016;46:122–131. doi: 10.1097/shk.0000000000000604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mu J., Wang H., Wang X., Sun P. Expression of miR-124 in gastric adenocarcinoma and the effect on proliferation and invasion of gastric adenocarcinoma SCG-7901 cells. Oncol. Lett. 2019;17:3406–3410. doi: 10.3892/ol.2019.9981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li R., Hu Z., Wang Z., Zhu T., Wang G., Gao B., Wang J., Deng X. miR-125a-5p promotes gastric cancer growth and invasion by regulating the Hippo pathway. J. Clin. Lab. Anal. 2021;35:e24078. doi: 10.1002/jcla.24078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Uddin M.H., Al-Hallak M.N., Philip P.A., Mohammad R.M., Viola N., Wagner K.U., Azmi A.S. Exosomal microRNA in pancreatic cancer diagnosis, prognosis, and treatment: from bench to bedside. Cancers. 2021;13:2777. doi: 10.3390/cancers13112777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Chen Q., Wang H., Liu Y., Song Y., Lai L., Han Q., Cao X., Wang Q. 2012. Inducible microRNA-223 Down-Regulation Promotes TLR-Triggered IL-6 and IL-1β Production in Macrophages by Targeting STAT3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yang F., Wang T., Du P., Fan H., Dong X., Guo H. M2 bone marrow-derived macrophage-derived exosomes shuffle microRNA-21 to accelerate immune escape of glioma by modulating PEG3. Cancer Cell Int. 2020;20:93. doi: 10.1186/s12935-020-1163-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wan X., Xie B., Sun H., Gu W., Wang C., Deng Q., Zhou S. Exosomes derived from M2 type tumor-associated macrophages promote osimertinib resistance in non-small cell lung cancer through MSTRG.292666.16-miR-6836-5p-MAPK8IP3 axis. Cancer Cell Int. 2022;22:83. doi: 10.1186/s12935-022-02509-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jin K.-T., Lu Z.-B., Lv J.-Q., Zhang J.-G. The role of long non-coding RNAs in mediating chemoresistance by modulating autophagy in cancer. RNA Biol. 2020;17:1727–1740. doi: 10.1080/15476286.2020.1737787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Yeon M., Lee S., Lee J.-E., Jung H.S., Kim Y., Jeoung D. CAGE-miR-140-5p-Wnt1 Axis regulates autophagic flux, tumorigenic potential of mouse colon cancer cells and cellular interactions mediated by exosomes. Front. Oncol. 2019;9:1240. doi: 10.3389/fonc.2019.01240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wang Z., Chen K., Li D., Chen M., Li A., Wang J. miR-140-3p is involved in the occurrence and metastasis of gastric cancer by regulating the stability of FAM83B. Cancer Cell Int. 2021;21:537. doi: 10.1186/s12935-021-02245-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhao M., Liu Q., Liu W., Zhou H., Zang X., Lu J. MicroRNA-140 suppresses Helicobacter pylori-positive gastric cancer growth by enhancing the antitumor immune response. Mol. Med. Rep. 2019;20:2484–2492. doi: 10.3892/mmr.2019.10475. [DOI] [PubMed] [Google Scholar]
  • 112.Yang R., Wang D., Han S., Gu Y., Li Z., Deng L., Yin A., Gao Y., Li X., Yu Y., Wang X. MiR-206 suppresses the deterioration of intrahepatic cholangiocarcinoma and promotes sensitivity to chemotherapy by inhibiting interactions with stromal CAFs. Int. J. Biol. Sci. 2022;18:43–64. doi: 10.7150/ijbs.62602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Xie M., Dart D.A., Owen S., Wen X., Ji J., Jiang W. Insights into roles of the miR-1, -133 and -206 family in gastric cancer (Review) Oncol. Rep. 2016;36:1191–1198. doi: 10.3892/or.2016.4908. [DOI] [PubMed] [Google Scholar]
