Abstract
Cancer cells grow in an environment comprised of multiple components that support tumor growth and contribute to therapy resistance. Major cell types in the tumor micro-environment are fibroblasts, endothelial cells and infiltrating immune cells all of which communicate with cancer cells. One way that these cell types promote cancer progression is by altering expression of miRNAs, small noncoding RNAs that negatively regulate protein expression, either in the cancer cells or in associated normal cells. Changes in miRNA expression can be brought about by direct interaction between the stromal cells and cancer cells, by paracrine factors secreted by any of the cell types, or even through direct communication between cells through secreted miRNAs. Understanding the role of miRNAs in the complex interactions between the tumor and cells in its micro-environment is necessary if we are to understand tumor progression and devise new treatments.
Introduction
The tumor micro-environment
The tumor micro-environment (TME) is composed of fibroblasts, blood vessels, immune cells, support cells, signaling molecules, and the extracellular matrix (ECM). The proportion of this stroma in human cancers can be over 90% 1, 2. The tumor and the surrounding micro-environment affect each other through close and constant interactions and together play a significant role in treatment outcomes 3 (Figure 1).
Figure 1.

An overview of the role of miRNAs in the tumor microenvironment.
Endothelial cells line the interior surface of blood vessels and lymphatic vessels, forming an interface between circulating blood or lymph in the lumen and the rest of the vessel wall. In tumors, aggressive and unregulated growth of neoplastically transformed cells that overexpress pro-angiogenic factors leads to the development of disorganized blood vessel networks that are fundamentally different from normal vasculature 3.
Cancer associated fibroblasts (CAFs) are a major constituent of the tumor stroma 4, 5. CAFs isolated from cancer patients have a different morphology and function than normal fibroblasts and have been shown to promote the invasion and growth of tumor cells 5. CAFs produce growth factors (VEGF) and cytokines (TGFβ, Interleukin(IL)-6, IL-10) that activate the adjacent ECM, which then contributes to the growth of the cancer cells. Additionally, CAFs are the primary source of an altered ECM, containing fibronectin and collagen, that also contributes to tumor growth 4. Major secreted factors include proinflammatory cytokines such as IL-1β and IL-8 which have been associated with pro-tumorigenic effects. A prominent chemokine that is secreted by CAFs is SDF-1 α, which primarily signals through the CXC chemokine receptor 4 (CXCR4) which promotes proliferation 4. CAFs retain their characteristics (at least partially) when cultured in the absence of malignant cells, suggesting that they have undergone genetic or epigenetic alterations. A recent series of studies reported genetic alterations in microdissected CAFs derived from breast, head and neck, and ovarian cancer 4-6. However, careful analysis of breast and ovarian CAFs demonstrated that such genetic alterations are extremely rare 7. It is more likely that the stable phenotype of CAFs is regulated by epigenetic changes, as suggested by a genome-wide analysis of breast cancer stroma 8.
Many cancers are driven by inflammation mediated by bone-marrow derived cells, such as monocytes and macrophages. Macrophages are innate immune cells that play indispensable roles in the innate and adaptive immune response. They can be activated by a variety of stimuli and polarized to functionally different phenotypes. Two distinct subsets of macrophages are now recognized: classically activated (M1) and alternatively activated (M2) macrophages 9. M1 macrophages express a series of proinflammatory cytokines, chemokines, and effector molecules, such as TNF- α, IL-12, IL-23, iNOS and MHC class I and II. In contrast, M2 macrophages express a wide array of anti-inflammatory molecules, such as IL-10, TGF- β, and arginase. In most tumors, infiltrating macrophages are of the M2 phenotype, which provides an immunosuppressive micro-environment for tumor growth. In addition, these tumor-associated macrophages secrete many cytokines, chemokines, and proteases, which also promote tumor angiogenesis, growth, metastasis, and immunosuppression 9. This review is about the role that microRNAs play in the communication between tumor cells and components of the TME (Figure 1).
miRNAs and cancer
Micro (mi)RNAs are small, 19-22 nucleotide (nt) long, non-coding RNAs that inhibit gene expression at the posttranscriptional level. They are first transcribed as parts of longer molecules, up to several kilobases in length (pri-miRNA), that are processed in the nucleus into hairpin RNAs of 70-100 nt by the double-stranded RNA-specific ribonuclease, Drosha 10. The hairpin pre-miRNAs are then transported by exportin 5 to the cytoplasm, where they undergo further processing by a second, double-strand specific ribonuclease, called Dicer. In animals, single-stranded miRNAs are incorporated into RNA induced silencing complex (RISC) and primarily bind specific messenger RNA (mRNA) at specific sequence motifs, predominantly within the 3′ untranslated region (3′UTR) of the transcript, that are significantly, though not completely, complementary to the miRNA. The mRNA/miRNA duplex then inhibits translation by blocking initiation or through increased degradation of the mRNA. Given the frequency with which miRNA target motifs are conserved within 3′UTRs, it is estimated that almost all human genes are conserved targets of miRNAs, and that for each miRNA there are approximately 200 genes that carry conserved sequence motifs within the 3′UTR 10.
miRNAs, which are frequently deregulated in many types of cancer, have been shown to play roles in multiple facets of tumorigenesis. The cancer cell autonomous roles of miRNAs have been thoroughly reviewed elsewhere 11. In this review the focus will be on the non-cell autonomous roles of miRNAs in cancer cells, specifically in how alterations in miRNA expression in cancer cells promotes tumorigenesis by modifying the TME. The modification of the TME through the regulation of miRNA expression by oncogenes was first reported in 2006 12. Using a colonocyte model with stable expression of Ras or Ras and myc, Dews et al. demonstrated that myc upregulates the miR-17-92 family, therefore promoting blood perfusion and endothelial cell recruitment 12.
A strong relationship between miRNAs and human cancers has been established. A comparison of miRNA expression in normal and tumor tissues showed global changes in miRNA expression in various human malignancies 10. In addition, mapping 186 human miRNA genes revealed that they are frequently located at fragile sites and other cancer-associated chromosomal regions 13. Consequently, miRNAs have been identified either as oncogenes (e.g. miR-155, miR-17-5p, miR-21, miR-221/222) or tumor suppressors (e.g. miR-34, miR-15a, miR-16-1, let-7) 10, 14. Thus, while miRNA deregulation in cancer cells plays a known role in tumor progression, metastasis, and therapy resistance, miRNAs have recently been shown to participate in reprogramming components of the TME to provide assistance for the cancer organ.
Reprogramming of somatic cells by miRNAs
Until 9 years ago, it was believed that most differentiated tissues were irreversibly locked into their differentiation states and that tissues could only be generated with pluripotent stem cells. This changed in 2006, with the demonstration that the expression of 4 transcription factors could reprogram somatic fibroblasts into induced pluripotent stem (iPS) cells 15. Since then, miRNAs were identified that are expressed only in embryonic stem cells and it was reported that miR-302 16 could reprogram somatic cells into iPS cells 17. In addition, it was demonstrated that expression of two miRNAs (miR-9/9* and miR-124) could convert fibroblasts into neurons 18, proving the power of miRNAs to reprogram adult fibroblasts. These data showed that the genome has much more plasticity than previously expected and that miRNAs are powerful regulators of this plasticity. In the context of cancer, the idea developed that investigating the reprogramming of cancer associated stroma cells could lead to a better understanding of cancer progression and to new treatments. This review highlights a number of miRNAs and their roles in reprogramming major components of the TME and discuss miRNAs that change in the cancer cells in response to interaction with components of the TME.
The role of miRNAs in cancer associated endothelial cells (CAEs)
Since endothelial cells are critical for angiogenesis and regulating tumor metastasis, their recruitment by cancer cells can substantially promote overall tumorigenesis. Through manipulation of the miRNA expression of endothelial cells, the cancer cells are able to reprogram these cells to enhance their angiogenic potential (Figure 2).
Figure 2.

Deregulated miRNAs in cancer associated endothelial cells (CAE) or cancer cells interacting with CAE. When known miRNA targets are given. BrCa: breast cancer; HCC, hepatocellular carcinoma; OvCa: ovarian cancer; CRC: colorectal cancer. References: [1], 21; [2], 19; [3], 20; [4], 22; [5], 23; [6], 25; [7], 24; [8], 36; [9], 12; [10], 29; [11], 27; [12], 30; [13], 31; [14], 37; [15], 32; [16], 33; [17], 39; [18], 34; [19], 35.
In two different cancer models, a similar approach was taken to determine how changes in miRNA expression in endothelial cells affect co-cultured cancer cells. When endothelial cells were co-cultured with hepatocellular carcinoma (HCC) cells, 4 miRNAs were significantly altered (>1.5 fold) in the endothelial cells, with miR-146a, miR-181a*, and miR-140-5p upregulated and miR-302c downregulated 19. The upregulation of miR-146a was found to promote endothelial cell migration and proliferation and to promote tumor growth and vascularization 19. This activity occurred through miR-146a directly targeting BRCA-1, which then negatively regulated expression of PDGFRA through targeting the PDGFRA promoter 19. When endothelial cells were co-cultured with glioma cell lines, a miRNA array analysis identified 12 miRNAs that were downregulated (miR-181a, miR-101, miR-30b, miR-27a, miR-21, miR-22, miR-23b, miR-31, miR-103, miR-126, miR-29a, and miR-125b) and one upregulated (miR-296) 20. miR-296 was found to promote angiogenesis through increasing endothelial tube formation and migration. The glioma cells reprogrammed the endothelial cells to upregulate miR-296 through VEGF and EGF 20. It was also shown that VEGF upregulates expression of miR-10b and miR-196b in murine breast and lung cancer models 21. These two miRNAs were found to be critical for angiogenesis and tumor growth in these cancers 21.
miR-125b is another miRNA that clearly has an important, though at present difficult to define, role in endothelial cell reprogramming during tumorigenesis. In gliomas a miRNA array detected the downregulation of miR-125b, which was confirmed to have a role in endothelial migration 22, 23. Once again VEGF played a role in reprogramming the endothelial cells through altering miRNA expression. VEGF downregulated miR-125b, leading to the upregulation of MAZ (Myc-associated zinc finger protein), which is a transcription factor that binds the promoter of VEGF. This process in the endothelial cells clearly promoted vascularization and angiogenesis 23. However, in another report miR-125b was shown to be upregulated by VEGF, FGF, and hypoxia in murine lung cancer models resulting in reduced vascularization and tumorigenesis 22. Whether these contradictions concerning the role of miR-125b in the reprogramming of endothelial cells are based on differences in cancer model or on experimental reasons needs to be further explored.
