Open Access

Role of microRNAs in remodeling the tumor microenvironment (Review)

  • Authors:
    • Zhaoji Pan
    • Yiqing Tian
    • Guoping Niu
    • Chengsong Cao
  • View Affiliations

  • Published online on: December 24, 2019     https://doi.org/10.3892/ijo.2019.4952
  • Pages: 407-416
  • Copyright: © Pan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are short non‑coding RNAs that are known to regulate gene expression at the post‑transcriptional level. miRNA expression is often deregulated in several human cancers, affecting the communication between tumor stroma and tumor cells, among other functions. Understanding the role of miRNAs in the tumor microenvironment is crucial for fully elucidating the molecular mechanisms underlying tumor progression and exploring novel diagnostic biomarkers and therapeutic targets. The present review focused on the role of miRNAs in remodeling the tumor microenvironment, with an emphasis on their impact on tumor growth, metastasis and resistance to treatment, as well as their potential clinical applications.

1. Introduction

Over the past decade, several studies have demonstrated that cancer initiation and progression are determined not only by cancer cells, but also by the tumor microenvironment (TME) (1-4), which includes fibroblasts, immune cells and endothelial cells, among others (5). Mounting evidence suggests that microRNAs (miRNAs) play a key role in shaping the biology and function of tumor stromal cells (5-8). The deregulation of miRNAs has been associated with almost every aspect of cancer initiation and progression (9-11). In 2006, Volinia et al first identified a miRNA signature in human cancers and demonstrated that the predicted targets of these deregulated miRNAs were classic oncogenes or tumor suppressors (12). One of the oncogenic miRNAs identified was miR-21, which was found to be highly expressed in breast and colon cancer, and its overexpression was correlated with poor patient survival. Furthermore, miR-21 was demonstrated to target programmed cell death 4, an apoptosis-inducing protein, thus promoting tumor growth (13,14). Additionally, miR-34a was identified as a downstream target of the p53 tumor suppressor gene (15). With the recent advances in miRNA detection techniques, cancer cell-derived miRNAs have emerged as promising diagnostic biomarkers and therapeutic targets, as has been described in detail elsewhere (16-18).

Recent studies have highlighted the significance of miRNAs in the TME (19-24). Aprelikova et al identified 11 miRNAs that were differentially expressed between cancer-associated fibroblasts (CAFs) isolated from human endometrial cancer and normal endometrial fibroblasts (25). Mesenchymal stem cells (MSCs) were previously isolated from gastric cancer (GC) tissues (GC-MSCs) and paired adjacent non-cancerous gastric tissues (GCN-MSCs), and 114 upregulated and 85 downregulated miRNAs were identified in GC-MSCs (26). Liu et al compared the miRNA expression profiles of myeloid-derived suppressor cells (MDSCs) from breast cancer-bearing mice and their counterparts from tumor-free mice, and found that 3 miRNAs (miR-494, miR-882 and miR-361) were upregulated, whereas 5 miRNAs (miR-466, let-7e, miR-133b, miR-713 and miR-322) were downregulated in the MDSCs from the tumor-bearing mice. Furthermore, 5 miRNAs were found to be upregulated and 7 miRNAs downregulated in cancer-associated endothelial cells compared with normal endothelial cells (27). The aim of the present review was to summarize the latest advances in understanding the roles of miRNAs in TME remodeling, which ultimately affects tumor progression and may be of value in the clinical setting.

2. Role of miRNAs in regulating TME cells

miRNAs in regulating CAFs

Fibroblasts are one of the major components of the TME. At the primary tumor site, fibroblasts acquire distinct phenotypic characteristics and become CAFs through a miRNA-mediated regulation of multiple signaling pathways. CAFs differ from normal fibroblasts (NFs) in their high expression of α-smooth muscle actin and their pro-tumorigenic properties (28). CAFs secrete a wide range of pro-inflammatory molecules, including interleukins, chemo-kines and extracellular matrix (ECM) components, ultimately promoting tumor growth by modulating tumor-associated inflammation or directing cell-to-cell communication (29). Broniszet al reported that miR-320 expression was significantly reduced when phosphatase and tensin homolog (PTEN) on chromosome 10 was ablated, which induced the transition of NFs to CAFs in breast cancer (30). Loss of the PTEN gene in stromal fibroblasts results in the activation of an oncogenic secretome. The downregulation of miR-320 and upregulation of one of its direct targets, ETS proto-oncogene 2, transcription factor, are critical events in PTEN-deficient stromal fibroblasts that induce the oncogenic secretome, which in turn promotes tumor angiogenesis and tumor cell invasion (30).

In breast cancer, the pro-metastatic miRNA miR-9 was shown to induce a switch in human breast fibroblasts from a normal towards a CAF phenotype, thereby contributing to tumor progression (31). In addition, the downregulation of miR-200s was reported to play an important role in reprogramming NFs into CAFs. miR-200s target friend leukemia virus integration 1 (Fli-1) and transcription factor 12 (TCF12) in the breast cancer microenvironment (32,33). Moreover, decreased miR-205 was shown to convert breast NFs into CAFs by promoting Yes-associated protein 1 (YAP1) expression, which has been proven to be involved in angiogenesis (34).

In ovarian cancer, low expression of miR-214 and high expression of miR-155 are involved in reprogramming quiescent fibroblasts to CAFs. miR-214 directly targets C-C motif ligand 5 (CCL5), which is essential for CAF function. The over-expression of miR-155 in NFs induces fibroblasts to develop CAF-like phenotypes. The disruption of these miRNAs is sufficient to reverse the functions of CAFs, thereby reducing ovarian cancer growth and metastasis (32). Furthermore, miR-155 was shown to convert NFs into CAFs in pancreatic cancer by targeting p53-inducible nuclear protein 1 (35).

In melanoma, melanocytes are specialized in producing pigment vesicles, referred to as melanosomes. Melanosomes have been shown to carry miRNAs, including miR-211, into primary fibroblasts, triggering changes such as increased proliferation, migration and pro-inflammatory gene expression, all of which are known characteristics of CAFs (36).

In lung cancer, downregulation of miR-1 and miR-206, and upregulation of miR-31, may promote the NF-CAF switch by stimulating CCL2/vascular endothelial growth factor A (VEGFA) expression (37). Taken together, these findings underline the importance of miRNAs in driving the conversion of NFs into CAFs in several tumor entities.

Role of miRNAs in regulating tumor-associated MSCs

MSCs are progenitor cells that are known to participate in tumor stroma formation and tumor progression. Bone marrow-derived MSCs (BM-MSCs) can migrate to tumor sites, where they exert tumor-promoting and pro-metastatic effects. We previously demonstrated that miR-155-5p downregulation induced BM-MSCs to acquire the phenotype of GC-MSCs through the activation of nuclear factor-κB p65 (NF-κB p65). In that study, NF-κB p65 and the inhibitor of NF-κB kinase subunit ε were identified as the targets of miR-155-5p (38). miR-214, miR-221 and miR-222 were upregulated in GC-MSCs compared with GCN-MSCs. GC-MSCs significantly promoted the proliferation and migration of GC cells. Targeted inhibition of miR-221 in GC-MSCs was found to inhibit its tumor-promoting effects (26).

