Skip to main content
Erschienen in: Molecular Cancer 1/2021

Open Access 01.12.2021 | Review

Non-coding RNAs regulation of macrophage polarization in cancer

verfasst von: Swati Mohapatra, Carlotta Pioppini, Bulent Ozpolat, George A. Calin

Erschienen in: Molecular Cancer | Ausgabe 1/2021

Abstract

Noncoding RNA (ncRNA) transcripts that did not code proteins but regulate their functions were extensively studied for the last two decades and the plethora of discoveries have instigated scientists to investigate their dynamic roles in several diseases especially in cancer. However, there is much more to learn about the role of ncRNAs as drivers of malignant cell evolution in relation to macrophage polarization in the tumor microenvironment. At the initial stage of tumor development, macrophages have an important role in directing Go/No-go decisions to the promotion of tumor growth, immunosuppression, and angiogenesis. Tumor-associated macrophages behave differently as they are predominantly induced to be polarized into M2, a pro-tumorigenic type when recruited with the tumor tissue and thereby favoring the tumorigenesis. Polarization of macrophages into M1 or M2 subtypes plays a vital role in regulating tumor progression, metastasis, and clinical outcome, highlighting the importance of studying the factors driving this process. A substantial number of studies have demonstrated that ncRNAs are involved in the macrophage polarization based on their ability to drive M1 or M2 polarization and in this review we have described their functions and categorized them into oncogenes, tumor suppressors, Juggling tumor suppressors, and Juggling oncogenes.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BC
Breast cancer
Cav-1
Caveolin-1
CCAT1
Colon cancer-associated transcript-1
COX-2
Cyclooxygenase-2
C/EBPβ
CCAAT/enhancer-binding protein β
CPEB4
Cytoplasmic polyadenylation element binding protein 4
CRC
Colorectal cancer
CSC
Cancer stem cells
EC
Endometrial cancer
ECM
Extra cellular matrix
EMT
Epithelial-mesenchymal transition
eRNAs
Enhancer-like ncRNAs
FGF-2
Fibroblast growth factor-2
FOXO1
Forkhead box protein O1
Fra-1
Fos-related antigen-1
GAS5
Growth arrest specific 5
GATA3
GATA Binding protein 3
GC
Gastric cancer
HCC
Hepatocellular carcinoma
HIF
Hypoxia-inducible factor
HMGA2
High Mobility Group AT-Hook 2
HDAC-11
Histone deacetylase 11
HNC
Head and neck cancer
IC
Immune Complex
IFN
Interferon
IKB
Inhibitor NF-Kappa B
IL
Interleukins
INOS
Inducible nitric oxide synthase
IRF
Interferon regulatory factor
JAK2
Janus Kinase 2 protein
JMJD3
Jumonji domain-containing protein D3
KLF
Kruppel-like factor
LC
Lung cancer
LIF
Leukemia inhibitory factor
LINC
Long intergenic ncRNAs
LNA
Locked nucleic acid
lncRNA
Long noncoding RNA
LPS
Lipopolysaccharide
MALAT1
Metastasis-Associated Lung Adenocarcinoma Transcript 1
MCP1
Monocyte chemoattractant protein-1
M-CSF
Macrophage colony-stimulating factor
MCT-1
Multiple Copies in T-cell Malignancy 1
MDSC
Myeloid-derived suppressor cells
MHC
Major histocompatibility complex
miRNA
MicroRNA
miRNAs
MicroRNAs
MM
Multiple myeloma
MMP
Metalloproteinase
MRC-1
Mannose receptor C-type 1
MSN-c
Met- c-Met via MSN-mediated protein stabilization
NATs
Natural antisense transcripts
ncRNA
Noncoding RNA
NEAT1
Nuclear paraspeckle assembly transcript 1
NECAB3
N-terminal EF-hand calcium- binding protein 3
NF-kB
Nuclear factor kappa-light-chain-enhancer of activated B cells
NOTCH2
Neurogenic locus notch homolog protein 2
NSCLC
Non-small cell lung cancer
OC
Ovarian cancer
OS
Osteosarcoma
OSCC
Oral squamous cell carcinoma
PACER
p50-associated COX-2 extragenic RNA
PANC
Pancreatic cancer