In breast cancer, alterations in miRNA expression have been shown to regulate angiogenesis through direct effects on endothelial cells and immune infiltration. Specifically, the upregulation of miR-155 and miR-93 24, 25 or the downregulation of miR-126 and miR-29b 26, 27 have been linked to angiogenesis through effects on endothelial cells, while downregulation of miR-92a may play a role in macrophage infiltration 28. miR-126 was shown to directly target both IGFBP2, which is a secreted endothelial recruitment factor and PITPNC1, the phosphatidylinositol transfer protein that regulates IGFBP2 secretion. Thus, the downregulation of miR-126 enhances endothelial recruitment and tumor growth by increasing IGFBP2 secretion. miR-126 was also shown to employ another distinct mechanism for modifying the chemotactic gradient through targeting MERTK, which when secreted binds to and neutralizes the negative chemotactic factor, GAS6. Therefore, the downregulation of miR-126 resulted in the upregulation of MERTK, which, by neutralizing physiological levels of GAS6, led to enhanced endothelial cell chemotaxis 27. Indeed, miR-126 downregulation was found to be necessary for endothelial cell recruitment in this breast cancer model 27. The upregulation of miR-155 has also been shown to be important in breast cancer, by promoting angiogenesis through the enhanced migration of endothelial cells and macrophages 25. miR-155 directly targets VHL, which is critical for the proangiogenic effects of the increased expression of miR-155 25. Additionally, miR-93 was found to be upregulated in breast carcinoma patients 24. Increased expression of miR-93 enhanced endothelial tube formation, increased blood vessel density in vivo, and promoted the formation of lung metastases through targeting of LATS2 24. The altered expression of miR-29b, which was downregulated through the loss of GATA3, also promoted tumorigenesis through enhanced angiogenesis, resulting in a poor prognosis in luminal breast cancer. Downregulation of miR-29b enhanced TGF-β signaling and promoted an overall epithelial-to-mesenchymal transition (EMT) 29. Furthermore, expression of VEGFA, ITGA6, ANGPTL4, and LOX, known mediators of angiogenesis, were inversely correlated with expression of miR-29b 29. Moreover, miRNA changes in cancer cells, as well as endothelial cells are important in breast cancer. The downregulation of miR-92a in the cancer cells correlated with a worse prognosis and reduced macrophage infiltration 28.
miR-29b, miR-214, and miR-195 are also downregulated in hepatocellular carcinoma (HCC) and coordinate the TME through the regulation of endothelial cells. As is the case in breast cancer, downregulation of miR-29b in HCC enhanced tumorigenesis through increased microvessel density and endothelial tube formation 26. This effect may be due to an increase in MMP-2 expression caused by the downregulation of miR-29b, which promotes VEGFR (also known as Flt-1) signaling in the endothelial cells 26. Another miRNA, miR-195, directly targets VEGF in HCC 30. The downregulation of miR-195 increased expression of VEGF, leading to enhanced endothelial cell branching and tube formation during co-culture 30. An analysis of a miRNA array of tumors from HCC patients, found that miR-214 was highly downregulated and that this downregulation was indicative of a worse prognosis 31. miR-214 was found to regulate endothelial cell branching through targeting HDGF, which is known to be involved with angiogenesis 31.
In lung cancer, downregulation of miR-519c, miR-126, and let-7b was found to promote angiogenesis through effects on endothelial cells. Additionally, downregulation of miR-200b was shown to promote metastasis through the regulation of CAFs and the regulation of endothelial cell recruitment in renal and ovarian cancer as well as lung cancer. miR-519c, which is downregulated by HGF, regulated angiogenesis by targeting HIF-1α 32. The downregulation of miR-519c in lung cancer promoted the secretion of proangiogenic factors, VEGF-A, bFGF, and IL-8, which enhanced endothelial tube formation and increased blood vessel density within the TME 32. Additionally, it was shown that downregulation of miR-126 and let-7b in both the tumor and the stroma was associated with increased microvessel density and a worse prognosis 33. While the mechanism and mediators of this increased microvessel density are not known, this observation suggests a role of miRNAs in angiogenesis in lung cancer.
Downregulation of miR-200 family members has been found to affect endothelial cell activity, and has been shown, through analysis of the TGCA dataset, to indicate a worse prognosis in lung, renal, and ovarian cancer 34. While the role of miR-200 in EMT is well known, it was shown that the downregulation of miR-200 family members reduced the amount of endothelial cells in the TME through targeting IL-8 34. Furthermore, increased expression of miR-200 in endothelial cells reduced migration and tube formation indicating that miR-200 is a regulator of angiogenesis in the TME 34.
Evidence of epigenetic modification of miRNA expression to promote angiogenesis was seen in ovarian cancer, melanoma, and glioblastoma. In addition to the downregulation of miR-200 in ovarian cancer, miR-484 was found to be downregulated in ovarian cancer patient tumor samples and was linked to an increase in blood vessels, possibly through targeting VEGFB and VEGFR2 35. In melanoma, in vivo passaging of cancer cells to generate highly metastatic clones, indicated that miR-1908, miR-214, miR-199a-5p, and miR-199-3p were upregulated and functional mediators of metastasis 36. Increased expression of miR-1908, miR-199a-5p, and miR-199a-3p indicated a worse prognosis 36. These miRNAs were shown to regulate microvessel density, endothelial cell recruitment, and melanoma metastasis and their increased expression was linked to a worse prognosis. Their mechanism of action was through targeting DNAJA4 and also ApoE, which reduces endothelial cell recruitment by binding the LRP8 receptor on endothelial cells 36. A similar approach, using in vivo passaging, was taken with glioblastoma. A human glioblastoma line was passaged in nude mice to identify clones with different dormancy rates determined by rate of outgrowth upon challenge 37. In the fast growing glioblastoma lines, miR-580, miR-588, and miR-190 were downregulated, which increased proangiogenic factors TGFα, HIF-1α, FGF2, TIMP-3, and Kras 37. Reconstitution of miR-588 in fast growing glioblastoma cell lines reduced recruitment of endothelial cells and MDSCs 37. Furthermore, increased expression of miR-93 in glioblastoma promoted endothelial cell proliferation and migration, possibly through targeting integrin B8 38.
In summary, many of the epigenetic changes in miRNA expression in cancer involve the recruitment of endothelial cells, which promote angiogenesis. However, a large number of studies identified other cells within the TME that change miRNA expression in the setting of cancer.
The role of miRNAs in cancer associated fibroblasts (CAFs)
Fibroblasts regulate angiogenesis, metastasis, EMT and extracellular matrix composition within the tumor. Not surprisingly, miRNAs are key regulators of the tumor promoting functions of CAFs and it is becoming more readily apparent that cancer directed changes regulate this miRNA network. In a large number of cancers CAFs have been shown to promote tumorigenesis by upregulating and downregulating specific miRNAs (Figure 3).
Figure 3.

Deregulated miRNAs in cancer associated fibroblasts (CAF) or cancer cells interacting with CAF. When known miRNA targets are given. BrCa: breast cancer; OvCa: ovarian cancer; CRC: colorectal cancer; Panc: pancreatic cancer; Gastr: gastric cancer; PrCa: prostate cancer; Eso squam: esophageal squamous cell carcinoma. References: [1], 49; [2], 48; [3], 40; [4], 41; [5], 42; [6], 43; [7], 46]; [8], 44; [9], 52; [10], 53; [11], 51; [12], 50; [13], 117.
During tumorigenesis miRNAs are frequently downregulated in what has been considered a dedifferentiation mechanism. More specifically, lung cancer cells downregulate miR-200b, which targets Flt-1 to promote CAF migration that increase metastasis 39. While the majority of miRNAs that are altered in the micro-environment are downregulated, the increase of several miRNAs of note has been shown to promote tumorigenesis. These include well known oncomirs such as miR-21 and miR-155. miR-21 has been shown to be highly upregulated in fibroblasts from patients in multiple tumor types, including colorectal cancer 40-42, squamous cell carcinoma 43, and pancreatic cancer 44. Increased expression of miR-21 by fibroblasts was shown to upregulate α -SMA, resulting in the acquisition of a more activated myofibroblast phenotype 40, 45. Cancer cells regulated miR-21 through soluble factors, including TGF- β, which has been shown to be relevant in several different cancer models 40, 44, 45. For example, TGF- β was shown to upregulate miR-143 in CAFs in scirrhous type gastric cancer 46. These results highlight that regulation of tumorigenesis by “oncomirs” is not cancer autonomous, but rather a function of the entire tumor organ.
As previously mentioned, miR-155 is often upregulated in CAFs. miR-155 has a prominent role in inflammation, since it is directly regulated by NF- κ B 47, a well-known regulator of cancer associated fibroblast function. We recently reported that upregulation of miR-155 contributed to the transition of normal omental fibroblasts into CAFs in ovarian cancer 48. Gene expression analysis of these CAFs and the effects of the upregulation of miR-155 showed an increase in the expression of chemokines and other immune modulators which may be congruent with the effects of miR-155 and its known regulator, NF- κ B. However further dissection of the role of miR-155 and its overlap with NF- κ B in the promotion of tumor growth by CAFs will need to be pursued further, since this study focused on the coordinated deregulation of 3 miRNAs (the upregulation of miR-155 with the downregulation of both miR-31 and miR-214) 48.