Let-7 is downregulated in prostate cancer-associated MSCs, which exhibit a stronger propensity for migration and invasion compared with normal MSCs. Interleukin (IL)-6 was identified as a direct target gene of let-7. The overexpression of let-7 reduces IL-6 expression and represses the metastasis-promoting activity of cancer-associated MSCs (39). miR-146a overexpression leads to increased secretion of chemo-tactic proteins, including CXCL1, interferon gamma-induced protein 10, CCL5 and IL-6, which are crucial for the growth and migration of multiple myeloma cells. The Notch pathway was also shown to be involved in the miR-146a-induced cytokine and chemokine secretion in MSCs (40). In myeloid neoplasms, miR-7977 in MSCs was found to induce aberrant reduction of hematopoietic growth factors, including Jagged-1, stem cell factor and angiopoietin-1, thereby reducing the hematopoietic-supporting capacity of bone marrow CD34+ cells (41). This evidence indicates that the aberrant expression of miRNAs can promote the evolution of MSCs in the TME, facilitating tumor progression.

Role of miRNAs in regulating tumor-associated macrophages

Tumor progression is intrinsically linked to immune evasion. The activation of certain immune cells drives potent antitumor responses, whereas the suppression of these cells promotes tumor progression and metastasis. Cancer cells can regulate miRNA expression in infiltrating immune cells, thereby suppressing the local antitumor immune response and ultimately leading to tumor progression (42-47). Tumor-associated macrophages (TAMs) have both pro- and anti-tumorigenic properties, depending on whether they belong to the classical M1 or alternative M2 subtype. At the late stage of human cancers, most of the infiltrating macrophages display an M2 phenotype, creating an immunosuppressive microenvironment that favors tumor progression. Increased miR-21-5p delivery by MSC-derived extracellular vesicles was recently reported to promote the polarization of macrophages towards an M2 phenotype (48). miR-125b binds to the 3’ untranslated region of tumor necrosis factor-α (TNF-α), inhibiting its production and sustaining an M1 phenotype (49). By contrast, miR-146 directly inhibits the expression of the adaptor protein tumor necrosis factor receptor-associated factor 6 and IL-1 receptor-associated kinase-1 in the NF-κB pathway, thus reducing pro-inflammatory cytokine production and promoting alternative M2 activation (5).

In addition to regulating the polarization and phenotype of TAMs, miRNAs have also been shown to regulate other functionalities of TAMs, including infiltration, immune responses and tumor-promoting effects. Ke et al reported that low miR-148b levels in hepatocellular carcinoma (HCC) cells enhanced the expression of colony-stimulating factor-1, promoting TAM infiltration and HCC metastasis (50). Moreover, another crucial miRNA involved in the modulation of immune responses is miR-155, which is persistently downregulated in TAMs (51,52). Soluble factors in HCC can downregulate miR-155 in TAMs. When the expression of miR-155 is restored in macrophages, both direct and indirect antitumor responses are promoted through the enhancement of T-cell function (51). Using a transgenic mouse model, Zhao et al demonstrated that miR-125a, a downstream molecule of Notch signaling pathway, could reprogram macrophages in the TME and restore their anti-tumor potential by targeting certain factors, including factor inhibiting hypoxia-inducible factor-1, interferon regulatory factor 4, and RING1- and YY1-binding protein (53). A recent report indicated that miR-511-3p exerted regulatory effects on TAMs, and that miR-511-3p overexpression in TAMs inhibited tumor growth in vivo, suggesting that miR-511-3p limits the tumor-promoting function of TAMs (54). miR-21 and miR-29a are delivered from cancer cells to macrophages via microvesicles and subsequently bind to intracellular toll-like receptors (TLR7 or TLR8). This miRNA-TLR interaction results in the activation of the NF-κB pathway and increased production of the pro-inflammatory cytokines IL-6 and TNF-α in TAMs, which in turn promotes cancer metastasis (55). Unexpectedly, apoptotic breast cancer cell-derived miR-375, which is released as a non-exosome entity, is taken up by macrophages via the CD36 receptor and enhances the migration and infiltration of macrophages towards tumor spheroids by targeting tensin 3 and paxillin (56). Therefore, the miRNAs of TME play a key role in regulating the phenotype and function of TAMs through several pathways.

Role of miRNAs in regulating T cells

miRNAs play a key role in T-cell maturation, activation and function. The immuno-suppressive FOXP3+ regulatory T cells (Tregs) are enriched in tumor tissues and are associated with cancer development and progression (57,58). Exosomal miR-24-3p inhibits T-cell proliferation and Th1 and Th17 differentiation, and promotes Treg development through the repression of fibroblast growth factor 11 (59). Tumor-secreted miR-214 can induce Tregs to secrete higher levels of IL-10 by downregulating PTEN, leading to immune suppression and rapid tumor growth. The inhibition of cancer cell-secreted miR-214 has been shown to block Treg induction and tumor growth, suggesting that anti-miR-214 therapy can abolish tumor-induced Treg expansion and suppress tumor growth (58). The silencing of miR-21 has been reported to significantly reduce the proliferation of chemokine receptor 6-positive Tregs by targeting PTEN and the subsequently activated Akt pathway, which is crucial for the induction and functional sustainability of CD4+ FOXP3+ Tregs (60). miR-126 silencing was also shown to impair the expression of FOXP3 and inhibit the expression of functional molecules in Tregs.

Moreover, in a murine mammary cancer model, the silencing of miR-126 in Tregs resulted in enhanced antitumor activity of CD8+ T cells (61). Additionally, miR-149-3p was found to enhance T-cell proliferation and secretion of cytokines, indicative of increased T-cell activation, which may reverse CD8+ T-cell exhaustion and promote CD8+ T-cell-mediated killing of 4T1 mouse breast tumor cells (62). Notably, in human colon cancer, miR-448 could also suppress CD8+ T-cell apoptosis and enhance CD8+ T-cell response by inhibiting indoleamine 2,3-dioxygenase 1 enzyme function (63). These results suggest that miRNAs are key regulators of the functions of T cells in the TME.

Role of miRNAs in regulating endothelial cells

Angiogenesis is crucial for tumor progression and metastasis. Mangala et al isolated endothelial cells from high-grade serous ovarian cancer and normal ovarian tissues and identified deregulated miRNAs in cancer-associated endothelial cells (64); they demonstrated that one of the deregulated miRNAs, miR-29a/c, was a regulator of VEGF. The downregulation of miR-29a/c was shown to promote GC progression by increasing VEGF expression (65). In breast cancer, tumor cells can release miR-939 to endothelial cells via exosomes, and miR-939 was shown to suppress the expression of its target gene vascular endothelial-cadherin in endothelial cells, increasing vascular permeability (66). Moreover, miR-205 suppression in other cells of the TME, including CAFs, was reported to enhance tubule formation and sprouting of endothelial cells by regulating YAP1-mediated IL-11 and IL-15 expression and secretion (34). In prostate cancer, miR-218 downregulation was shown to promote tumor angiogenesis through the regulatory-associated protein-independent companion of mammalian target of rapamycin complex 2/VEGFA axis (67). In lung cancer, miR-143/145 markedly promotes tumor growth by stimulating the proliferation of endothelial cells (68). In glioma, the downregulation of miR-125b may inhibit the expression of Myc-associated zinc finger protein, thereby attenuating primary human brain endothelial cell migration and tubule formation in vitro (69). The upregulation of miR-93 was reported to enhance tumor growth via integrin-β8-mediated angiogenesis (70), as well as to enhance endothelial tube formation and increase blood vessel densityin vivo (71). The downregulation of miR-29b was also shown to promote VEGFR signaling in endothelial cells, enhancing angiogenesis (72). Another miRNA involved in angiogenesis is miR-638, which was shown to suppress HCC growth by inhibiting VEGF signaling (73). These findings demonstrate that miRNAs play diverse roles in regulating endothelial cell activity and tumor angiogenesis.