  • 114.Liu N., Wang X., Steer C.J., Song G. MicroRNA-206 promotes the recruitment of CD8+ T cells by driving M1 polarisation of Kupffer cells. Gut. 2022;71:1642–1655. doi: 10.1136/gutjnl-2021-324170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ye S.B., Zhang H., Cai T.T., Liu Y.N., Ni J.J., He J., Peng J.Y., Chen Q.Y., Mo H.Y., Jun C., et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J. Pathol. 2016;240:329–340. doi: 10.1002/path.4781. [DOI] [PubMed] [Google Scholar]
  • 116.Wang J., Huang F., Shi Y., Zhang Q., Xu S., Yao Y., Jiang R. RP11-323N12.5 promotes the malignancy and immunosuppression of human gastric cancer by increasing YAP1 transcription. Gastric Cancer. 2021;24:85–102. doi: 10.1007/s10120-020-01099-9. [DOI] [PubMed] [Google Scholar]
  • 117.Lou Q., Liu R., Yang X., Li W., Huang L., Wei L., Tan H., Xiang N., Chan K., Chen J., Liu H. miR-448 targets IDO1 and regulates CD8+ T cell response in human colon cancer. J. Immunother. Cancer. 2019;7:210. doi: 10.1186/s40425-019-0691-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Xie M., Yu T., Jing X., Ma L., Fan Y., Yang F., Ma P., Jiang H., Wu X., Shu Y., Xu T. Exosomal circSHKBP1 promotes gastric cancer progression via regulating the miR-582-3p/HUR/VEGF axis and suppressing HSP90 degradation. Mol. Cancer. 2020;19:112. doi: 10.1186/s12943-020-01208-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Piao H.Y., Guo S., Wang Y., Zhang J. Exosome-transmitted lncRNA PCGEM1 promotes invasive and metastasis in gastric cancer by maintaining the stability of SNAI1. Clin. Transl. Oncol. 2021;23:246–256. doi: 10.1007/s12094-020-02412-9. [DOI] [PubMed] [Google Scholar]
  • 120.Qiu S., Xie L., Lu C., Gu C., Xia Y., Lv J., Xuan Z., Fang L., Yang J., Zhang L., et al. Gastric cancer-derived exosomal miR-519a-3p promotes liver metastasis by inducing intrahepatic M2-like macrophage-mediated angiogenesis. J. Exp. Clin. Cancer Res. 2022;41:296. doi: 10.1186/s13046-022-02499-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Huang X.Y., Huang Z.L., Zhang P.B., Huang X.Y., Huang J., Wang H.C., Xu B., Zhou J., Tang Z.Y. CircRNA-100338 is associated with mTOR signaling pathway and poor prognosis in hepatocellular carcinoma. Front. Oncol. 2019;9:392. doi: 10.3389/fonc.2019.00392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Liu H., Liu Y., Sun P., Leng K., Xu Y., Mei L., Han P., Zhang B., Yao K., Li C., et al. Colorectal cancer-derived exosomal miR-106b-3p promotes metastasis by down-regulating DLC-1 expression. Clin. Sci. 2020;134:419–434. doi: 10.1042/cs20191087. [DOI] [PubMed] [Google Scholar]
  • 123.Qu M., Li J., Hong Z., Jia F., He Y., Yuan L. The role of human umbilical cord mesenchymal stem cells-derived exosomal microRNA-431-5p in survival and prognosis of colorectal cancer patients. Mutagenesis. 2022;37:164–171. doi: 10.1093/mutage/geac007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Liang Z.X., Liu H.S., Wang F.W., Xiong L., Zhou C., Hu T., He X.W., Wu X.J., Xie D., Wu X.R., Lan P. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death Dis. 2019;10:829. doi: 10.1038/s41419-019-2077-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Chen K., Wang Q., Liu X., Wang F., Yang Y., Tian X. Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. Int. J. Biol. Sci. 2022;18:1220–1237. doi: 10.7150/ijbs.67675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.