A miRNA array was also used to identify altered signaling pathways in fibroblasts from breast cancer patients. miR-221-5p, miR-221-3p and miR-31-3p were found to be upregulated in these CAFs 49. The authors then performed gene ontology analysis, searching for pathways that were regulated by multiple deregulated miRNAs. Many pathways were identified, including MAPK, IL-6, tight junction, insulin, chemokine, TGF- β, and HGFR signaling 49. Additional analysis validated the activation of the IL-6 pathway and downstream signaling of TGF- β with increased MMP-1, MMP-2, and MMP-9 in CAFs from breast cancer 49. However, the role and significance of these upregulated miRNAs in regulating CAFs will need to be further dissected because miR-205, miR-200c, miR-200b, miR-141, miR-101, miR-342-3p, let-7g, and miR-26b were also shown to be significantly downregulated 49.
Interestingly, the study discussed above, which performed a miRNA array to identify deregulated expression in of CAFs in breast cancer found that miR-31 was upregulated 49 while the report on ovarian cancer metastases found that miR-31 was downregulated. 48. Further profiling of miRNA expression in endometrial cancer showed that miR-31 is downregulated in fibroblasts 50. This downregulation of miR-31 permitted the expression of SATB2, promoting endometrial cancer invasiveness and growth (49). The apparent paradox of the different regulation of miR-31 in different types of cancer may reflect the distinct tissue origin that determines the unique context for the growth of each individual tumor. Interestingly, in endometrial cancer the downregulation of miR-148a, as well as miR-31, in CAFs is critical to the promotion of tumorigenesis. The downregulation of miR-148 in CAFs increases Wnt activity through elevated expression of Wnt10B, which is a target of miR-148 51.
Another study that performed miRNA array analysis on CAFs in breast cancer reported 6 miRNAs that were downregulated more then 10-fold, including miR-7f, let-7g, miR-107, miR-15b, miR-26b, and miR-30b 52. The downregulation of miR-26b in CAFs promoted the migration of fibroblasts, which is a functional determinant of the CAF phenotype 52. Furthermore, the downregulation of miR-26b in CAFs promoted the invasion of the human breast cancer cell line, MCF-7 52. Additionally, PTEN expression was reduced in the stroma of invasive breast cancer patients, which was correlated with miR-320 expression 53, 54. Loss of PTEN expression in stromal fibroblasts led to a protumorigenic secretome, which was partially mediated by the downregulation of miR-320. Furthermore, the authors determined that miR-320 directly regulated the transcription factor ETS2, and ETS2 binding sites were found in 20 genes of the 54 gene secretome identified through PTEN deletion 53. The downregulation of miR-320 in fibroblasts promoted cancer cell invasion and endothelial proliferation through direct targeting of MMP-9 and EMILIN2, respectively. EMILIN2 is a secreted factor important for angiogenesis through the promotion of endothelial cell proliferation 55. That miR-320 regulated expression of MMP-9 and EMILIN2 both directly and indirectly was indicated by the presence of ETS2 binding sites that are responsible for their regulation. Thus, miR-320 targets the upstream transcription factor and the downstream mediators regulating the protumorigenic secretome of fibroblasts 53.
In prostate cancer, as in breast cancer, miRNAs were downregulated in CAFs to promote tumorigenesis. In prostate cancer the downregulation of miR-15 and miR-16 in CAFs was likely mediated through activation of the AKT and ERK pathways, promoting prostate cancer migration and neovascularization (55). This data adds to our awareness of the diversity of miRNA regulation in fibroblasts, based on different tumor types and other unknown physiological factors.
The role of miRNAs in cancer associated mesothelial cells
While ovarian cancer interacts with CAFs, the primary micro-environment for ovarian cancer cells is the mesothelial cell. These cells cover the peritoneum, the small and large bowel serosa and the omentum. The most common sites of metastasis from the primary ovarian tumor are the abdominal peritoneum and the omentum. Metastasis is mediated by binding of ovarian cancer cells to mesothelial cells before they can access to the fibroblasts located underneath. Recently, it was shown that TGF β -stimulated human primary mesothelial cells, which induced expression of MMP-2 and MMP-9, were able to promote cancer cell attachment and proliferation through downregulating members of the miR-200 family. Consequently, the delivery of miR-200 family miRNAs to mesothelial cells in mice inhibited ovarian cancer cell implantation and dissemination 56. We recently showed that the direct interaction of OvCa cells with mesothelial cells, which cover the surface of the omentum, causes a DNA methyltransferase 1 (DNMT1) mediated decrease in the expression of miR-193b in the cancer cells 57. The reduction in miR-193b resulted in the upregulation of its target urokinase-type plasminogen activator (uPA), a known tumor associated protease. The paracrine signals emitted from the TME resulted in the deregulation of a key miRNA in the cancer cells, promoting the initial steps of OvCa metastatic colonization.
The deregulation of miRNAs in tumor-associated macrophages promotes tumorigenesis
Cancer cells reprogram and regulate the miRNA expression of infiltrating immune cells, not only to suppress an anti-tumor immune response but to highjack the wound healing response of these cells to promote tumorigenesis. Antigen presenting cells, especially macrophages, have been shown to be a primary target of this conversion. They represent an ideal target, given their juxtaposition between the innate and adaptive immune system. Macrophages that infiltrate the TME adopt an M2 polarized phenotype, which promotes tumor growth more than the inflammatory M1 phenotype. As expected, cancer cells upregulate and downregulate several miRNAs in immune cells to limit their anti-tumor response and reprogram them to tumorigenesis (Figure 4).
Figure 4.

Deregulated miRNAs in cancer associated macrophages (CAM) or cancer cells interacting with CAM. When known miRNA targets are given. BrCa: breast cancer; HCC, hepatocellular carcinoma; OvCa: ovarian cancer; CRC: colorectal cancer; Panc: pancreatic cancer; Gastr: gastric cancer. References: [1], 70; [2], 69; [3], 67; [4], 92; [5], 60; [6], 62; [7], 64; [8], 71; [9], 87; [10], 88; [11], 72; [12], 58; [13], 68; [14], 63.
miR-155 is a well-known regulator of the immune response, involving T cells, NK cells, B cells, and antigen presenting cells (macrophages and dendritic cells). Notably, miR-155 has been found to be persistently downregulated in tumor associated macrophages (TAMs) 58, 59. Soluble factors from HCC were shown to downregulate miR-155, increasing expression of the transcription factor C/EPB β and increasing production of IL-10 58. When expression of miR-155 was restored in macrophages or dendritic cells, both direct and indirect anti-tumor responses were promoted through the enhancement of T cell function. miR-155 targeted SOCS1, C/EPB β and possibly Ship1, which are transcriptional suppressors of Th1 type cytokine production (e.g. TNF α, IL-12, IFN γ, CCL5) 58, 60, 61. Several other miRNAs were also found to be downregulated within TAMs, including miR-142-3p 62, miR-125b 63, and miR-19a-3p 64. Downregulation of miR-142-3p limited the tumor infiltration of macrophages and reduced the therapeutic effect of adoptive T cell transfer 62. Restoring miR-125b expression in macrophages promoted an anti-tumor response by targeting IRF-4, a known promotor of the M2 macrophage phenotype 63, 65. Macrophages have been found to downregulate miR-19a-3p within the TME, possibly through a cancer derived soluble factor 64. The downregulation of miR-19a-3p promoted STAT3 signaling, because miR-19a-3p directly targets Fra-1 (Fos-related antigen), a member of the AP1 family involved in macrophage polarization and the upstream regulation of STAT3 64, 66. STAT3 expression in macrophages, which promotes immune suppression, was rescued upon overexpression of miR-19a-3p 64. In contrast, in pancreatic cancer miR-146a is upregulated in monocytes to prevent their differentiation and expression of costimulatory molecules 67. This is a similar mechanism of immune evasion as is seen with the downregulation of miR-142-3p, which is not only critical for macrophage infiltration but also for macrophage differentiation 62. Additionally, elevated levels of miR-200 in myeloid cells reduced angiogenesis through the targeting of VEGF that limited tumor growth in a murine lung cancer model 68.
miR-511-3p is part of an interesting negative feedback loop in TAMs. in which it is coordinately regulated with the mannose receptor, a marker for TAMs. However, instead of promoting tumorigenesis, miR-511-3p negatively regulates protumoral gene expression 69. This indicates that not all miRNA reprogramming in macrophages will drive tumorigenesis, but can be the result of negative feedback limiting the protumorigenic capabilities of TAMs.
The role of miRNAs in myeloid derived suppressor cells
Myeloid derived suppressor cells (MDSCs) are components of the myeloid compartment that, like TAMs, regulate immunosuppression within the TME. Therefore, these MDSCs can, through cancer directed miRNA reprogramming, suppress immune responses and promote tumor growth. A miRNA array identified 8 deregulated miRNAs in tumor infiltrating MDSCs in a murine breast cancer model. miR-494, miR-882, and miR-361 were upregulated and let-7e, miR-713, miR-133b, miR-466j, and miR-322 downregulated 70. miR-494 was the most significantly upregulated miRNA, and promoted breast cancer invasion through suppression of anti-tumor CD8 T cell responses, possibly in response to TGF- β, 70. miR-494 was shown to target PTEN in MDSCs, which is responsible for the enhanced immune suppression of CD8 T cells 70. Further evidence suggests that several miRNAs are downregulated in MDSCs to promote the differentiation of myeloid cells into MDSCs and to regulate immunosuppressive signaling pathways. The downregulation of miR-223 in the bone marrow were critical if myeloid progenitors were to differentiate into MDSCs in a murine breast cancer model. Stable overexpression of miR-223 prevented the differentiation into MDSCs and the ability to promote tumor growth through immune suppression 71. miR-17-5p and miR-20a regulated the immunosuppressive potential of MDSCs through targeting of STAT3 72. Cancers downregulate these miRNAs, increasing the expression of STAT3, which in turn enhanced immunosuppression, partially through STAT3 induction of reactive oxygen species production72.