3. Role of TME-derived miRNAs in regulating tumor progression

TME-associated miRNAs in regulating tumor growth

Tumor growth is highly dependent on the communication between tumor cells and cells of the TME. During tumor progression, miRNAs participate in the intricate interactions between tumor cells and tumor stromal cells, such as CAFs, endothelial cells and infiltrating immune cells. For example, fibroblasts provide a stromal framework for tumor cells during early tumor growth (29,74). Recent studies have revealed that the upregulated miR-7 suppresses RAS-association domain family member 2 expression in CAFs, which in turn enhances the proliferation of head and neck cancer cells (75). Wang et al also observed that loss of CAF-derived exosomal miR-3188 may affect the proliferation and apoptosis of tumor cells (76). Melanoma cell-derived miR-211 was shown to induce CAF formation by directly targeting insulin-like growth factor 2 and activating mitogen-activated protein kinase signaling, which in turn enhances the growth of melanoma (36). TAMs play pivotal roles in tumor growth (77-79). Yin et al reported that M2 macrophage-derived exosomal miR-501-3p facilitated tumor formation in vivo via regulating the transforming growth factor (TGF)-β signaling pathway (80). Macrophages transfected with a miR-125a mimic exhibited increased phagocytic activity and repressed lung cancer growth in mice (53).

Additionally, the increased levels of miR-27a inhibited dendritic cell-mediated differentiation of Th1 and Th17 cells and enhanced the growth of melanoma cells (81). The downregulation of miR-638 was demonstrated to promote angiogenesis and HCC growth by targeting VEGF, whereas its overexpression inhibited tumor angiogenesis (73). miR-143/145 in the lung cancer microenvironment markedly promoted tumor growth by targeting calcium/calmodulin-dependent protein kinase 1D, an inhibitory kinase, the overexpression of which prevents the mitotic entry of endothelial cells (68). Thus, TME-associated miRNAs play key roles in tumor growth.

Role of TME-associated miRNAs in regulating tumor metastasis

Metastasis accounts for ~90% of cancer-related deaths (82,83). The metastatic ability of tumor cells is enhanced by their interactions with tumor stromal cells. miRNAs participate in several steps of the metastatic process. In a mouse xenograft model, miR-126/miR-126* independently inhibited the sequential recruitment of MSCs and inflammatory mono-cytes into the tumor stroma, thus suppressing lung metastasis through the downregulation of stromal cell-derived factor-1 α and CCL2 (84). Tumor-secreted miR-9 may be transferred via exosomes to the recipient NFs, and miR-9, in turn, is secreted by fibroblasts to stimulate breast cancer cell migration through the reduction of E-cadherin (31). Recently, low levels of miR-200s in CAFs were found to increase Fli-1 and TCF12 expression and promote stromal ECM remodeling to drive tumor metastasis through the upregulation of fibronectin and lysyl oxidase in breast cancer (33). Furthermore, low-level CAF-derived exosomal miR-148b may be transferred to endometrial cancer cells and enhance endometrial cancer metastasis by targeting DNA-methyltransferase 1, which is an important regulator of tumor metastasis (85). MSC-induced expression of miRNA-199a was shown to enhance the stem cell properties of breast cancer cells, thereby promoting tumor initiation and metastasis (86). The aberrantly expressed miRNAs in TAMs also play pivotal roles in tumor metastasis. miR-28-5p deficiency was shown to upregulate the expression of IL-34 to recruit TAMs, consequently promoting tumor metastasis (87). M2 macrophage-derived exosomal miR-501-3p was found to facilitate the liver and lung metastasis of pancreatic ductal adenocarcinoma (PDAC) cells (80). In summary, miRNAs in the TME are well-established crucial mediators of tumor metastasis.

Role of TME-associated miRNAs in regulating drug resistance

Resistance to chemotherapy and targeted therapy is a major challenge in clinical practice. Accumulating evidence suggests that miRNAs from the TME may affect drug resistance. In HCC, exosomal miR-122 (122-Exo) from adipose tissue-derived MSCs can be delivered into HCC cells, increasing the chemosensitivity of HCC cells by inhibiting the expression of cyclin G1, a disintegrin and metallopro-tease 10, and insulin-like growth factor receptor 1 (IGF1R). Intratumoral injection of 122-Exo significantly increased the antitumor efficacy of sorafenib in HCC in vivo (88). Moreover, the chemotherapeutic agent sorafenib can inhibit miR-101 expression and enhance dual-specificity phosphatase 1 expression, leading to the inhibition of macrophage-induced HCC growth (89). In GC, miR-21 was transferred from macrophages to GC cells via exosomes and resulted in a significant reduction in the sensitivity of GC cells to cisplatin chemotherapy in vitro and in vivo, partially through the regulation of the PTEN/phosphoinositide 3-kinase/AKT signaling pathway (90). The levels of miR-27a/b in the serum were significantly higher in patients with esophageal cancer compared with those in healthy volunteers, and high expression levels of miR-27a/b were shown to induce the transformation of NFs into CAFs through upregulation of TGF-β, leading to chemoresistance in esophageal cancer cells (91). These results suggest that TME-derived miRNAs play a critical role in drug resistance.

4. Clinical applications of tumor-associated miRNAs

miRNAs as cancer diagnostic biomarkers

miRNAs have exhibited great potential in cancer diagnosis and prognosis. Circulating miRNAs in body fluids, such as plasma, serum and urine, have high diagnostic potential as cancer biomarkers due to their combination with argonaute protein or high-density lipoprotein, which protects them from degradation by extreme pH, high temperature and RNase. The vast majority of studies conducted thus far have focused on miRNAs in the tumor cells or the TME. Bryant et al reported that miR-107 and miR-574-3p were present at higher concentrations in the urine of men with prostate cancer compared with healthy individuals, indicating their potential as non-invasive biomarkers (92). Leeet al reported that circulating miR-146b and miR-155 levels were significantly higher in patients with papillary thyroid carcinoma compared with those with benign tumors. The levels of miR-146b and miR-155 were also positively associated with tumor size (93).