PC
Prostate cancer
PDCD4
Programmed cell death protein 4
PIK3IP1
Phosphoinositide-3-kinase interacting protein 1
piRNA
Piwi-interacting RNA
PKC
Protein kinase C
PPAR
Peroxisome proliferator-activated receptor
PTBP3
Polypyrimidine tract binding protein 3
RPPH1
Ribonuclease P RNA component H1
rRNA
Ribosomal RNA
siRNAs
Small interfering RNA
snoRNA
Small nucleolar RNA
snRNA
Small nuclear RNA
SOCS1
Suppressor of cytokine signaling 1 protein
STAT
Signal transducer and activator of transcription protein
TAMs
Tumor-associated macrophages
TCF-4
T-cell specificTranscription factor 4
TERF2IP
Telomeric repeat-binding factor2-interacting protein 1
TH1
Type I T helper cells
TME
Tumor microenvironment
TGFβ
Tumor growth factor-beta
TNFα
Tumor necrosis factor alpha
T-PYKs
Transcribed pyknons
TRIB1
Tribbles homolog 1
tRNAs
Transfer RNAs
TUBB3
Tubulin Beta 3 Class III protein
T-UCRs
Transcribed ultra-conserved regions
VEGF-A
Vascular endothelial growth factor-A
XIST
X-Inactive Specific Transcript

Background

The high throughput state-of-art technologies have made the transcriptome more easily accessible, which allowed the discovery of the RNA transcripts that do not encode for proteins termed “non-coding RNAs (ncRNAs)”, which constitute up to 98% of the transcribed human genome [1]. The complex interactions between various types of ncRNAs and other DNA/RNAs, proteins, and lipids [2] can explain their role as regulators in many cellular pathways including immunological processes. Furthermore, they are abnormally regulated in several dreadful diseases including many cancer types [3]. The broad classification of ncRNAs is based on their size and divided into two major groups: small ncRNA (< 200 bp) and long ncRNAs (lncRNAs) (> 200 bp) [4]. The small ncRNAs class includes mainly microRNAs (miRNAs), small nucleolar RNA (snoRNA), small interfering RNA (siRNAs), small nuclear RNA (snRNA), transfer RNAs (tRNAs), and Piwi-interacting RNAs (piRNAs) [5]. While small ncRNAs like miRNAs are highly studied for almost two decades, investigation on lncRNAs realm started in the last decade. Depending on their location in the genome, lncRNAs can be classified into multiple categories, such as natural antisense transcripts (NATs) overlapping in antisense usually protein coding genes, enhancer-like ncRNAs (eRNAs) from the enhancer regions, transcribed ultra-conserved regions (T-UCRs) from the ultra-conserved elements, transcribed pyknons (T-PYKs from human-specific regions) [6] and long intergenic ncRNAs (LINC) from regions between protein-coding genes [79]. lncRNAs have important roles as gene regulators in post-transcriptional and transcriptional, epigenetic chromatin remodeling, protein functioning, small RNAs’ maturation, and other significant biological processes including macrophage polarization occurring from monocytes [10, 11]. Monocytes are precursor cells of macrophages produced from progenitor cells in the bone marrow called monoblasts that are produced from hematopoietic stem cells [12]. Monocytes are released into the bloodstream and migrate to the tissues at the periphery, thereby undergoing differentiation into macrophages and dendritic cells depending on their unveiling to the microbial compounds, inflammation-favoring cytokines, and growth factors [10].
In this review, we summarize recent findings regarding ncRNAs (miRNAs and lncRNAs) that mechanistically regulate the differentiation process of macrophages, based on their ability to drive M1 or M2 polarization in different cancers and broadly categorize them into oncogenes, tumor suppressors, Juggling tumor suppressors, and Juggling oncogenes (Fig. 2).