It is evident, therefore, that cancer cells reprogram the myeloid compartment to evade the immune system and promote tumorigenesis. These miRNA alterations directly limit immune responses through polarizing macrophages from a M1 to a M2 phenotype and indirectly through reprogramming macrophages and MDSCs to inhibit adaptive immune responses. However, there is now growing evidence that immune cells within the TME are, themselves, able to regulate miRNA expression in cancer cells. Soluble factors secreted from macrophages have been shown to downregulate miR-7 in gastric cancer and promote tumorigenesis 73. Additionally, co-culture experiments pairing astrocytes with three different tumor types that commonly metastasize to the brain (melanoma, breast, and lung) showed that astrocytes downregulate miR-768-3p when exposed to any cancer cells 74. In addition, the downregulation of miR-768-3p was found to promote chemo resistance and cancer stem cell properties 74. In a model of ovarian cancer, MDSCs were shown to upregulate miR-101 and promote cancer “stemness” and miR-101 was shown to target CtBP2, which bound to the promoters of Sox2 and Nanog 75. Follicular dendritic cells promoted chemoresistance in follicular lymphoma and mantle cell lymphoma through upregulation of miR-181a 76. Follicular dendritic cells are commonly found adhered to lymphoma cells. This interaction was shown to upregulate miR-181a, which targets the BH3 only protein Bim, making the lymphoma cells less sensitive to mitoxantrone 76.
The role of miRNAs in tumor infiltrating T cells (TILs)
Components of the adaptive immune system paradoxically play two roles in cancer with CD8 T cells and CD4 T cells performing tumor surveillance while T regulatory cells (Tregs) suppress anti-tumor responses. To avoid immune detection and elimination cancer cells directly suppress T cells through multiple mechanisms including deregulating miRNA expression. Further, cancer cells indirectly subvert the immune system through enhancing the suppressive functions of adaptive immune cells like Tregs and NKT cells through altering miRNA expression in these cells. In addition to regulating miRNAs in TILs, cancer cells have been shown to alter their own miRNA programming in order to avoid immune detection.
CD4 T cells isolated from patients with glioblastoma multiforme (GBM) show repressed expression of miR-17∼92 family members, while the miR-17∼92 family was shown to be generally upregulated in Th1 type CD4 T cells 77. This suggests a skewing of T cells towards a Th2 phenotype in the TME. Additionally, TGF- β, an immune suppressive cytokine commonly elevated in cancer, suppresses anti-tumor T cell responses by upregulating miR-23a in TILs, which targets BLIMP-1 reducing CTL expression of anti-tumor effector molecules, granzyme B and IFN γ 78. As previously discussed miR-155, is a miRNA associated with inflammation in the TME. Increased expression of miR-155 in T cells was shown to promote anti-tumor responses 77, 79. miR-155 expression in T cells negatively correlated with SOCS1 and SHIP1, regulators of T cell activation and function 78, 79. Furthermore, miR-155 in conjunction with miR-146a in T cells regulates IFN γ production 79. miRNAs are not only deregulated in effector T cells. A miRNA array comparison between peripheral Tregs and tumor infiltrating Tregs demonstrated deregulation of several miRNAs which may be regulated by Foxp3 80. These studies demonstrate that cancer cells regulate miRNAs in T cells that are important for anti-tumor T cell responses.
Avoidance of immune surveillance has recently been included as a hallmark of cancer. Cancer cells alter expression of transcription factors, surface receptors, soluble chemokines/cytokines and now it appears miRNAs to subvert the immune system. In GBM miR-124 was significantly downregulated compared to normal brain tissue. Downregulation of miR-124 increased Treg infiltration and reduced TIL cytokine production potentially through altered expression of STAT-3, a target of miR-124 81. Further in portal vein tumor thrombosis, TGF- β was shown to downregulate miR-34a, which targets the Treg recruitment chemokine CCL-22 82. Increasing expression of CCL-22 promoted Treg infiltration 82. Thus, demonstrating that deregulation of miRNAs within the cancer cells can alter the TME through manipulation of Tregs. In addition to Tregs, NKT cells may suppress the TILs as well 83. Glioma cells were shown to increase the proportion of IL-10 and IL-6 producing NK cells possibly through the upregulation of miR-92a, which suppressed NKT responsiveness and possibly CD8 T cell function 84. HCC cell lines were shown to alter expression of miR-222 and miR-339 to target ICAM-1 reducing anti-tumor T cell responses 85. In addition to cancer cells altering miRNA expression to avoid the immune system, cancer cells may regulate miRNA expression to use the immune system to promote metastasis. T cells that infiltrated prostate cancer produced FGF11, which upregulated miR-541 in prostate cancer cells. The upregulation of miR-541 enhanced in vitro invasion of prostate cancer potentially through MMP-9 and androgen receptor signaling 86. Collectively these data support the notion that the widespread changes in miRNA expression in cancer cells promote tumorigenesis and determine the behavior of cancer including, immune evasion.
miRNAs, MSCs, and cancer cells
Mesenchymal stem cells (MSCs) are routinely recruited in solid tumors in order to support tumorigenesis through enhancing angiogenesis, metastasis, and immune modulation. Given that miRNAs regulate protein expression, influencing the differentiation and activation of cells, it is not surprising that cancer cells direct changes in miRNA expression in MSCs to reprogram them into tumor promoting cells. MSCs from breast adipose tissue were found to downregulate miR-15b, miR-27b, and miR-29b in response to conditioned media from human breast cancer cell lines. The downregulation of these miRNAs, which promoted MSC migration, was induced by soluble factors 87. Interestingly, miRNA arrays of breast cancer cell lines with differing degrees of metastatic potential indicated that miR-126/126* were significantly downregulated in the more metastatic cell lines 88. Since these miRNAs target SDF-1, a chemokine known to recruit MSCs to the TME, their downregulation promoted breast cancer metastasis by increasing the recruitment of MSCs 88-90. Collectively, these studies demonstrate that, in breast cancer, miRNA changes in both the MSCs and cancer cells foster enhanced recruitment of MSCs to the cancer. Furthermore, MSCs downregulate miR-200b and miR-200c in MCF-7 cells through TGF- β, resulting in the upregulation of ZEB1 and ZEB2, suggesting a role for MSCs in promoting EMT 91. Thus, miRNA reprogramming in both MSCs and breast cancer cells is involved in both the recruitment of MSCs to the TME and the protumorigenic effects of MSCs in the TME. Moreover, the ability of MSCs to reprogram cancer cells is not limited to breast cancer. In HCC, MSCs upregulated miR-155 through secretion of S100A4, a calcium binding protein that has a known role in promoting metastasis, thereby promoting tumorigenesis 92. Based on this collective evidence, miRNA reprogramming of both the cancer cells and MSCs plays a role in the regulation of tumorigenesis.
Direct communication between the tumor micro-environment and cancer cells through transfer of miRNAs
Exosomes (and other microvesicles) provide the means for both the micro-environment and cancer to alter epigenetics within the tumor through the direct transfer of miRNAs 93. This direct transfer introduces miRNAs that subsequently target and functionally alter the cells they enter.
Several studies have highlighted the transfer of miRNAs by exosomes from cancer cells to endothelial cells to promote angiogenesis and metastasis. In a murine breast cancer model, miR-210 was found to be upregulated in cancer derived exosomes, promoting endothelial tubing and branching 94. This transfer of miR-210 through exosomes promoted tumorigenesis through increased endothelial cells in the TME 94. Furthermore, exosomes from a human derived breast cancer cell line upregulated expression of miR-105 in endothelial cells, promoting metastasis to the lungs and brain 95. The exosomes reduced endothelial barriers through miR-105 targeting of ZO-1 95. miR-105 was also found to be elevated in exosomes isolated from the serum of breast cancer patients 95. Similar observations were made in multiple other types of cancers. Through coculturing 5 different tumor cell lines including non-small cell lung cancer, glioblastoma, pancreatic cancer, colorectal cancer and melanoma with endothelial cells and performing a miRNA array on the endothelial cells miR-9, miR-96, miR-182, and miR-183 were found to be consistently upregulated 96. Specifically, cancer mediated secretion of miR-9 in exosomes promoted migration and in vivo endothelial density, which, in turn, promoted tumor growth 96. The upregulation of miR-9 in endothelial cells through exosome delivery promoted angiogenesis through targeting SOCS5, resulting in increased STAT1 and STAT3 activation 96. In glioblastoma, several well-known miRNAs that are highly expressed (let-7a, miR-15b, miR-16, miR19b, miR-21, miR-26a, miR-27a, miR-92, miR-93, miR-320, and miR-20) were found in the microvesicles isolated from patients 97. Furthermore, these microvesicles promoted endothelial tube formation in an RNA dependent manner suggesting that the miRNAs in these microvesicles advance angiogenesis 97. Additionally, it appears that cancers may downregulate miRNAs to limit their incorporation into exosomes that would constrain tumorigenesis. An example of tumor inhibition from miRNA transfer through exosomes is the transfer of miR-1 in microvesicles resulting from the reconstitution of miR-1 in glioblastoma, which acted to reduce tumor growth 98. The upregulation of miR-1 in endothelial cells reduced endothelial cell recruitment and branching through miR-1 targeting of ANXA2 (annexin A2), which is highly expressed in some cancers and promotes tumorigenesis 98, 99. Interestingly, miRNA composition varied between renal cancer stem cells and renal cancer cells. By utilizing a miRNA array of microvesicles from renal cancer patients and sorting cancer stem cells with CD105 expression it was shown that 24 miRNAs were upregulated and 32 miRNAs downregulated in renal cancer stem cells vs. renal non-cancer stem cells 100. Furthermore, these microvesicles promoted endothelial adhesion and invasion in an RNA dependent manner, illustrating the potential for differences in miRNA composition of exosomes within the same tumor from different types of cancer cells 100. The transfer of miRNAs to endothelial cells is not limited to solid cancers. Exosomes from leukemia upregulated expression of the miR-17-92 family in endothelial cells during co-culture and promoted endothelial branch formation and migration 101. Collectively, these studies demonstrate that cancer cells directly alter the miRNA composition of endothelial cells through exosomal transfer of cancer-derived miRNAs to modify the TME. These cancer-mediated changes in miRNA expression in endothelial cells promote angiogenesis and tumor invasion.