Moreover, Yi et al identified circulating miR-31-5p as a potentially novel and non-invasive biomarker for the early diagnosis of nasopharyngeal carcinoma (94). Nobuyoshi et al observed higher miR-92 levels in patients with colorectal cancer compared with those patients with inflammatory bowel diseases or in healthy volunteers; hence, it may be used as a biomarker to detect colorectal cancer (95). Few studies have explored the diagnostic potential of TME-derived miRNAs. In acute myeloid leukemia (AML), bone marrow stromal cell-derived miRNAs, including miR-150, -155 and -1246, were present in serum exosomes and were able to improve the sensitivity and specificity for detecting residual or recurrent AML, thereby providing a reliable cell-free marker that is unaffected by chemotherapy (96). We also previously demonstrated that miR-221 was highly expressed in MSCs from human GC tissues. The expression of MSC-derived miR-221 was positively associated with the proliferation and migration of GC cells, suggesting that it may be a new potential biomarker for GC diagnosis (26). In breast cancer, the expression of miR-21 in CCR6-expressing Tregs is very high, whereas silencing of miR-21 enhances the expression of PTEN, which inhibits the proliferation of Tregs in the tumor tissues and endows CD8+ T cells with effective antitumor functions, indicating a new biomarker for breast cancer prognosis (60). In PDAC, the strong expression of miR-21 in CAFs was found to be associated with decreased overall survival in PDAC patients receiving 5-fluoro-uracil, but not gemcitabine. Thus, miR-21 may serve as a marker to guide chemotherapy options in PDAC patients (97). However, the number of TME-associated miRNAs identified as cancer biomarkers is currently limited and further research is required.

miRNAs in cancer therapy

According to the literature, miRNA-based antitumor therapy can be broadly divided into two distinct approaches, namely miRNA silencing by anti-miRNAs and miRNA restoration. Cubillos-Ruiz et al reported that nanoparticles carrying oligonucleotide duplexes markedly augmented miR-155 activity and transformed tumor-associated dendritic cells from the immunosuppressive phenotype to highly immunostimulatory cells, eliciting potent antitumor responses that eliminate ovarian cancer cells (98). In a mouse breast cancer model, let-7b delivered through a nucleic acid delivery system expeditiously reprogrammed the functions of TAMs and tumor-infiltrating dendritic cells, leading to the inhibition of tumor growth. This strategy may represent a new approach to cancer immunotherapy (99). Additionally, miR-21 depletion in TAMs promoted tumori-cidal M1 polarization by upregulating Janus kinase 2 and Signal transducer and activator of transcription 1, and this, combined with programmed cell death protein 1 blockade, may exert synergistic effects and exhibit superior antitumor activity (100).

Moreover, the targeted interference of deregulated miRNAs in cancer-associated endothelial cells was shown to decrease vascular permeability. Pi et al reported that the elevated expression of miR-302/367 significantly suppressed tumor growth by restricting sprout angiogenesis and decreasing vascular permeability (101). Schnittert et al reported that anti-miR-199a, delivered via peptide-based nanocomplexes, may inhibit human-derived pancreatic stellate cell (hPSC) differentiation into CAFs and repress the size of 3D hetero-spheroids, which consist of hPSCs and tumor cells (102). In summary, targeting miRNAs has emerged as a novel, promising approach to cancer treatment.

5. Conclusion

The present review summarized the roles of miRNAs in the communication between tumor cells and tumor stromal cells. Deregulated expression of several miRNAs has been observed in both types of cells, clearly highlighting the crucial roles of miRNAs in cancer development and progression (Fig. 1; Table I). Numerous examples in which miRNAs were demonstrated to regulate the critical aspects of TME involved in tumor angiogenesis, tumor cell growth and metastasis were herein summarized. Previous studies elucidated some of the mechanisms by which these small RNAs in the TME considerably affect tumor biology. Undoubtedly, miRNA-related research has great potential for the identification of important biomarkers and for promoting the development of novel cancer therapies. As it has been reported in previous studies, miRNA delivery to tumors is an attractive yet challenging opportunity to improve therapeutic strategies for cancer (103,104). However, more cancer-related pathways must be identified as specific targets of miRNAs, and additional efforts are required to ultimately design an optimal delivery system for miRNAs. Several novel siRNA and miRNA delivery systems are currently under development. However, despite the enormous potential of microRNAs in cancer diagnosis and anticancer clinical practice, microRNAs are not yet widely applied as cancer biomarkers and therapy targets in the clinical setting, as numerous obstacles must be overcome before their widespread application.

Table I

MicroRNAs in tumor microenvironment remodeling.

Table I

MicroRNAs in tumor microenvironment remodeling.

MicroRNAsCancersFunction(Refs.)
Regulation of CAFs
 miR-9Breast cancerEnhancing the switch to CAF phenotype(29)
 miR-200sBreast cancerInhibiting CAF activation by targeting Fli-1 and TCF12(31)
 miR-205Breast cancerInhibiting the switch to CAFs by regulating YAP1(34)
 miR-155Ovarian cancer Pancreatic cancerPromoting conversion of the fibroblasts to a CAF-like phenotype by targeting TP53INP1(30,32)
 miR-214Ovarian cancerInhibiting CAF activation by targeting CCL5(30)
 miR-211MelanomaEnhancing the switch to CAF phenotype by targeting IGF2R and leading to MAPK signaling activation(33)
 miR-1, miR-206, miR-31Lung cancerReprograming NF-CAF conversion via affecting CCL2/VEGFA expression(34)
Regulation of cancer-associated
MSCs
 miR-155-5pGastric cancerInhibiting the conversion of BM-MSCs to GC-MSCs by targeting NF-κB p65(35)
 miR-221Gastric cancerPlaying a tumor-supporting role(24)
 Let-7Prostate cancerInhibiting the adipogenic differentiation and metastasis-promoting activity of cancer-associated MSCs by targeting IL-6(36)
 miR-146aMyelomaInducing cytokine and chemokine secretion in MSCs(37)
 miR-7977Myeloid neoplasmsDecreasing hematopoiesis-supporting capacity of bone marrow CD34+ cells.(38)
Regulation of TAM
 miR-125bLymphomaEnhancing macrophage responsiveness to IFNγ and increasing surface expression of its cognate receptor(45)
 miR-155Gastric cancerSuppressing cytokine production in tumor-activated monocytes/macrophages by targeting C/EBPβ(46)
 miR-511-3pLung cancerInhibiting the tumor-promoting functions of TAMs(49)
 miR-21, miR-29aLung cancerInducing the secretion of the proinflammatory cytokines TNF-α and IL-6 by TLR8-mediated activation of NF-κB(50)
 miR-21-5pLung cancerPromoting the polarization of M2 macrophages(48)
 miR-148bHepatocellular carcinomaInhibiting TAM infiltration and HCC metastasis by downregulating the colony stimulating factor-1 expression(50)
Regulation of T cells
 miR-24-3pNasopharyngeal carcinomaInducing differentiation of Tregs by targeting FGF11(53)
 miR-214Lung cancerInducing the secretion of IL-10 from Tregs and modulating Treg induction(52)
 miR-21Breast cancerReducing the proliferation of Tregs by targeting the PTEN and Akt pathways(54)
 miR-149-3pBreast cancerEnhancing T-cell proliferation and activation(62)
 miR-448Colon cancerSuppressing the apoptosis of CD8+ T cells and enhancing CD8+ T-cell response(63)
Regulation of endothelial cells
 miR-29a/cGastric cancerSuppressing angiogenesis by targeting VEGF in the gastric tumor microenvironment.(57)
 miR-939Breast cancerIncreasing HUVEC monolayer permeability by targeting vascular endothelial-cadherin.(58)
 miR-143/145Lung cancerStimulating the proliferation of endothelial cells and regulating endothelial cell migration(60)
 miR-125bGlioblastomaMediating VEGF-induced angiogenesis by targeting Myc-associated zinc finger protein(61)
 miR-93Breast cancerEnhancing endothelial tube formation by increasing blood vessel density(62)
miR-638Hepatocellular carcinomaSuppressing angiogenesis by inhibiting VEGF signaling(65)
 miR-205Breast cancerRegulating YAP1-mediated IL-11 and IL-15 expression and secretion in CAFs, which enhance tubule formation(34)
Regulation of tumor growth
 miR-7Head and neck cancerCAF-mediated tumor-promoting effect by targeting RASSF2(67)
 miR-211MelanomaCAF-mediated tumor-promoting effect by targeting IGF2R(33)
 miR-125aLung cancerTAM-mediated antitumor activity by targeting FIH1 and IRF4(48)
 miR-638Hepatocellular carcinoma Angiogenesis-mediated antitumor activity by targeting VEGF(65)
 miR-143/145Lung cancerEndothelial cell-mediated tumor-promoting effect by targeting CAMK1D(60)
 miR-3188Head and neck cancerCAF-derived exosomal expression regulating tumor growth(76)
 miR-501-3pPancreatic ductalM2 macrophage-derived exosomal miR-501-3p adenocarcinoma facilitating tumor formation by regulating the TGF-β signaling pathway(80)
Regulation of tumor metastasis
 miR-126/miR-126*Breast cancerSuppressing lung metastasis by inhibiting the recruitment of MSCs and inflammatory monocytes(74)
 miR-9Breast cancerCAF-mediated tumor cell migration by reducing E-cadherin(29)
 miR-200sBreast cancerCAF-mediated tumor cell invasion and metastasis by targeting Fli-1 and TCF12.(31)
 miRNA-199aBreast cancerMSC-mediated tumor initiation and metastasis(75)
 miR-28-5pHepatocellular carcinomaTAM-mediated metastasis by miR-28-5p-IL-34 signaling(76)
 miR-148bCAF-derived exosomal expression enhances tumor metastasis by targeting DNMT1(85)
Regulation of drug resistance
 miR-122 (MSCs)Hepatocellular carcinomaIncreasing the chemosensitivity of HCC cells by negative regulation of CCNG1, ADAM10 and IGF1R(77)
 miR-101 (macrophage)Hepatocellular carcinomaInhibited by sorafenib and enhancing DUSP1, leading to the inhibition of macrophage-induced HCC growth(78)
 miR-21 (macrophage)Gastric cancerReducing the sensitivity of gastric cancer cells to cisplatin through the regulation of PTEN/PI3K/AKT(79)