Macrophage polarization is an event of plasticity

Macrophages differentiate into specific phenotypes in response to various microenvironmental stimuli and have specific biological functions [11]. Human peripheral blood monocytes (HPBM) are classified on the basis of surface markers CD14 and CD16 into three distinct subgroups: classical monocytes with high CD14 and nil CD16 expression; non-classical monocytes having high CD16 but with comparatively lower CD14 expression; and intermediate type having higher CD14 and lower CD16 expression [13]. Depending on the activation stage and functional status of the macrophages, they have been classified into M1 and M2 types (Table 1) [14]. In fact, all the three subsets mentioned above if incubated with granulocyte macrophage-colony stimulating factor (GM-CSF) induces M1 type polarization while macrophage colony-stimulating factor (M-CSF) induces M2 type polarization [15]. M1 macrophages are the predominant phenotype in normal immunological responses and involved in TH1 (type I T helper cells) response against different pathogens and produce pro-inflammatory cytokines with tumor-cell and microbe-killing activities [15, 16]. IFN-γ or lipopolysaccharide (LPS) are involved in the classical activation of M1 and in the production of proinflammatory cytokines that leads to phagocytosis of microbes, thereby initiating an immune response while IL-4, IL-13, or IL-10 cytokines help in activating M2 macrophages [17]. M2s induce immunosuppression, tumorigenesis, elimination of parasites, and are involved in wound repair [17]. The oncogene MCT-1 (Multiple Copies in T-cell Malignancy 1) stimulates the secretion of IL-6 that enhances the polarization of THP-1 monocytes into the M2 subtype [18]. The polarization of macrophages can occur at any point during the inflammatory processes and is triggered by various factors including epigenetic, tissue microenvironment; and extrinsic factors like microbial byproducts and inflammatory cytokines [11]. M2 macrophages can be divided into three subgroups namely M2a, M2b, M2c, and M2d (Fig. 1) [1922]. The M2a subtype differentiation is triggered by mast cells’ secretion of IL-4 and IL-13, Th2 lymphocytes, and basophils [22]. Several mediators such as pro-inflammatory molecules (tumor necrosis factor-alpha (TNF-α), IFN-γ, IL-6, IL-12, IL-1β) and superoxide anions are negatively regulated by IL-4 and IL-13 cytokines, and M2a cells are reported to be involved in tissue repair and wound healing [22]. M2b subtype has anti-inflammatory and immune regulatory roles. In fact, their differentiation is mediated by the interaction between immune complexes (IC) with LPS or IL-1R ligand that reduces the synthesis of IL-12 and increases the production of IL-10 [23]. M2c subtype differentiation is promoted by TGF-β, IL-10, and glucocorticoids. M2c plays an important role in immunosuppression and tissue remodeling. The last subtype, M2d, is activated by the presence of tumor-associated factors. They promote tumor growth and angiogenesis and hence, this subtype is a major constituent of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) [24]. TME is composed of malignant cells, non-malignant cells, such as immune and stromal cells (i.e, cancer stem cells (CSC), cancer-associated fibroblasts, cancer-associated adipocytes), endothelial cells, fibroblasts, neurons [25, 26], and non-cellular components include extracellular matrix (ECM) proteins, growth factors, cytokines, and metabolites that promotes tumor proliferation, angiogenesis, and metastasis [2729]. The exact composition of TME differs between different types of cancers. TAMs play an important role in suppressing the immune response and favoring tissue remodeling and consequentially metastasis, and drug resistance [30]. They originate from monocytes after being recruited by factors (i.e, MCP1) released from the stromal and neoplastic cells at the tumor site [16]. Majority of TAMs are associated with Th2 response, secreting IL-10, CCL17, CCL2, CCL22, and TGF-β that favor cancer cell survival, and CCL22 that suppresses antitumor immunity by Treg action [16, 30].