miRNA transfer via exosomes from cancer is not restricted to endothelial cells. Fibroblasts, bone marrow derived stromal cells, and macrophages uptake exosomes from cancer, which leads to the upregulation of cancer-derived miRNAs that alter the function of these cells. In a prostate cancer model, “oncosomes” isolated from prostate epithelial cells transformed with Kras infection showed increased levels of miR-1227. The oncosomes were taken up by CAFs, which induced their mobility 102. A miRNA array of exosomes isolated from a melanoma cell line identified 154 miRNAs within the exosomes 103. Bone marrow stromal cells in co-culture took up these exosomes, which increased migration 103. Cancer produced exosomes also regulate the immune system to promote tumorigenesis through TLR signaling. Exosomes from lung cancer cell lines were shown to bind to human TLR8 and to murine Tlr7 which is critical for tumorigenesis in a murine lung cancer model 104. Specifically, miR-21 and miR-29a were secreted in exosomes from lung cancer cell lines and bound to TLR8 104. Additionally, miR-29 was shown to co-localize with exosome markers in human lung cancer samples 104. Therefore, the evidence indicates that cancer cells directly reprogram stromal cells to support cancer progression through miRNA transfer by means of exosomes. While more is known about the secretion of exosomes from cancer cells and their effects on tumorigenesis, stromal cells, including macrophages and MSCs, secrete exosomes that modulate miRNA expression in cancer cells, thereby regulating tumorigenicity. IL-4 activated macrophages, promoting an M2 phenotype, were shown to secrete exosomes containing miR-223. 105. The macrophage derived exosomes upregulated miR-223 during co-culture with a breast cancer cell line, which promoted breast cancer invasiveness 105. Exosomes secreted by MSCs isolated from the bone marrow of patients with multiple myeloma promoted proliferation and metastasis. However, MSCs from healthy patients inhibited tumorigenesis in multiple myeloma. When the miRNA composition of the exosomes from MSCs taken from multiple myeloma patients was compared with that from healthy patients, 16 miRNAs were found to be downregulated and 2 upregulated 106. Interestingly, miR-15, a known tumor suppressing miRNA, was downregulated in exosomes from MSCs isolated from multiple myeloma patients 106. This downregulation of miR-15, along with other miRNA changes within exosomes, resulted in protumorigenic effects of exosomes 106. In a model of gastric cancer, miR-214, miR-221, and miR-222 were shown to be highly upregulated and MSCs were found to transfer miR-221 via exosomes 107. However, MSCs also reduce tumorigenesis through exosomal secretion. In a murine breast cancer model, exosomes from MSCs transferred miR-16 to breast cancer cells reducing angiogenic capacity 108. miR-16 reduced angiogenesis through targeting VEGF, interfering with the ability of breast cancer cells to promote endothelial branching, migrate, and increase microvessel density 108.
Transfer of miRNAs between cells is not limited to microvesicle packaging. It has been demonstrated that miRNAs are transferred through direct cell contact via gap junctions, functionally altering protein expression through miRNA targeting of the mRNA. A recent study that may be relevant to our understanding of the immune micro-environment showed that miRNAs from macrophages were transferred to HCC cells reducing proliferation 109. Specifically, miR-142 and miR-223, which are highly expressed in macrophages, are transferred to HCC cells through direct cell contact mediated by gap junctions 109.
Conclusion
This review highlights the emerging relevance of miRNAs in the communication between cancer cells and the TME. The addition of another level of complexity, represented by the interactions between cancer and stromal cells involving the regulation of miRNAs, presents yet another obstacle to a comprehensive understanding of tumor growth and progression, while also providing new and promising therapeutic targets. While there are many cancer specific deregulations of miRNA in both the cancer cells and cells of the TME there are a few miRNAs that have been reported to be deregulated in TME components in more than one cancer (Figure 5). These could play a more fundamental role in the bidirectional communication between cancer cells and the TME.
Figure 5.

miRNA changes in the components of the TME that occur in more than one cancer. References: [1], 40; [2], 41; [3], 42; [4], 43; [5], 44; [6], 52; [7], 48; [8], 50; [9], 21; [10], 62. CAF, cancer associated fibroblast; CAEs, cancer associated endothelial cells.
The importance of miRNAs and their ability to reprogram cells to regulate function within the TME will be further elucidated, as our understanding of miRNA biology progresses and the complicated network of the TME is revealed. The studies in this review confirm that there is an active communication between cancer cells and the stroma mediated through miRNA changes. However, the extent of this communication has yet to be fully explored. Currently, the majority of the miRNA changes detected in the TME are dictated by the cancer cells. However, there is plenty of evidence that endothelial cells, fibroblasts, and infiltrating immune cells influence tumorigenesis. It is likely that we will find that miRNAs are significant factors in the influence of stromal cells on cancer progression
Clinically altering gene expression and targeting RNAs has been met with limited success, However it is an active area of research. The primary complication with RNA targeting therapies, like most cancer therapies, is specificity for the TME and cancer cells. Understanding how cancer cells communicate with the stroma within the TME, especially through exosomes, may uncover an avenue for specific targeting. The specificity, packaging, and proximity of miRNA containing exosomes within the TME are still unknown. However, these features of exosomes are likely be actively explored, since understanding them could lead to distinct clinical benefits. If we can determine if certain miRNA containing exosome are destined to be taken up by specific cells, we could provide a mechanism and pathway for specific cell targeting, allowing for delivery of chemotherapeutics and gene therapies.
That miRNAs and proteins are not randomly packaged within exosomes is evidenced by the differences in proportions of miRNAs contained with exosomes as compared to the overall cell content 97, 110-115. The miRNA content of exosomes isolated from the peripheral blood from OvCa patients is significantly altered from that of healthy patients, suggesting that the miRNA profile of exosomes might be used for diagnostics 116.
The question remains whether the exosomes that are found in the blood accurately correspond to the exosomes released in the TME. While cancer cells are thought to produce the majority of miRNA containing exosomes, as illustrated in this review, endothelial cells and immune infiltrating cells secrete exosomes in the TME as well. It is possible that once the communication between cancer cells and the stroma is decoded, these miRNA communication pathways may be exploited in strategies for early cancer detection, monitoring therapeutic responses, or, potentially, for targeted cancer therapy.
Acknowledgments
FK was supported by grants T32 CA070085 and F32 CA180677. MEP and EL were supported by an OCRF program project development grant FP049318/PPD/UC.
Abbreviations
- BRCA-1
breast cancer 1, early onset
- VE-cadherin
vascular endothelial cadherin
- MAZ
Myc-associated zinc finger protein
- HGF
hepatocyte growth factor
- IGFBP2
Insulin-like growth factor-binding protein 2
- PITPNC1
phosphatidylinositol transfer protein, cytoplasmic 1
- MERTK
c-mer proto-oncogene tyrosine kinase
- VHL
von Hippel-Lindau tumor suppressor, E3 ubiquitin protein ligase
- LATS2
large tumor suppressor kinase 2
- APOE
apolipoprotein E
- DNAJA4
DnaJ (Hsp40) homolog, subfamily A, member 4
- MMP2
matrix metallopeptidase 2
- VEGF
vascular endothelial growth factor
- HIF1A
hypoxia inducible factor 1, alpha subunit
- FLT1
fms-related tyrosine kinase 1
- IL
interleukin
- VEGFR
vascular endothelial growth factor receptor
- TGFB
transforming growth factor beta
- ETS2
v-ets avian erythroblastosis virus E26 oncogene homolog 2
- MMP9
matrix metallopeptidase 9
- EMILIN2
elastin microfibril interfacer 2
- CCL5
chemokine (C-C motif) ligand 5
- SATB2
Special AT-Rich Sequence-Binding Protein 2
- WNT10B
wingless-type MMTV integration site family, member 10B
- PTEN
phosphatase and tensin homolog
- SOCS1
suppressor of cytokine signaling 1
- SDF1
stromal cell-derived factor 1
- STAT3
signal transducer and activator of transcription 3
- CEBPB
CCAAT/enhancer binding protein (C/EBP), beta
- IRF4
interferon regulatory factor 4
- TME
Tumor Micro-environment
- CAF
cancer associated fibroblasts
- ECM
extracellular matrix
- CXCR4
CXC chemokine receptor 4
- TNF- α
tumor necrosis factor alpha
- NF- κ B
nuclear factor-kappa B
- iNOS
Inducible nitric oxide synthase
- MHC
major histocompatibility complex
- iPS
induced pluripotent stem cells
- HCC
hepatocellular carcinoma
- PDGFRA
platelet-derived growth factor receptor, alpha polypeptide
- EGF
epidermal growth factor
- FGF
fibroblast growth factor
- GAS6
growth arrest-specific 6
- GATA3
GATA binding protein 3
- ITGA6