Funding

The present study was supported by the Special Foundation for Young Scientists of Jiangsu Province (grant no. QNRC2016379), Xuzhou Central Hospital and Xinyi People’s Hospital.

Availability of data and materials

The datasets used and analyzed during the present study are available from the corresponding author on reasonable request.

Authors’ contributions

ZP and YT made substantial contributions to conception and design and wrote the manuscript. GN and CC critically revised the manuscript for important intellectual content. ZP, YT, GN and CC reviewed and edited the manuscript. All authors have read and approved the final version of the manuscript for publication.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Wenrong Xu and Xu Zhang (Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China) for their ideas and support.

References

1 

Naito Y, Yoshioka Y, Yamamoto Y and Ochiya T: How cancer cells dictate their microenvironment: Present roles of extracellular vesicles. Cell Mol Life Sci. 74:697–713. 2017. View Article : Google Scholar :

2 

Dachs GU and Chaplin DJ: Microenvironmental control of gene expression: Implications for tumor angiogenesis, progression, and metastasis. Semin Radiat Oncol. 8:208–216. 1998. View Article : Google Scholar : PubMed/NCBI

3 

Whiteside TL: Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin Immunol. 35:69–79. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Chou J, Shahi P and Werb Z: microRNA-mediated regulation of the tumor microenvironment. Cell Cycle. 12:3262–3271. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Li X, Wu Z, Fu X and Han W: A microRNA component of the neoplastic Microenvironment: microregulators with far-reaching impact. Biomed Res Int. 2013:7621832013.PubMed/NCBI

7 

Zhang Y, Yang P and Wang XF: Microenvironmental regulation of cancer metastasis by miRNAs. Trends Cell Biol. 24:153–160. 2014. View Article : Google Scholar

8 

Xu SJ, Hu HT, Li HL and Chang S: The role of miRNAs in immune cell development, immune cell activation, and tumor immunity: With a focus on macrophages and natural killer cells. Cells. 8:pii: E1140. 2019. View Article : Google Scholar

9 

Musumeci M, Coppola V, Addario A, Patrizii M, Maugeri-Saccà M, Memeo L, Colarossi C, Francescangeli F, Biffoni M, Collura D, et al: Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene. 30:4231–4242. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Iorio MV and Croce CM: MicroRNA dysregulation in cancer: Diagnostics, monitoring and therapeutics. A comprehensive review EMBO Mol Med. 9:8522017. View Article : Google Scholar

11 

Que KT, Zhou Y, You Y, Zhang Z, Zhao XP, Gong JP and Liu ZJ: MicroRNA-31-5p regulates chemosensitivity by preventing the nuclear location of PARP1 in hepatocellular carcinoma. J Exp Clin Cancer Res. 37:2682018. View Article : Google Scholar : PubMed/NCBI

12 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S and Allgayer H: MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 27:2128–2136. 2008. View Article : Google Scholar

14 

Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A and Lund AH: Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem. 283:1026–1033. 2008. View Article : Google Scholar

15 

He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Zender L, Magnus J, Ridzon D, et al: A microRNA component of the p53 tumour suppressor network. Nature. 447:1130–1134. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Corsini LR, Bronte G, Terrasi M, Amodeo V, Fanale D, Fiorentino E, Cicero G, Bazan V and Russo A: The role of microRNAs in cancer: Diagnostic and prognostic biomarkers and targets of therapies. Expert Opin Ther Targets. 16(Suppl 2): S103–S109. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Nana-Sinkam SP and Croce CM: Clinical applications for microRNAs in cancer. Clin Pharmacol Ther. 93:98–104. 2013. View Article : Google Scholar

18 

Pichler M and Calin GA: MicroRNAs in cancer: From developmental genes in worms to their clinical application in patients. Br J Cancer. 113:569–573. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Soon P and Kiaris H: MicroRNAs in the tumour microenvironment: Big role for small players. Endocr Relat Cancer. 20:R257–R267. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Rupaimoole R, Calin GA, Lopez-Berestein G and Sood AK: miRNA deregulation in cancer cells and the tumor microenvi-ronment. Cancer Discov. 6:235–246. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Kuninty PR, Schnittert J, Storm G and Prakash J: MicroRNA targeting to modulate tumor microenvironment. Front Oncol. 6:32016. View Article : Google Scholar : PubMed/NCBI

22 

Fanini F and Fabbri M: Cancer-derived exosomic microRNAs shape the immune system within the tumor microenvironment: State of the art. Semin Cell Dev Biol. 67:23–28. 2017. View Article : Google Scholar :

23 

Curtale G: MiRNAs at the crossroads between innate immunity and cancer: Focus on macrophages. Cells. 7:pii: E12. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Bandini E, Rossi T, Gallerani G and Fabbri F: Adipocytes and microRNAs crosstalk: A key tile in the mosaic of breast cancer microenvironment. Cancers (Basel). 11:pii: E1451. 2019. View Article : Google Scholar