Table 1
Important features that help in differentiating M1 from M2 macrophages
Factors
M1 Macrophage
M2 Macrophage
Action
Pro-inflammatory, anti-microbial
Anti-inflammatory,
Induce extracellular matrix
Action in tumor-microenvironment
Anti-tumorigenic
Pro-tumorigenic
Activation
Classical (Th1)
Alternative (Th2)
Stimuli
IFN-y, LPS, GM-CSF
IL-4, IL-13, M-CSF, Glucocorticoids
Cytokines and chemokines release
IFN-γ, IL-8, TNF-α, IL-1β, RANTES (CCL5), CXCL10
IL-10, IL-13, CCL17, CCL18, CCL22
Antigen presentation
Yes
No
Nitric oxide and ROS production
High
Low
From a mechanistic point of view, it has been demonstrated that the equilibrium between activating pathways of STAT1 and STAT3/STAT6 are responsible for the regulation of macrophage polarization and functions [31]. The activation of M1 macrophage polarization is promoted by the increased expression of NF-κB and STAT1, conferring cytotoxic and inflammatory functions. On the other side, the activation of M2 macrophage polarization is mediated by a predominance of STAT3 and STAT6, resulting in immune suppression and tumor progression [32]. Furthermore, peroxisome proliferator-activated receptors (PPAR) such as PPARγ and PPARδ are involved in M2 macrophage activation and oxidative metabolism while KLF4, cooperating with STAT6, induces M2 genes such as Arg-1, Mrc1, Fizz1, PPARγ and inhibits M1 genes (i.e, TNFa, Cox-2, CCL5, iNOS) by sequestering NF-κB coactivators. Then, KLF2 majorly regulates macrophage activation by promoting the inhibition of NF-κB/HIF-1α activities [33]. IL-4 leads to induction of c-Myc, thereby providing control over genes such as Mrc1, Scarb1, and Alox15 involved in activating M2 type including other downstream pathways [32]. Also, IL-4 inhibits IRF5-mediated M1 polarization by inducing the M2-polarizing Jmjd3-IRF4 axis. Thereby, IL-10 helps in promoting M2 type polarization by way of inducing the c-Maf, STAT3, and p50 NF-κB homodimer activities [31].
Various reports in cancer have demonstrated a difference in NF-kB activation between early and late stages of cancer progression: a higher NF-kB in M1 macrophage has antitumoral action in initial stages while in the most advanced stages TAMs have a lower NF-kB activation but higher immunosuppression activity [34]. TAM-M2s secrete various pro-angiogenic factors including vascular endothelial growth factor-A (VEGF-A), TNFα and produces different factors responsible for the induction of lymphangiogenesis [35], thereby supporting tumor cell proliferation, angiogenesis, invasion, and cancer stem cell functions [36]. Recent studies have shown ncRNAs to be one of the key regulators of TME, cancer progression, and cancer cell signaling through exosomes [27, 37]. Exosomes are a type of extracellular vesicles that play a significant role in establishing intercellular communication network between cancer cells, tumor immune and stromal cells [38]. Exosomes carrying tumor associated cargo such as ncRNAs are demonstrated to have the potential of becoming novel biomarkers and targets in progression of cancer as they play significant roles in inducing angiogenesis, metastasis, immune response, and therapeutic resistance [38].

microRNAs regulating M1 or M2 macrophage-type polarization in different cancers

miR-16 expression was reported to be upregulated by epigallocatechin gallate (one of the polyphenols in green tea having anti-tumorigenesis effect) in 4 T1 murine breast cancer (BC) cells and involved in repolarization of M1 macrophages by regulating the TAMs in the TME through reduction of IL-6 and TGF-β and high TNFα [39]. The findings elucidated the anti-tumor activity of miR-16 by inhibiting TAM infiltration and M2 polarization thereby promoting the repolarization of M1-like macrophages [39].
miR-19a-3p is involved in inducing the M2- like macrophage phenotype by regulating the Fra-1 proto-oncogene involved in breast tumor progression and invasion [40]. Overexpression of miR-19a-3p induces inhibition of BC progression and metastasis through macrophage regulation, indicating the potential of therapeutic applications using this miRNA [41]. miR-19a-3p is also involved in the promotion of colitis-associated colorectal cancer (CRC) by regulating the TNFα loops which were majorly found in macrophage and dendritic cells [40]. However, the mechanism behind miR-19a-3p in the induction of M2-like macrophages has not been elucidated to date [40].
miR-21 containing exosomes increased the M2 polarization compared to M1 and were found to be engulfed by CD14+ human monocytes [42]. miR-21 knockdown in the Snail-expressing head and neck cancer (HNC) cells also reduced the Snail-induced M2 polarization which further mitigated the tumor growth and angiogenesis. miR-21 repressed the expression of programmed cell death protein 4 (PDCD4) and IL12A which resulted in M2-type macrophage polarization. The authors provided significant findings that shed light on epithelial to mesenchymal(EMT)-mediated M2 polarization mechanisms potentially serving as a tumor progression biomarker and a good target for the TME remodeling mediated through EMT [42].