integrin, alpha 6
- ANGPTL4
angiopoietin-like 4
- LOX
lysyl oxidase
- TCGA
the cancer genome atlas
- EMT
epithelial-mesenchymal transition
- MDSC
myeloid derived suppressor cell
- MAPK
mitogen-activated protein kinase
- AKT
v-akt murine thymoma viral oncogene homolog
- ERK
extracellular-signal-regulated kinases
- OvCa
ovarian cancer
- DNMT1
DNA methyl transferase 1
- uPA
urokinase-type plasminogen activator
- TAM
tumor associated macrophage
- IFN γ
interferon gamma
- Th1
T helper 1
- Fra-1
Fos-related antigen
- AP1
adaptor-related protein complex 1
- CTBP2
C-terminal binding protein 2
- SOX2
SRY (sex determining region Y)-box 2
- MSC
mesenchymal stem cell
- ZEB1
zinc finger E-box binding homeobox 1
- ZEB2
zinc finger E-box binding homeobox 2
- ZO-1
zonula occludens
- ANXA2
annexin A2
- TLR
toll like receptor
- CCL-22
chemokine (C-C motif) ligand 22
- FGF11
fibroblast growth factor 11
- ICAM-1
intercellular adhesion molecule 1
- BLIMP-1
B-lymphocyte-induced maturation protein 1
References
- 1.Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309–322. doi: 10.1016/j.ccr.2012.02.022. [DOI] [PubMed] [Google Scholar]
- 2.Connolly JL, Schnitt SJ, Wang HH, Longtine JA, Dvorak A, Dvorak HF. BC Decker Inc.; Hamilton (ON): 2003. Tumor Structure and Tumor Stroma Generation. vol. http://www.ncbi.nlm.nih.gov/books/NBK13447/ [Google Scholar]
- 3.Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501:346–354. doi: 10.1038/nature12626. [DOI] [PubMed] [Google Scholar]
- 4.Orimo A, Gupta P, Sgroi D, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis throught elevated SDF-1/CXCL 12 secretion. Cell. 2005;121:335–348. doi: 10.1016/j.cell.2005.02.034. [DOI] [PubMed] [Google Scholar]
- 5.Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401. doi: 10.1038/nrc1877. [DOI] [PubMed] [Google Scholar]
- 6.Tuhkanen H, Anttila M, Kosma VM, Heinonen S, Juhola M, Helisalmi S, et al. Frequent gene dosage alterations in stromal cells of epithelial ovarian carcinomas. Int J Cancer. 2006;119:1345–1353. doi: 10.1002/ijc.21785. [DOI] [PubMed] [Google Scholar]
- 7.Qiu W, Hu M, Sridhar A, Opeskin K, Fox S, Shipitsin M, et al. No evidence of clonal somatic genetic alterations in cancer-associated fibroblasts from human breast and ovarian carcinomas. Nat Genet. 2008;40:650–655. doi: 10.1038/ng.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hu M, Yao J, Cai L, Bachman KE, van den Brule F, Velculescu V, et al. Distinct epigenetic changes in the stromal cells of breast cancers. Nat Genet. 2005;37:899–905. doi: 10.1038/ng1596. [DOI] [PubMed] [Google Scholar]
- 9.Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61. doi: 10.1016/j.immuni.2014.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Schickel R, Boyerinas B, Park SM, Peter ME. MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene. 2008;27:5959–5974. doi: 10.1038/onc.2008.274. [DOI] [PubMed] [Google Scholar]
- 11.Di Leva G, Garofalo M, Croce CM. MicroRNAs in cancer. Ann Rev Pathol. 2014;9:287–314. doi: 10.1146/annurev-pathol-012513-104715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dews M, Homayouni A, Yu D, Murphy D, Sevignani C, Wentzel E, et al. Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat Genet. 2006;38:1060–1065. doi: 10.1038/ng1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A. 2004;101:2999–3004. doi: 10.1073/pnas.0307323101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lujambio A, Lowe SW. The microcosmos of cancer. Nature. 2012;482:347–355. doi: 10.1038/nature10888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 16.Lin SL, Chang DC, Lin CH, Ying SY, Leu D, Wu DT. Regulation of somatic cell reprogramming through inducible mir-302 expression. Nucleic Acids Res. 2011;39:1054–1065. doi: 10.1093/nar/gkq850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Barroso-delJesus A, Romero-Lopez C, Lucena-Aguilar G, Melen GJ, Sanchez L, Ligero G, et al. Embryonic stem cell-specific miR302-367 cluster: human gene structure and functional characterization of its core promoter. Mol Cell Biol. 2008;28:6609–6619. doi: 10.1128/MCB.00398-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature. 2011;476:228–231. doi: 10.1038/nature10323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhu K, Pan Q, Zhang X, Kong LQ, Fan J, Dai Z, et al. MiR-146a enhances angiogenic activity of endothelial cells in hepatocellular carcinoma by promoting PDGFRA expression. Carcinogenesis. 2013;34:2071–2079. doi: 10.1093/carcin/bgt160. [DOI] [PubMed] [Google Scholar]
- 20.Wurdinger T, Tannous BA, Saydam O, Skog J, Grau S, Soutschek J, et al. miR-296 regulates growth factor receptor overexpression in angiogenic endothelial cells. Cancer Cell. 2008;14:382–393. doi: 10.1016/j.ccr.2008.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Plummer PN, Freeman R, Taft RJ, Vider J, Sax M, Umer BA, et al. MicroRNAs regulate tumor angiogenesis modulated by endothelial progenitor cells. Cancer Res. 2013;73:341–352. doi: 10.1158/0008-5472.CAN-12-0271. [DOI] [PubMed] [Google Scholar]
- 22.Muramatsu F, Kidoya H, Naito H, Sakimoto S, Takakura N. microRNA-125b inhibits tube formation of blood vessels through translational suppression of VE-cadherin. Oncogene. 2013;32:414–421. doi: 10.1038/onc.2012.68. [DOI] [PubMed] [Google Scholar]
- 23.Smits M, Wurdinger T, van het Hof B, Drexhage JA, Geerts D, Wesseling P, et al. Myc-associated zinc finger protein (MAZ) is regulated by miR-125b and mediates VEGF-induced angiogenesis in glioblastoma. FASEB J. 2012;26:2639–2647. doi: 10.1096/fj.11-202820. [DOI] [PubMed] [Google Scholar]
- 24.Fang L, Du WW, Yang W, Rutnam ZJ, Peng C, Li H, et al. MiR-93 enhances angiogenesis and metastasis by targeting LATS2. Cell Cycle. 2012;11:4352–4365. doi: 10.4161/cc.22670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kong W, He L, Richards EJ, Challa S, Xu CX, Permuth-Wey J, et al. Upregulation of miRNA-155 promotes tumour angiogenesis by targeting VHL and is associated with poor prognosis and triple-negative breast cancer. Oncogene. 2014;33:679–689. doi: 10.1038/onc.2012.636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Fang JH, Zhou HC, Zeng C, Yang J, Liu Y, Huang X, et al. MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology. 2011;54:1729–1740. doi: 10.1002/hep.24577. [DOI] [PubMed] [Google Scholar]
- 27.Png KJ, Halberg N, Yoshida M, Tavazoie SF. A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells. Nature. 2012;481:190–194. doi: 10.1038/nature10661. [DOI] [PubMed] [Google Scholar]
- 28.Nilsson S, Moller C, Jirstrom K, Lee A, Busch S, Lamb R, et al. Downregulation of miR-92a is associated with aggressive breast cancer features and increased tumour macrophage infiltration. PLoS One. 2012;7:e36051. doi: 10.1371/journal.pone.0036051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chou J, Lin JH, Brenot A, Kim JW, Provot S, Werb Z. GATA3 suppresses metastasis and modulates the tumour microenvironment by regulating microRNA-29b expression. Nat Cell Biol. 2013;15:201–213. doi: 10.1038/ncb2672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang R, Zhao N, Li S, Fang JH, Chen MX, Yang J, et al. MicroRNA-195 suppresses angiogenesis and metastasis of hepatocellular carcinoma by inhibiting the expression of VEGF, VAV2, and CDC42. Hepatology. 2013;58:642–653. doi: 10.1002/hep.26373. [DOI] [PubMed] [Google Scholar]
- 31.Shih TC, Tien YJ, Wen CJ, Yeh TS, Yu MC, Huang CH, et al. MicroRNA-214 downregulation contributes to tumor angiogenesis by inducing secretion of the hepatoma-derived growth factor in human hepatoma. J Hepatol. 2012;57:584–591. doi: 10.1016/j.jhep.2012.04.031. [DOI] [PubMed] [Google Scholar]
- 32.Cha ST, Chen PS, Johansson G, Chu CY, Wang MY, Jeng YM, et al. MicroRNA-519c suppresses hypoxia-inducible factor-1alpha expression and tumor angiogenesis. Cancer Res. 2010;70:2675–2685. doi: 10.1158/0008-5472.CAN-09-2448. [DOI] [PubMed] [Google Scholar]
- 33.Jusufovic E, Rijavec M, Keser D, Korosec P, Sodja E, Iljazovic E, et al. let-7b and miR-126 are down-regulated in tumor tissue and correlate with microvessel density and survival outcomes in non--small--cell lung cancer. PLoS One. 2012;7:e45577. doi: 10.1371/journal.pone.0045577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Pecot CV, Rupaimoole R, Yang D, Akbani R, Ivan C, Lu C, et al. Tumour angiogenesis regulation by the miR-200 family. Nature Commun. 2013;4:2427. doi: 10.1038/ncomms3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Vecchione A, Belletti B, Lovat F, Volinia S, Chiappetta G, Giglio S, et al. A microRNA signature defines chemoresistance in ovarian cancer through modulation of angiogenesis. Proc Natl Acad Sci U S A. 2013;110:9845–9850. doi: 10.1073/pnas.1305472110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Pencheva N, Tran H, Buss C, Huh D, Drobnjak M, Busam K, et al. Convergent multi-miRNA targeting of ApoE drives LRP1/LRP8-dependent melanoma metastasis and angiogenesis. Cell. 2012;151:1068–1082. doi: 10.1016/j.cell.2012.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Almog N, Ma L, Schwager C, Brinkmann BG, Beheshti A, Vajkoczy P, et al. Consensus micro RNAs governing the switch of dormant tumors to the fast-growing angiogenic phenotype. PLoS One. 2012;7:e44001. doi: 10.1371/journal.pone.0044001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fang L, Deng Z, Shatseva T, Yang J, Peng C, Du WW, et al. MicroRNA miR-93 promotes tumor growth and angiogenesis by targeting integrin-beta8. Oncogene. 2011;30:806–821. doi: 10.1038/onc.2010.465. [DOI] [PubMed] [Google Scholar]