25 

Aprelikova O, Yu X, Palla J, Wei BR, John S, Yi M, Stephens R, Simpson RM, Risinger JI, Jazaeri A and Niederhuber J: The role of miR-31 and its target gene SATB2 in cancer-associated fibroblasts. Cell Cycle. 9:4387–4398. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Wang M, Zhao C, Shi H, Zhang B, Zhang L, Zhang X, Wang S, Wu X, Yang T, Huang F, et al: Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: Novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 110:1199–1210. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Liu Y, Lai L, Chen Q, Song Y, Xu S, Ma F, Wang X, Wang J, Yu H, Cao X and Wang Q: MicroRNA-494 is required for the accumulation and functions of tumor-expanded myeloid-derived suppressor cells via targeting of PTEN. J Immunol. 188:5500–5510. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Erez N, Truitt M, Olson P, Arron ST and Hanahan D: Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 17:135–147. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Kalluri R and Zeisberg M: Fibroblasts in cancer. Nat Rev Cancer. 6:392–401. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Bronisz A, Godlewski J, Wallace JA, Merchant AS, Nowicki MO, Mathsyaraja H, Srinivasan R, Trimboli AJ, Martin CK, Li F, et al: Reprogramming of the tumour microenvironment by stromal PTEN-regulated miR-320. Nat Cell Biol. 14:159–167. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Baroni S, Romero-Cordoba S, Plantamura I, Dugo M, D’Ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone MG and Iorio MV: Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 7:pp. e23122016, View Article : Google Scholar : PubMed/NCBI

32 

Mitra AK, Zillhardt M, Hua Y, Tiwari P, Murmann AE, Peter ME and Lengyel E: MicroRNAs reprogram normal fibroblasts into cancer-associated fibroblasts in ovarian cancer. Cancer Discov. 2:1100–1108. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Tang X, Hou Y, Yang G, Wang X, Tang S, Du YE, Yang L, Yu T, Zhang H, Zhou M, et al: Stromal miR-200s contribute to breast cancer cell invasion through CAF activation and ECM remod-eling. Cell Death Differ. 23:132–145. 2016. View Article : Google Scholar

34 

Du YE, Tu G, Yang G, Li G, Yang D, Lang L, Xi L, Sun K, Chen Y, Shu K, et al: MiR-205/YAP1 in activated fibroblasts of breast tumor promotes VEGF-independent angiogenesis through STAT3 signaling. Theranostics. 7:3972–3988. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Pang W, Su J, Wang Y, Feng H, Dai X, Yuan Y, Chen X and Yao W: Pancreatic cancer-secreted miR-155 implicates in the conversion from normal fibroblasts to cancer-associated fibroblasts. Cancer Sci. 106:1362–1369. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Dror S, Sander L, Schwartz H, Sheinboim D, Barzilai A, Dishon Y, Apcher S, Golan T, Greenberger S, Barshack I, et al: Melanoma miRNA trafficking controls tumour primary niche formation. Nat Cell Biol. 18:1006–1017. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Shen H, Yu X, Yang F, Zhang Z, Shen J, Sun J, Choksi S, Jitkaew S and Shu Y: Reprogramming of normal fibroblasts into cancer-associated fibroblasts by miRNAs-mediated CCL2/VEGFA signaling. PLoS Genet. 12:e10062442016. View Article : Google Scholar : PubMed/NCBI

38 

Zhu M, Wang M, Yang F, Tian Y, Cai J, Yang H, Fu H, Mao F, Zhu W, Qian H and Xu W: miR-155-5p inhibition promotes the transition of bone marrow mesenchymal stem cells to gastric cancer tissue derived MSC-like cells via NF-κB p65 activation. Oncotarget. 7:16567–16580. 2016.PubMed/NCBI

39 

Sung SY, Liao CH, Wu HP, Hsiao WC, Wu IH, Jinpu Yu, Lin SH and Hsieh CL: Loss of let-7 microRNA upregulates IL-6 in bone marrow-derived mesenchymal stem cells triggering a reactive stromal response to prostate cancer. PLoS One. 8:pp. e716372013, View Article : Google Scholar : PubMed/NCBI

40 

De Veirman K, Wang J, Xu S, Leleu X, Himpe E, Maes K, De Bruyne E, Van Valckenborgh E, Vanderkerken K, Menu E and Van Riet I: Induction of miR-146a by multiple myeloma cells in mesenchymal stromal cells stimulates their protumoral activity. Cancer Lett. 377:17–24. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Horiguchi H, Kobune M, Kikuchi S, Yoshida M, Murata M, Murase K, Iyama S, Takada K, Sato T, Ono K, et al: Extracellular vesicle miR-7977 is involved in hematopoietic dysfunction of mesenchymal stromal cells via poly(rC) binding protein 1 reduction in myeloid neoplasms. Haematologica. 101:437–447. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Jang JY, Lee JK, Jeon YK and Kim CW: Exosome derived from epigallocatechin gallate treated breast cancer cells suppresses tumor growth by inhibiting tumor-associated macrophage infiltration and M2 polarization. BMC Cancer. 13:4212013. View Article : Google Scholar : PubMed/NCBI

43 

Xu Z, Zhao L, Zhu LY, He M, Zheng L and Wu Y: MicroRNA-17, 20a regulates the proangiogenic function of tumor-associated macrophages via targeting hypoxia-inducible factor 2α. PLoS One. 8:pp. e778902013, View Article : Google Scholar

44 

Lagrange B, Martin RZ, Droin N, Aucagne R, Paggetti J, Largeot A, Itzykson R, Solary E, Delva L and Bastie JN: A role for miR-142-3p in colony-stimulating factor 1-induced monocyte differentiation into macrophages. Biochim Biophys Acta. 1833:1936–1946. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Baj-Krzyworzeka M, Mytar B, Szatanek R, Surmiak M, Węglarczyk K, Baran J and Siedlar M: Colorectal cancer-derived microvesicles modulate differentiation of human monocytes to macrophages. J Transl Med. 14:362016. View Article : Google Scholar : PubMed/NCBI

46 

Li Y, Zhao L, Shi B, Ma S, Xu Z, Ge Y, Liu Y, Zheng D and Shi J: Functions of miR-146a and miR-222 in tumor-associated macrophages in breast cancer. Sci Rep. 5:186482015. View Article : Google Scholar : PubMed/NCBI

47 

Huber V, Vallacchi V, Fleming V, Hu X, Cova A, Dugo M, Shahaj E, Sulsenti R, Vergani E, Filipazzi P, et al: Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J Clin Invest. 128:5505–5516. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Ren W, Hou J, Yang C, Wang H, Wu S, Wu Y, Zhao X and Lu C: Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J Exp Clin Cancer Res. 38:622019. View Article : Google Scholar : PubMed/NCBI

49 

Chaudhuri AA, So AY, Sinha N, Gibson WS, Taganov KD, O’Connell RM and Baltimore D: MicroRNA-125b potentiates macrophage activation. J Immunol. 187:5062–5068. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Ke M, Zhang Z, Cong L, Zhao S, Li Y, Wang X, Lv Y, Zhu Y and Dong J: MicroRNA-148b-colony-stimulating factor-1 signaling-induced tumor-associated macrophage infiltration promotes hepatocellular carcinoma metastasis. Biomed Pharmacother. 120:1095232019. View Article : Google Scholar : PubMed/NCBI