miR-29a-3p is often upregulated in the exosomes derived from oral squamous cell carcinoma (OSCC) cells. Exosomes enriched in miR-29a-3p are transported to the unpolarized macrophages and are differentiated into M2-subtype through the activation of SOCS1/STAT6 signaling, eventually leading to invasion and tumor cell proliferation [43].
miR-34a was reported for the inhibition of MCT-1-promoted IL-6R expression and the polarization of M2 which depicts the potential of miR-34a in driving M1 polarization [44, 45]. Another report also demonstrated that the expression of miR-34a in triple-negative BC mediate M1 polarization while antagomiR-34a promotes M2 plasticity [18]. miR-34a is also known for stimulating CRC invasion and metastasis by the IL6R/STAT3/miR-34a feedback loop through its interaction with STAT3 and IL-6R [45].
miR-142-3p is a tumor-suppressor and supports the M1 phenotype of macrophage polarization. This miRNA is often overexpressed in the gliomas and has been demonstrated to be involved in apoptosis of M2 macrophage and inhibits glioma-infiltrating macrophages which in turn inhibits glioma growth [46]. It was reported that miR-142-3p is downregulated in glioblastoma infiltrating macrophages and M2 type showed lower expression of miR-142-3p [46]. M2 macrophages overexpressing miR-142 selectively modulate the transforming growth factor beta receptor 1 that triggered apoptosis of M2 type subset [46].
miR-145 plays an important role in polarizing M2-like macrophages in CRC cells [47]. CRC cells secreting miR-145 in extracellular vesicles were involved in promoting the M2-like phenotype by downregulating the class of enzymes that remove acetyl groups from histone (including histone deacetylase 11) [47]. These have been further demonstrated to be involved in growth of tumors in mice and miR-145 was found to be acting as a communication tool between the TAMs and cancer cells, leading to induction of tumor-promoting TME [47].
miR-155 has been one of the most extensively studied miRNAs in different cancers and is found to be involved in driving the M1 polarization of the macrophages which are generally known to be pro-inflammatory in action [48, 49]. Recently, exosome-mediated expression of miR-155 and miR-125b-2 were reported to be involved in repolarization to M1 macrophages in the transfected pancreatic cancer (PANC) cells [50]. Furthermore, it was shown that modified tumor derived exosomes can reprogramme macrophage polarization in PANC TME [50].
miR-195-5p is involved in tumor suppression in CRC by directly regulating the NOTCH2 expression and thereby inhibiting CRC metastasis and M2-like polarization of macrophages. In turn, miR-195-5p/NOTCH2 was found to inhibit the M2-like TAMs polarization by suppressing the GATA3-mediated IL-4 secretion pathway in CRC cells, in turn helping in M1-type polarization. In summary, miR-195-5p/NOTCH2-GATA3/IL-4 axis can lead to be an impending therapeutic target [51].
miRNA-224 is involved in the inhibition of the progression of prostate cancer (PC) by downregulation of TRIB1 [52]. It was further reported that TRIB1 is responsible for the induction of M2 like-macrophages by IKB-zeta inhibition in the PC that was validated both in vitro and in vivo in mice models [53]. In summary, miR-224 downregulates TRIB1, which induces M2 phenotype macrophages and leads to the inhibition of the IKB-zeta in PC.
miR-301a-3p: The hypoxic exosomal miR-301a-3p promotes metastasis phenotype in PANC cells by inducing differentiation of M2 macrophage from the stromal macrophage [54]. The level of miR-301a-3p was enhanced under hypoxia conditions in both the PC cell-derived exosomes and PANC cells [54]. The genes responsible for the abovementioned regulation were HIF-1a and HIF-2a [54]. Further, knocking down miR-301a-3p in exosomes weakened the polarization of macrophages into M2 type significantly, thereby reducing invasion and the migration capability, and metastatic ability of the PANC cells both in vivo and ex vivo [54].