- 39.Roybal JD, Zang Y, Ahn YH, Yang Y, Gibbons DL, Baird BN, et al. miR-200 Inhibits lung adenocarcinoma cell invasion and metastasis by targeting Flt1/VEGFR1. Mol Cancer Res. 2011;9:25–35. doi: 10.1158/1541-7786.MCR-10-0497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bullock MD, Pickard KM, Nielsen BS, Sayan AE, Jenei V, Mellone M, et al. Pleiotropic actions of miR-21 highlight the critical role of deregulated stromal microRNAs during colorectal cancer progression. Cell Death Dis. 2013;4:e684. doi: 10.1038/cddis.2013.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yamamichi N, Shimomura R, Inada K, Sakurai K, Haraguchi T, Ozaki Y, et al. Locked nucleic acid in situ hybridization analysis of miR-21 expression during colorectal cancer development. Clin Cancer Res. 2009;15:4009–4016. doi: 10.1158/1078-0432.CCR-08-3257. [DOI] [PubMed] [Google Scholar]
- 42.Nielsen BS, Jorgensen S, Fog JU, Sokilde R, Christensen IJ, Hansen U, et al. High levels of microRNA-21 in the stroma of colorectal cancers predict short disease-free survival in stage II colon cancer patients. Clin Exp Metastasis. 2011;28:27–38. doi: 10.1007/s10585-010-9355-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nouraee N, Van Roosbroeck K, Vasei M, Semnani S, Samaei NM, Naghshvar F, et al. Expression, tissue distribution and function of miR-21 in esophageal squamous cell carcinoma. PLoS One. 2013;8:e73009. doi: 10.1371/journal.pone.0073009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kadera BE, Li L, Toste PA, Wu N, Adams C, Dawson DW, et al. MicroRNA-21 in pancreatic ductal adenocarcinoma tumor-associated fibroblasts promotes metastasis. PLoS One. 2013;8:e71978. doi: 10.1371/journal.pone.0071978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yao Q, Cao S, Li C, Mengesha A, Kong B, Wei M. Micro-RNA-21 regulates TGF-beta-induced myofibroblast differentiation by targeting PDCD4 in tumor-stroma interaction. Int J Cancer. 2011;128:1783–1792. doi: 10.1002/ijc.25506. [DOI] [PubMed] [Google Scholar]
- 46.Naito Y, Sakamoto N, Oue N, Yashiro M, Sentani K, Yanagihara K, et al. MicroRNA-143 regulates collagen type III expression in stromal fibroblasts of scirrhous type gastric cancer. Cancer Sci. 2014;105:228–235. doi: 10.1111/cas.12329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gatto G, Rossi A, Rossi D, Kroening S, Bonatti S, Mallardo M. Epstein-Barr virus latent membrane protein 1 trans-activates miR-155 transcription through the NF-kappaB pathway. Nucleic Acids Res. 2008;36:6608–6619. doi: 10.1093/nar/gkn666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mitra AK, Zillhardt M, Hua Y, Tiwari P, Murmann AE, Peter ME, et al. MicroRNAs reprogram normal fibroblasts into cancer-associated fibroblasts in ovarian cancer. Cancer Discov. 2012;2:1100–1108. doi: 10.1158/2159-8290.CD-12-0206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhao L, Sun Y, Hou Y, Peng Q, Wang L, Luo H, et al. MiRNA expression analysis of cancer-associated fibroblasts and normal fibroblasts in breast cancer. Int J Biochem Cell Biol. 2012;44:2051–2059. doi: 10.1016/j.biocel.2012.08.005. [DOI] [PubMed] [Google Scholar]
- 50.Aprelikova O, Yu X, Palla J, Wei BR, John S, Yi M, et al. The role of miR-31 and its target gene SATB2 in cancer-associated fibroblasts. Cell Cycle. 2010;9:4387–4398. doi: 10.4161/cc.9.21.13674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Aprelikova O, Palla J, Hibler B, Yu X, Greer YE, Yi M, et al. Silencing of miR-148a in cancer-associated fibroblasts results in WNT10B-mediated stimulation of tumor cell motility. Oncogene. 2013;32:3246–3253. doi: 10.1038/onc.2012.351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Verghese ET, Drury R, Green CA, Holliday DL, Lu X, Nash C, et al. MiR-26b is down-regulated in carcinoma-associated fibroblasts from ER-positive breast cancers leading to enhanced cell migration and invasion. J Pathol. 2013;231:388–399. doi: 10.1002/path.4248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bronisz A, Godlewski J, Wallace JA, Merchant AS, Nowicki MO, Mathsyaraja H, et al. Reprogramming of the tumour microenvironment by stromal PTEN-regulated miR-320. Nat Cell Biol. 2012;14:159–167. doi: 10.1038/ncb2396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Trimboli AJ, Cantemir-Stone CZ, Li F, Wallace JA, Merchant A, Creasap N, et al. Pten in stromal fibroblasts suppresses mammary epithelial tumours. Nature. 2009;461:1084–1091. doi: 10.1038/nature08486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mongiat M, Marastoni S, Ligresti G, Lorenzon E, Schiappacassi M, Perris R, et al. The extracellular matrix glycoprotein elastin microfibril interface located protein 2: a dual role in the tumor microenvironment. Neoplasia. 2010;12:294–304. doi: 10.1593/neo.91930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sugiyama K, Kajiyama H, Shibata K, Yuan H, Kikkawa F, Senga T. Expression of the miR200 family of microRNAs in mesothelial cells suppresses the dissemination of ovarian cancer cells. Mol Cancer Ther. 2014;13:2081–2091. doi: 10.1158/1535-7163.MCT-14-0135. [DOI] [PubMed] [Google Scholar]
- 57.Mitra AK, Chiang CY, Tiwari P, Tomar S, Watters KM, Peter ME, et al. Microenvironment induced downregulation of miR-193b drives ovarian cancer metastasis. Oncogene. 2014 doi: 10.1038/onc.2015.43. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.He M, Xu Z, Ding T, Kuang DM, Zheng L. MicroRNA-155 regulates inflammatory cytokine production in tumor-associated macrophages via targeting C/EBPbeta. Cell Mol Immunol. 2009;6:343–352. doi: 10.1038/cmi.2009.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zonari E, Pucci F, Saini M, Mazzieri R, Politi LS, Gentner B, et al. A role for miR-155 in enabling tumor-infiltrating innate immune cells to mount effective antitumor responses in mice. Blood. 2013;122:243–252. doi: 10.1182/blood-2012-08-449306. [DOI] [PubMed] [Google Scholar]
- 60.Cubillos-Ruiz JR, Baird JR, Tesone AJ, Rutkowski MR, Scarlett UK, Camposeco-Jacobs AL, et al. Reprogramming tumor-associated dendritic cells in vivo using miRNA mimetics triggers protective immunity against ovarian cancer. Cancer Res. 2012;72:1683–1693. doi: 10.1158/0008-5472.CAN-11-3160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cai X, Yin Y, Li N, Zhu D, Zhang J, Zhang CY, et al. Re-polarization of tumor-associated macrophages to pro-inflammatory M1 macrophages by microRNA-155. J Mol Cell Biol. 2012;4:341–343. doi: 10.1093/jmcb/mjs044. [DOI] [PubMed] [Google Scholar]
- 62.Sonda N, Simonato F, Peranzoni E, Cali B, Bortoluzzi S, Bisognin A, et al. miR-142-3p prevents macrophage differentiation during cancer-induced myelopoiesis. Immunity. 2013;38:1236–1249. doi: 10.1016/j.immuni.2013.06.004. [DOI] [PubMed] [Google Scholar]
- 63.Chaudhuri AA, So AY, Sinha N, Gibson WS, Taganov KD, O'Connell RM, et al. MicroRNA-125b potentiates macrophage activation. J Immunol. 2011;187:5062–5068. doi: 10.4049/jimmunol.1102001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yang J, Zhang Z, Chen C, Liu Y, Si Q, Chuang TH, et al. MicroRNA-19a-3p inhibits breast cancer progression and metastasis by inducing macrophage polarization through downregulated expression of Fra-1 proto-oncogene. Oncogene. 2013;33:3014–3023. doi: 10.1038/onc.2013.258. [DOI] [PubMed] [Google Scholar]
- 65.El Chartouni C, Schwarzfischer L, Rehli M. Interleukin-4 induced interferon regulatory factor (Irf) 4 participates in the regulation of alternative macrophage priming. Immunobiology. 2010;215:821–825. doi: 10.1016/j.imbio.2010.05.031. [DOI] [PubMed] [Google Scholar]
- 66.Wang Q, Ni H, Lan L, Wei X, Xiang R, Wang Y. Fra-1 protooncogene regulates IL-6 expression in macrophages and promotes the generation of M2d macrophages. Cell Res. 2010;20:701–712. doi: 10.1038/cr.2010.52. [DOI] [PubMed] [Google Scholar]
- 67.Du J, Wang J, Tan G, Cai Z, Zhang L, Tang B, et al. Aberrant elevated microRNA-146a in dendritic cells (DC) induced by human pancreatic cancer cell line BxPC-3-conditioned medium inhibits DC maturation and activation. Med Oncol. 2012;29:2814–2823. doi: 10.1007/s12032-012-0175-2. [DOI] [PubMed] [Google Scholar]
- 68.Chang SH, Lu YC, Li X, Hsieh WY, Xiong Y, Ghosh M, et al. Antagonistic function of the RNA-binding protein HuR and miR-200b in post-transcriptional regulation of vascular endothelial growth factor-A expression and angiogenesis. J Biol Chem. 2013;288:4908–4921. doi: 10.1074/jbc.M112.423871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Squadrito ML, Pucci F, Magri L, Moi D, Gilfillan GD, Ranghetti A, et al. miR-511-3p modulates genetic programs of tumor-associated macrophages. Cell Rep. 2012;1:141–154. doi: 10.1016/j.celrep.2011.12.005. [DOI] [PubMed] [Google Scholar]
- 70.Liu Y, Lai L, Chen Q, Song Y, Xu S, Ma F, et al. MicroRNA-494 is required for the accumulation and functions of tumor-expanded myeloid-derived suppressor cells via targeting of PTEN. J Immunol. 2012;188:5500–5510. doi: 10.4049/jimmunol.1103505. [DOI] [PubMed] [Google Scholar]
- 71.Liu Q, Zhang M, Jiang X, Zhang Z, Dai L, Min S, et al. miR-223 suppresses differentiation of tumor-induced CD11b(+) Gr1(+) myeloid-derived suppressor cells from bone marrow cells. Int J Cancer. 2011;129:2662–2673. doi: 10.1002/ijc.25921. [DOI] [PubMed] [Google Scholar]