51 

He M, Xu Z, Ding T, Kuang DM and Zheng L: MicroRNA-155 regulates inflammatory cytokine production in tumor-associated macrophages via targeting C/EBPbeta. Cell Mol Immunol. 6:343–352. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Zonari E, Pucci F, Saini M, Mazzieri R, Politi LS, Gentner B and Naldini L: A role for miR-155 in enabling tumor-infiltrating innate immune cells to mount effective antitumor responses in mice. Blood. 122:243–252. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Zhao JL, Huang F, He F, Gao CC, Liang SQ, Ma PF, Dong GY, Han H and Qin HY: Forced activation of notch in macrophages represses tumor growth by upregulating miR-125a and disabling tumor-associated macrophages. Cancer Res. 76:1403–1415. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Squadrito ML, Pucci F, Magri L, Moi D, Gilfillan GD, Ranghetti A, Casazza A, Mazzone M, Lyle R, Naldini L and De Palma M: miR-511-3p modulates genetic programs of tumor-associated macrophages. Cell Rep. 1:141–154. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Lovat F, Fadda P, Mao C, Nuovo GJ, et al: MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci USA. 109:E2110–2116. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Frank AC, Ebersberger S, Fink AF, Lampe S, Weigert A, Schmid T, Ebersberger I, Syed SN and Brüne B: Apoptotic tumor cell-derived microRNA-375 uses CD36 to alter the tumor-associated macrophage phenotype. Nat Commun. 10:11352019. View Article : Google Scholar : PubMed/NCBI

57 

Xu L, Xu W, Wen Z and Xiong S: In situ prior proliferation of CD4+ CCR6+ regulatory T cells facilitated by TGF-β secreting DCs is crucial for their enrichment and suppression in tumor immunity. PLoS One. 6:e202822011. View Article : Google Scholar

58 

Yin Y, Cai X, Chen X, Liang H, Zhang Y, Li J, Wang Z, Chen X, Zhang W, Yokoyama S, et al: Tumor-secreted miR-214 induces regulatory T cells: A major link between immune evasion and tumor growth. Cell Res. 24:1164–1180. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Ye SB, Zhang H, Cai TT, Liu YN, Ni JJ, He J, Peng JY, Chen QY, Mo HY, Jun-Cui, 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. 240:329–340. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Hu Y, Wang C, Li Y, Zhao J, Chen C, Zhou Y, Tao Y, Guo M, Qin N, Ren T, et al: MiR-21 controls in situ expansion of CCR6+ regulatory T cells through PTEN/AKT pathway in breast cancer. Immunol Cell Biol. 93:753–764. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Qin A, Wen Z, Zhou Y, Li Y, Li Y, Luo J, Ren T and Xu L: MicroRNA-126 regulates the induction and function of CD4(+) Foxp3(+) regulatory T cells through PI3K/AKT pathway. J Cell Mol Med. 17:252–264. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Zhang M, Gao D, Shi Y, Wang Y, Joshi R, Yu Q, Liu D, Alotaibi F, Zhang Y, Wang H, et al: miR-149-3p reverses CD8+ T-cell exhaustion by reducing inhibitory receptors and promoting cyto-kine secretion in breast cancer cells. Open Biol. 9:1900612019. View Article : Google Scholar

63 

Lou Q, Liu R, Yang X, Li W, Huang L, Wei L, Tan H, Xiang N, Chan K, Chen J and Liu H: miR-448 targets IDO1 and regulates CD8+ T cell response in human colon cancer. J Immunother Cancer. 7:2102019. View Article : Google Scholar :

64 

Mangala LS, Wang H, Jiang D, Wu SY, Somasunderam A, Volk DE, Lokesh GLR, Li X, Pradeep S, Yang X, et al: Improving vascular maturation using noncoding RNAs increases antitumor effect of chemotherapy. JCI Insight. 3:pii: 122387. 2018.PubMed/NCBI

65 

Zhang H, Bai M, Deng T, Liu R, Wang X, Qu Y, Duan J, Zhang L, Ning T, Ge S, et al: Cell-derived microvesicles mediate the delivery of miR-29a/c to suppress angiogenesis in gastric carcinoma. Cancer Lett. 375:331–339. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Di Modica M, Regondi V, Sandri M, Iorio MV, Zanetti A, Tagliabue E, Casalini P and Triulzi T: Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Lett. 384:94–100. 2017. View Article : Google Scholar

67 

Guan B, Wu K, Zeng J, Xu S, Mu L, Gao Y, Wang K, Ma Z, Tian J, Shi Q, et al: Tumor-suppressive microRNA-218 inhibits tumor angiogenesis via targeting the mTOR component RICTOR in prostate cancer. Oncotarget. 8:8162–8172. 2017. View Article : Google Scholar :

68 

Dimitrova N, Gocheva V, Bhutkar A, Resnick R, Jong RM, Miller KM, Bendor J and Jacks T: Stromal expression of miR-143/145 promotes neoangiogenesis in lung cancer development. Cancer Discov. 6:188–201. 2016. View Article : Google Scholar :

69 

Smits M, Wurdinger T, van het Hof B, Drexhage JA, Geerts D, Wesseling P, Noske DP, Vandertop WP, de Vries HE and Reijerkerk A: Myc-associated zinc finger protein (MAZ) is regulated by miR-125b and mediates VEGF-induced angiogenesis in glioblastoma. FASEB J. 26:2639–2647. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Fang L, Deng Z, Shatseva T, Yang J, Peng C, Du WW, Yee AJ, Ang LC, He C, Shan SW and Yang BB: MicroRNA miR-93 promotes tumor growth and angiogenesis by targeting integrin-β8. Oncogene. 30:806–821. 2011. View Article : Google Scholar

71 

Fang L, Du WW, Yang W, Rutnam ZJ, Peng C, Li H, O'Malley YQ, Askeland RW, Sugg S, Liu M, et al: MiR-93 enhances angio-genesis and metastasis by targeting LATS2. Cell Cycle. 11:4352–4365. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Szczyrba J, Nolte E, Hart M, Döll C, Wach S, Taubert H, Keck B, Kremmer E, Stöhr R, Hartmann A, et al: Identification of ZNF217, hnRNP-K, VEGF-A and IPO7 as targets for microRNAs that are downregulated in prostate carcinoma. Int J Cancer. 132:775–784. 2013. View Article : Google Scholar

73 

Cheng J, Chen Y, Zhao P, Liu X, Dong J, Li J, Huang C, Wu R and Lv Y: Downregulation of miRNA-638- promotes angiogenesis and growth of hepatocellular carcinoma by targeting VEGF. Oncotarget. 7:30702–30711. 2016.PubMed/NCBI

74 

Schauer IG, Sood AK, Mok S and Liu J: Cancer-associated fibroblasts and their putative role in potentiating the initiation and development of epithelial ovarian cancer. Neoplasia. 13:393–405. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Shen Z, Qin X, Yan M, Li R, Chen G, Zhang J and Chen W: Cancer-associated fibroblasts promote cancer cell growth through a miR-7-RASSF2-PAR-4 axis in the tumor microenvi-ronment. Oncotarget. 8:1290–1303. 2017.