miR-503 was shown to play a vital role in promoting brain metastasis by programming the microglia inducing M1 to M2 macrophage polarization in BC patients [55]. All these events are initiated when XIST (X-inactive-specific transcript) expression is decreased in BC that activates MSN-c-Met which increases tumor cell stemness [55]. XIST knockout in the mammary glands of mice was significantly promoting the primary tumor growth and metastasis in the brain. These investigations validated the role of miR-503 in regulating M2 polarization in the microglia [55].
miR-1246: Hypoxic glioma cell-derived exosomes dispense miR-1246 in inducing the M2-like macrophage polarization [56], while in ovarian cancer (OC) miR-1246 confers chemoresistance by targeting through the M2-type macrophage polarization [57]. The target of miR-1246 in glioma cancer cells was TERF2IP via the NF-kB and STAT3 pathways [56] while in OC it was mediated through Cav1/p-gp/M2-type macrophage axis [57].
miRNA Let-7b has an important function in the modulation of macrophage polarization, which in turn enhances the presence of TAMs in PC [58]. The upregulation of miR-let-7b was a signature event in PC-TAMs and downregulation in TAMs changed the inflammatory cytokines expression profiles [58]. The magnitude of this function was high, and it led to a reduction of the mobility and angiogenesis of prostate carcinoma cells when treated with let-7b inhibitors [58]. Let-7 expression can be suppressed by STAT3 through the direct instigation of LIN28A/LIN28B expression by the process of inflammation-stimulated EMT [59]. Let-7c regulates inflammation and associated cytokines through sputum macrophages in the initiation and progression of lung cancer (LC) [60]. Recently it was reported that the Lin-28B-let-7-HMGA2 axis was involved in inducing BC stem cells by M1 macrophage activation [61]. miR-let-7a is shown to be involved in M2 macrophage polarization and enhances oxidative phosphorylation activity in the hypoxic melanoma B16-F0 cells which were further engulfed into bone marrow macrophages [62].

Long non-coding RNAs regulating M1 or M2 macrophage-type polarization in cancers

lncRNA ANCR, an anti-differentiation ncRNA, is significantly upregulated in gastric cancer (GC). The overexpressed lncRNA ANCR inhibited FOXO1 expression and affected macrophage polarization [63]. Previous studies showed that FOXO1 is mediator of the production of inflammatory factors by macrophages favoring M1 macrophage polarization and hence, leading to anti-tumor environments [64]. The upregulation of lncRNA ANCR in GC enhanced the migration and the invasion of the tumor, inhibiting macrophage M1 by targeting FOXO1 and by reducing the concentrations of IL-6 and IL-1β in the cells [63].
lncRNA BCRT1 is upregulated and correlated with poor prognosis through its involvement in tumor growth and metastasis in BC patients. It was shown that lncRNA BCRT1 acts as a tumor promoter by competitively binding to miR-1303 and thereby preventing the degradation of the miRNA target gene PTBP3, a tumor promoter in BC [65]. Overexpression of lncRNA BCRT1 promotes M2 macrophage polarization through exosome-mediated transfer experiments [65].
lncRNA CCAT1(colon cancer-associated transcript-1) has been studied in several cancer types and shown to be overexpressed in GC, multiple myeloma (MM), and BC [66]. lncRNA CCAT1 promotes cell proliferation and migration by interacting with miR-148a and regulates PIK3IP1 in OS [67]. lncRNA CCAT1 is highly expressed by M1 macrophages than TAM (M2-like) macrophages. Furthermore, M1 macrophages are characterized also by a higher expression of PKCζ that is negatively correlated with miR-148a expression level because PKCζ is the target gene of that miRNA [68]. In PC, miR-148a has been reported to be upregulated [69], while the expression of lncRNA CCAT1 has been reported to be varying by IL-4 stimulation. lncRNA CCAT1 downregulation promotes polarization towards to M2 and tumor invasiveness by increasing miR-148a expression that favored M2 macrophage polarization by decreasing PKCζ expression level [68].