- 72.Zhang M, Liu Q, Mi S, Liang X, Zhang Z, Su X, et al. Both miR-17-5p and miR-20a alleviate suppressive potential of myeloid-derived suppressor cells by modulating STAT3 expression. J Immunol. 2011;186:4716–4724. doi: 10.4049/jimmunol.1002989. [DOI] [PubMed] [Google Scholar]
- 73.Kong D, Piao YS, Yamashita S, Oshima H, Oguma K, Fushida S, et al. Inflammation-induced repression of tumor suppressor miR-7 in gastric tumor cells. Oncogene. 2012;31:3949–3960. doi: 10.1038/onc.2011.558. [DOI] [PubMed] [Google Scholar]
- 74.Subramani A, Alsidawi S, Jagannathan S, Sumita K, Sasaki AT, Aronow B, et al. The brain microenvironment negatively regulates miRNA-768-3p to promote K-ras expression and lung cancer metastasis. Sci Rep. 2013;3:2392. doi: 10.1038/srep02392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Cui TX, Kryczek I, Zhao L, Zhao E, Kuick R, Roh MH, et al. Myeloid-derived suppressor cells enhance stemness of cancer cells by inducing microRNA101 and suppressing the corepressor CtBP2. Immunity. 2013;39:611–621. doi: 10.1016/j.immuni.2013.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lwin T, Lin J, Choi YS, Zhang X, Moscinski LC, Wright KL, et al. Follicular dendritic cell-dependent drug resistance of non-Hodgkin lymphoma involves cell adhesion-mediated Bim down-regulation through induction of microRNA-181a. Blood. 2010;116:5228–5236. doi: 10.1182/blood-2010-03-275925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sasaki K, Kohanbash G, Hoji A, Ueda R, McDonald HA, Reinhart TA, et al. miR-17-92 expression in differentiated T cells - implications for cancer immunotherapy. J Transl Med. 2010;8:17. doi: 10.1186/1479-5876-8-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Dudda JC, Salaun B, Ji Y, Palmer DC, Monnot GC, Merck E, et al. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity. 2013;38:742–753. doi: 10.1016/j.immuni.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huffaker TB, Hu R, Runtsch MC, Bake E, Chen X, Zhao J, et al. Epistasis between microRNAs 155 and 146a during T cell-mediated antitumor immunity. Cell Rep. 2012;2:1697–1709. doi: 10.1016/j.celrep.2012.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen L, Ma H, Hu H, Gao L, Wang X, Ma J, et al. Special role of Foxp3 for the specifically altered microRNAs in Regulatory T cells of HCC patients. BMC Cancer. 2014;14:489. doi: 10.1186/1471-2407-14-489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, et al. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res. 2013;73:3913–3926. doi: 10.1158/0008-5472.CAN-12-4318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yang P, Li QJ, Feng Y, Zhang Y, Markowitz GJ, Ning S, et al. TGF-beta-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell. 2012;22:291–303. doi: 10.1016/j.ccr.2012.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology. 2013;138:105–115. doi: 10.1111/imm.12036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Tang B, Wu W, Wei X, Li Y, Ren G, Fan W. Activation of glioma cells generates immune tolerant NKT cells. J Biol Chem. 2014;289:34595–34600. doi: 10.1074/jbc.M114.614503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ueda R, Kohanbash G, Sasaki K, Fujita M, Zhu X, Kastenhuber ER, et al. Dicer-regulated microRNAs 222 and 339 promote resistance of cancer cells to cytotoxic T-lymphocytes by down-regulation of ICAM-1. Proc Natl Acad Sci U S A. 2009;106:10746–10751. doi: 10.1073/pnas.0811817106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hu S, Li L, Yeh S, Cui Y, Li X, Chang HC, et al. Infiltrating T cells promote prostate cancer metastasis via modulation of FGF11-->miRNA-541-->androgen receptor (AR)-->MMP9 signaling. Mol Oncol. 2015;9:44–57. doi: 10.1016/j.molonc.2014.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Senst C, Nazari-Shafti T, Kruger S, Honer Zu Bentrup K, Dupin CL, Chaffin AE, et al. Prospective dual role of mesenchymal stem cells in breast tumor microenvironment. Breast Cancer Res Treat. 2013;137:69–79. doi: 10.1007/s10549-012-2321-0. [DOI] [PubMed] [Google Scholar]
- 88.Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15:284–294. doi: 10.1038/ncb2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997;185:111–120. doi: 10.1084/jem.185.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ponte AL, Marais E, Gallay N, Langonne A, Delorme B, Herault O, et al. The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities. Stem Cells. 2007;25:1737–1745. doi: 10.1634/stemcells.2007-0054. [DOI] [PubMed] [Google Scholar]
- 91.Xu Q, Wang L, Li H, Han Q, Li J, Qu X, et al. Mesenchymal stem cells play a potential role in regulating the establishment and maintenance of epithelial-mesenchymal transition in MCF7 human breast cancer cells by paracrine and induced autocrine TGF-beta. Int J Oncol. 2012;41:959–968. doi: 10.3892/ijo.2012.1541. [DOI] [PubMed] [Google Scholar]
- 92.Yan XL, Jia YL, Chen L, Zeng Q, Zhou JN, Fu CJ, et al. Hepatocellular carcinoma-associated mesenchymal stem cells promote hepatocarcinoma progression: role of the S100A4-miR155-SOCS1-MMP9 axis. Hepatology. 2013;57:2274–2286. doi: 10.1002/hep.26257. [DOI] [PubMed] [Google Scholar]
- 93.Kosaka N, Yoshioka Y, Hagiwara K, Tominaga N, Katsuda T, Ochiya T. Trash or Treasure: extracellular microRNAs and cell-to-cell communication. Front Genet. 2013;4:173. doi: 10.3389/fgene.2013.00173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kosaka N, Iguchi H, Hagiwara K, Yoshioka Y, Takeshita F, Ochiya T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J Biol Chem. 2013;288:10849–10859. doi: 10.1074/jbc.M112.446831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, et al. Cancer-Secreted miR-105 Destroys Vascular Endothelial Barriers to Promote Metastasis. Cancer Cell. 2014;25:501–515. doi: 10.1016/j.ccr.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, et al. Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012;31:3513–3523. doi: 10.1038/emboj.2012.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature Cell Biol. 2008;10:1470–1476. doi: 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bronisz A, Wang Y, Nowicki MO, Peruzzi P, Ansari KI, Ogawa D, et al. Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 2014;74:738–750. doi: 10.1158/0008-5472.CAN-13-2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zhai H, Acharya S, Gravanis I, Mehmood S, Seidman RJ, Shroyer KR, et al. Annexin A2 promotes glioma cell invasion and tumor progression. J Neurosci. 2011;31:14346–14360. doi: 10.1523/JNEUROSCI.3299-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71:5346–5356. doi: 10.1158/0008-5472.CAN-11-0241. [DOI] [PubMed] [Google Scholar]
- 101.Umezu T, Ohyashiki K, Kuroda M, Ohyashiki JH. Leukemia cell to endothelial cell communication via exosomal miRNAs. Oncogene. 2013;32:2747–2755. doi: 10.1038/onc.2012.295. [DOI] [PubMed] [Google Scholar]
- 102.Morello M, Minciacchi VR, de Candia P, Yang J, Posadas E, Kim H, et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle. 2013;12:3526–3536. doi: 10.4161/cc.26539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Rappa G, Mercapide J, Anzanello F, Pope RM, Lorico A. Biochemical and biological characterization of exosomes containing prominin-1/CD133. Mol Cancer. 2013;12:62. doi: 10.1186/1476-4598-12-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci U S A. 2012;109:E2110–2116. doi: 10.1073/pnas.1209414109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yang M, Chen J, Su F, Yu B, Lin L, Liu Y, et al. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol Cancer. 2011;10:117. doi: 10.1186/1476-4598-10-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 2013;123:1542–1555. doi: 10.1172/JCI66517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wang M, Zhao C, Shi H, Zhang B, Zhang L, Zhang X, et al. Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 2014;110:1199–1210. doi: 10.1038/bjc.2014.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lee JK, Park SR, Jung BK, Jeon YK, Lee YS, Kim MK, et al. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013;8:e84256. doi: 10.1371/journal.pone.0084256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Aucher A, Rudnicka D, Davis DM. 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]
- 110.Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol. 2009;11:1143–1149. doi: 10.1038/ncb1929. [DOI] [PubMed] [Google Scholar]
- 111.Guduric-Fuchs J, O'Connor A, Camp B, O'Neill CL, Medina RJ, Simpson DA. Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genomics. 2012;13:357. doi: 10.1186/1471-2164-13-357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Nolte-'t Hoen EN, Buermans HP, Waasdorp M, Stoorvogel W, Wauben MH, t Hoen PA. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012;40:9272–9285. doi: 10.1093/nar/gks658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21:575–581. doi: 10.1016/j.ceb.2009.03.007. [DOI] [PubMed] [Google Scholar]
- 114.Squadrito ML, Baer C, Burdet F, Maderna C, Gilfillan GD, Lyle R, et al. Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells. Cell Rep. 2014;8:1432–1446. doi: 10.1016/j.celrep.2014.07.035. [DOI] [PubMed] [Google Scholar]
- 115.Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 116.Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110:13–21. doi: 10.1016/j.ygyno.2008.04.033. [DOI] [PubMed] [Google Scholar]
- 117.Musumeci M, Coppola V, Addario A, Patrizii M, Maugeri-Sacca M, Memeo L, et al. Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene. 2011;30:4231–4242. doi: 10.1038/onc.2011.140. [DOI] [PubMed] [Google Scholar]