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;37:324. doi: 10.1186/s13046-018-0965-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Pang W., Yao W., Dai X., Zhang A., Hou L., Wang L., Wang Y., Huang X., Meng X., Li L. Pancreatic cancer-derived exosomal microRNA-19a induces β-cell dysfunction by targeting ADCY1 and EPAC2. Int. J. Biol. Sci. 2021;17:3622–3633. doi: 10.7150/ijbs.56271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Zhao M., Zhuang A., Fang Y. Cancer-associated fibroblast-derived exosomal miRNA-320a promotes macrophage M2 polarization in vitro by regulating PTEN/PI3Kγ signaling in pancreatic cancer. J. Oncol. 2022;2022:9514697. doi: 10.1155/2022/9514697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Shi L., Wang Z., Geng X., Zhang Y., Xue Z. Exosomal miRNA-34 from cancer-associated fibroblasts inhibits growth and invasion of gastric cancer cells in vitro and in vivo. Aging (Albany NY) 2020;12:8549–8564. doi: 10.18632/aging.103157. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 130.Xu G., Zhang B., Ye J., Cao S., Shi J., Zhao Y., Wang Y., Sang J., Yao Y., Guan W., et al. Exosomal miRNA-139 in cancer-associated fibroblasts inhibits gastric cancer progression by repressing MMP11 expression. Int. J. Biol. Sci. 2019;15:2320–2329. doi: 10.7150/ijbs.33750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Lin H., Zhang R., Wu W., Lei L. miR-4454 promotes hepatic carcinoma progression by targeting Vps4A and Rab27A. Oxid. Med. Cell. Longev. 2021;2021:9230435. doi: 10.1155/2021/9230435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lin H., Peng J., Zhu T., Xiong M., Zhang R., Lei L. Exosomal miR-4800-3p aggravates the progression of hepatocellular carcinoma via regulating the hippo signaling pathway by targeting STK25. Front. Oncol. 2022;12:759864. doi: 10.3389/fonc.2022.759864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Yu S., Zhou L., Fu J., Xu L., Liu B., Zhao Y., Wang J., Yan X., Su J. H-TEX-mediated signaling between hepatocellular carcinoma cells and macrophages and exosome-targeted therapy for hepatocellular carcinoma. Front. Immunol. 2022;13:997726. doi: 10.3389/fimmu.2022.997726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Zhang P.F., Gao C., Huang X.Y., Lu J.C., Guo X.J., Shi G.M., Cai J.B., Ke A.W. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol. Cancer. 2020;19:110. doi: 10.1186/s12943-020-01222-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Yuan P., Song J., Wang F., Chen B. Exosome-transmitted circ_002136 promotes hepatocellular carcinoma progression by miR-19a-3p/RAB1A pathway. BMC Cancer. 2022;22:1284. doi: 10.1186/s12885-022-10367-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Chen J., Wang S., Jia S., Ding G., Jiang G., Cao L. Integrated analysis of long non-coding RNA and mRNA expression profile in pancreatic cancer derived exosomes treated dendritic cells by microarray analysis. J. Cancer. 2018;9:21–31. doi: 10.7150/jca.21749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ren W., Zhang X., Li W., Feng Q., Feng H., Tong Y., Rong H., Wang W., Zhang D., Zhang Z., et al. Exosomal miRNA-107 induces myeloid-derived suppressor cell expansion in gastric cancer. Cancer Manag. Res. 2019;11:4023–4040. doi: 10.2147/cmar.S198886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Zhu J., Du S., Zhang J., Huang G., Dong L., Ren E., Liu D. microRNA-10a-5p from gastric cancer cell-derived exosomes enhances viability and migration of human umbilical vein endothelial cells by targeting zinc finger MYND-type containing 11. Bioengineered. 2022;13:496–507. doi: 10.1080/21655979.2021.2009962. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]

Articles from iScience are provided here courtesy of Elsevier

RESOURCES