lncRNA Cox-2 expression is induced by toll-like receptors (TLR) and it regulates the expression of different genes such as Ccl5, Tlr7, Stat1, IkB, and Icam1 involved in immune regulations [70]. It is also regulated by the nuclear antisense ncRNA PACER, transcribed 300 bp upstream COX-2 transcriptional start site, that interact with the homodimer NF-kB repressor p50/p50, inhibiting its interaction with the promoter and favoring the activating interaction of heterodimer p50/p65 that induce lncRNA Cox-2 transcription [71]. lncRNA Cox-2 has been studied in HCC and is highly expressed in M1 macrophages compared to M2 macrophages [72]. Inhibition of lncRNA Cox-2 decreases levels of certain cytokines including IL-12, iNOS, and TNFα in M1 macrophages and increases the levels of Arg-1, IL-10, and Fizz-1 in M2 macrophages [73, 74]. lncRNA Cox-2 normally inhibits EMT, evasion of immune cells, migratory, and invasive abilityof HCC cells advancing M1 macrophage polarization and inhibiting the M2 macrophage phenotype. Inhibition of Cox-2 can setback the inhibition of M1-type macrophages on tumor proliferation, metastasis, migration, and invasion while increasing the polarization to M2 macrophage in HCC simultaneously [72].
lncRNA GAS5 is involved in the inhibition of tumor progression in different cancer types such as CRC, non-small-cell lung carcinoma (NSCLC), low-grade gliomas, and GC [7578]. lncRNA GAS5 has a role in macrophage polarization in CRC macrophages and is highly expressed in M1 macrophages. Its inhibition has been shown to promote macrophages towards M2 phenotype, suggesting its role in polarization to M1. Nevertheless, recent reports demonstrated that lncRNA GAS5 inhibits the CRC cell proliferation, promotion of apoptosis and migration [76]. However, the mechanism of action of this lncRNA seems to be ambiguous and needs to be further elucidated [76]. Some studies showed that GAS5 is correlated with poor prognosis in several cancer types including NSCLC and GC [77, 78]. Later studies suggested that lncRNA GAS5 can be used as a predictor of patient survival in glioblastoma brain cancer patients [79, 80]. Low expression of lncRNA GAS5 has also been to be associated with poor survival in low-grade glioma [81].
lncRNA GNAS-AS1 is overexpressed in TAMs, tumor tissues, cell lines and positively correlates with poor patient survival and metastatic free survival in NSCLC patients [82]. lncRNA GNAS-AS1 can promote M2 polarization and enhance expression of TAM markers (IL-10 and Arg-1) [82]. lncRNA GNAS-AS1 induces NSCLC cell proliferation, migration, and invasion through its ability to inhibit miR-4319 and thereby promoting the expression of miR-4319 that targets as N-terminal EF-hand calcium-binding protein 3 (NECAB3) [82]. GNAS-AS1-overexpression induces macrophage differentiation of THP-1-monocytic cells and promotes cell viability, migration, and invasion of NSCLC cells [82]. The role of GNAS-AS1 as a promoter of M2 polarization has been also studied in ER-positive BC [83]. GNAS-AS1 is overexpressed in ER+ breast cancer tissues and M2 macrophages. GNAS-AS1 mediates sponging of miR-433-3p and accelerates M2 macrophage polarization and promotes cell proliferative, migration, and invasive ability of ER-positive breast cancer cells [83]. One of the targets of miR-433-3p is GATA3 that could be positively regulated by GNAS-AS1 [83].
lncRNA LINC00662 is mainly located in the cytoplasm and its expression is similar to the levels of miR-15a/16/107 [84]. LINC00662 acts as an oncogene in different human cancers such as acute myeloid leukemia (AML), GC, PC, LC [8487], and HCC [88]. This lncRNA has strong expression in HCC tissues when matched to normal liver tissues and its expression is associated with poor prognosis [88]. LINC00662 can interact as ceRNA that binds to miR-15a, miR-16, and miR-107, inhibiting their action on the target genes and inducing the activation of Wnt/β-catenin signaling in HCC cells and macrophages [88]. In HCC cells, activation of Wnt/β-catenin signaling occurs in an autocrine mode and it induces the proliferation and invasion ability in HCC cells while in macrophage the Wnt/β-catenin signaling is activated in paracrine mode, thereby promoting M2 polarization [88].