76 

Wang X, Qin X, Yan M, Shi J, Xu Q, Li Z, Yang W, Zhang J and Chen W: Loss of exosomal miR-3188 in cancer-associated fibroblasts contributes to HNC progression. J Exp Clin Cancer Res. 38:1512019. View Article : Google Scholar : PubMed/NCBI

77 

Biswas SK, Allavena P and Mantovani A: Tumor-associated macrophages: Functional diversity, clinical significance, and open questions. Semin Immunopathol. 35:585–600. 2013. View Article : Google Scholar : PubMed/NCBI

78 

De Palma M and Lewis CE: Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell. 23:277–286. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Epelman S, Lavine KJ and Randolph GJ: Origin and functions of tissue macrophages. Immunity. 41:21–35. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Yin Z, Ma T, Huang B, Lin L, Zhou Y, Yan J, Zou Y and 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. 38:3102019. View Article : Google Scholar

81 

Min S, Li L, Zhang M, Zhang Y, Liang X, Xie Y, He Q, Li Y, Sun J, Liu Q, et al: TGF-β-associated miR-27a inhibits dendritic cell-mediated differentiation of Th1 and Th17 cells by TAB3, p38 MAPK, MAP2K4 and MAP2K7. Genes Immun. 13:621–631. 2012. View Article : Google Scholar : PubMed/NCBI

82 

Nguyen DX, Bos PD and Massague J: Metastasis: From dissemination to organ-specific colonization. Nat Rev Cancer. 9:274–284. 2009. View Article : Google Scholar : PubMed/NCBI

83 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, Li C, Chong M, Ibrahim T, Mercatali L, et al: miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 15:284–294. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Li BL, Lu W, Qu JJ, Ye L, Du GQ and Wan XP: Loss of exosomal miR-148b from cancer-associated fibroblasts promotes endometrial cancer cell invasion and cancer metastasis. J Cell Physiol. 234:2943–2953. 2019. View Article : Google Scholar

86 

Cuiffo BG, Campagne A, Bell GW, Lembo A, Orso F, Lien EC, Bhasin MK, Raimo M, Hanson SE, Marusyk A, et al: MSC-regulated microRNAs converge on the transcription factor FOXP2 and promote breast cancer metastasis. Cell Stem Cell. 15:762–774. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Zhou SL, Hu ZQ, Zhou ZJ, Dai Z, Wang Z, Cao Y, Fan J, Huang XW and Zhou J: miR-28-5p-IL-34-macrophage feedback loop modulates hepatocellular carcinoma metastasis. Hepatology. 63:1560–1575. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Lou G, Song X, Yang F, Wu S, Wang J, Chen Z and Liu Y: Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 8:1222015. View Article : Google Scholar : PubMed/NCBI

89 

Wei X, Tang C, Lu X, Liu R, Zhou M, He D, Zheng D, Sun C and Wu Z: MiR-101 targets DUSP1 to regulate the TGF-β secretion in sorafenib inhibits macrophage-induced growth of hepatocar-cinoma. Oncotarget. 6:18389–18405. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Zheng P, Chen L, Yuan X, Luo Q, Liu Y, Xie G, Ma Y and Shen L: Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 36:532017. View Article : Google Scholar : PubMed/NCBI

91 

Tanaka K, Miyata H, Sugimura K, Fukuda S, Kanemura T, Yamashita K, Miyazaki Y, Takahashi T, Kurokawa Y, Yamasaki M, et al: miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts. Carcinogenesis. 36:894–903. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Bryant RJ, Pawlowski T, Catto JW, Marsden G, Vessella RL, Rhees B, Kuslich C, Visakorpi T and Hamdy FC: Changes in circulating microRNA levels associated with prostate cancer. Br J Cancer. 106:768–774. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Lee YS, Lim YS, Lee JC, Wang SG, Park HY, Kim SY and Lee BJ: Differential expression levels of plasma-derived miR-146b and miR-155 in papillary thyroid cancer. Oral Oncol. 51:77–83. 2015. View Article : Google Scholar

94 

Yi SJ, Liu P, Chen BL, Ou-Yang L, Xiong WM and Su JP: Circulating miR-31-5p may be a potential diagnostic biomarker in nasopharyngeal carcinoma. Neoplasma. 66:825–829. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Kosaka N, Iguchi H and Ochiya T: Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 101:2087–2092. 2010. View Article : Google Scholar : PubMed/NCBI

96 

Hornick NI, Huan J, Doron B, Goloviznina NA, Lapidus J, Chang BH and Kurre P: Serum exosome MicroRNA as a minimally-invasive early biomarker of AML. Sci Rep. 5:112952015. View Article : Google Scholar : PubMed/NCBI

97 

Donahue TR, Nguyen AH, Moughan J, Li L, Tatishchev S, Toste P and Farrell JJ: Stromal microRNA-21 levels predict response to 5-fluorouracil in patients with pancreatic cancer. J Surg Oncol. 110:952–959. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Cubillos-Ruiz JR, Baird JR, Tesone AJ, Rutkowski MR, Scarlett UK, Camposeco-Jacobs AL, Anadon-Arnillas J, Harwood NM, Korc M, Fiering SN, et al: Reprogramming tumor-associated dendritic cells in vivo using miRNA mimetics triggers protective immunity against ovarian cancer. Cancer Res. 72:1683–1693. 2012. View Article : Google Scholar : PubMed/NCBI

99 

Huang Z, Gan J, Long Z, Guo G, Shi X, Wang C, Zang Y, Ding Z, Chen J, Zhang J and Dong L: Targeted delivery of let-7b to reprogramme tumor-associated macrophages and tumor infiltrating dendritic cells for tumor rejection. Biomaterials. 90:72–84. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Xi J, Huang Q, Wang L, Ma X, Deng Q, Kumar M, Zhou Z, Li L, Zeng Z, Young KH, et al: miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immuno-therapy. Oncogene. 37:3151–3165. 2018. View Article : Google Scholar : PubMed/NCBI

101 

Pi J, Tao T, Zhuang T, Sun H, Chen X, Liu J, Cheng Y, Yu Z, Zhu HH, Gao WQ, et al: A MicroRNA302-367-Erk1/2-Klf2-S1pr1 pathway prevents tumor growth via restricting angiogenesis and improving vascular stability. Circ Res. 120:85–98. 2017. View Article : Google Scholar

102 

Schnittert J, Kuninty PR, Bystry TF, Brock R, Storm G and Prakash J: Anti-microRNA targeting using peptide-based nano-complexes to inhibit differentiation of human pancreatic stellate cells. Nanomedicine (Lond). 12:1369–1384. 2017. View Article : Google Scholar

103 

Pecot CV, Calin GA, Coleman RL, Lopez-Berestein G and Sood AK: RNA interference in the clinic: Challenges and future directions. Nat Rev Cancer. 11:59–67. 2011. View Article : Google Scholar :

104 

Ling H, Fabbri M and Calin GA: MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 12:847–865. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 56 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pan Z, Tian Y, Niu G and Cao C: Role of microRNAs in remodeling the tumor microenvironment (Review). Int J Oncol 56: 407-416, 2020
APA
Pan, Z., Tian, Y., Niu, G., & Cao, C. (2020). Role of microRNAs in remodeling the tumor microenvironment (Review). International Journal of Oncology, 56, 407-416. https://doi.org/10.3892/ijo.2019.4952
MLA
Pan, Z., Tian, Y., Niu, G., Cao, C."Role of microRNAs in remodeling the tumor microenvironment (Review)". International Journal of Oncology 56.2 (2020): 407-416.
Chicago
Pan, Z., Tian, Y., Niu, G., Cao, C."Role of microRNAs in remodeling the tumor microenvironment (Review)". International Journal of Oncology 56, no. 2 (2020): 407-416. https://doi.org/10.3892/ijo.2019.4952