lncRNA MALAT1 (Metastasis-Associated Lung Adenocarcinoma Transcript 1) plays a vital role in the developmental stages of various cancer types as liver cancer [89] and renal carcinoma [90]. It plays a role as a modulator of angiogenesis and immune response in several cancer types, including human neuroblastoma [91] and thyroid cancer by modulating the secretion of fibroblast growth factor (FGF2) from TAM promoting angiogenesis [92]. In HCC cells, MALAT1 and VEGF-A are both overexpressed compared to healthy normal cells. The interaction between VEGF-A and miR-140 and between miR-140 and MALAT1 is validated where lncRNA MALAT1 acts as a molecular sponge against miR-140 [93]. MALAT1 has a negative correlation with miR-140, hence, the inhibition of miR-140 in HCC induces angiogenesis in HCC cells while favoring macrophage M2 polarization [93].
lncRNA MM2P: According to the lncRNA microarray-based profiling assay, lncRNA-5730422e09Rik, then named lncRNA MM2P, has been shown to promote M2 polarization [94]. During polarization of M2-type macrophages, this lncRNA was upregulated while in contrast was downregulated during the M1-type polarization. Furthermore, knocking down of lncRNA MM2P blocked the cytokine-driven M2-type macrophage polarization and reduced the strength of the angiogenesis-promoting characteristics of M2-type macrophages by inhibiting the STAT6 phosphorylation [94]. lncRNA-MM2P may be able to maintain the activation of STAT6 and so it might be involved in macrophage M2 polarization [94].
lncRNA NEAT1 (nuclear paraspeckle assembly transcript 1) is overexpressed in many cancerous conditions and exerts oncogenic functions [95]. Furthermore, it regulates the accumulation of miR-214 by molecular sponging in endometrial carcinoma cells [96]. lncRNA NEAT1 induces tumor cell proliferation and induces M2 TAM polarization via molecular sponging of miR-214 and regulates the expression of B7-H3 (immune checkpoint regulator) [97]. B7-H3 can also regulate M2 macrophage polarization via the JAK2/STAT3 signaling but the underlying molecular mechanism has not been fully elucidated yet [98].
lncRNA NIFK-AS1 is often downregulated in the TAMs in endometrial cancer (EC) [99]. NIFK-AS1 overexpression inhibits IL4-induced M2 polarization by NIFK-AS1/miR-146a through targeting Notch1 that helps in the expression of pro-inflammatory cytokines in M1 macrophages [99]. Overexpression of miR-146a favors the expression of M2-inducing genes like IL-10 and Arg-1, thereby affecting tumor growth and TAM recruitment [100]. LncRNA NIFK-AS1 negatively regulates miR-146a, increasing the expression of Notch1 in macrophages and so it is involved in estrogen-induced proliferation, invasiveness, and migration of EC cells. However, overexpression of miR-146a reduces the inhibitory action of NIFK-AS1 on M2 polarization suggesting the important role of both these ncRNAs in the M2 polarization process [99].
LincRNA p21 has been studied in several diseases including cancers such as NSCLC where it’s involved in angiogenesis and has been linked to poor prognosis in resected NSCLC patients [101]. It also plays a vital role in atherosclerosis where it is found to modulate p53-dependent target genes [102]. LincRNA p21 plays a critical role in p53/MDM2 stability by directly interacting with p53 and abolishing MDM2 degradation to p53. This event favors the M2-like TAM polarizing phenotype in BC patients [103]. LincRNA p21 knockout reverses the M2-like TAM phenotype towards an anti-tumor function, mediated by the interaction of p53 with MDM2 leading to induction of NF-κB and STAT3 signaling pathway activation [104].
lncRNA RPPH1 is upregulated in CRCs and associated with poor patient prognosis [105]. RPPH1 can promote tumor cell migration and invasiveness in CRC. LncRNA RPPH1 interacts with TUBB3 and inhibits its ubiquitination, leading to enhancement of its stability. TUBB3 activates the Snail pathway and induces EMT that promotes metastasis. RPPH1 is also transported through exosome to macrophages in TME favoring their M2 polarization and its upregulation is observed in the plasma of CRC patients, suggesting its potential as a therapeutic target and diagnostic marker in CRC [105]. lncRNA RPPH1 acts as an oncogene also in BC [106] and acute myeloid leukemia (AML) [107].
lncRNA RP11-361F15.2 is highly expressed in osteosarcoma (OS) tissues and correlated with cytoplasmic polyadenylation element binding protein 4 (CPEB4) [108]. lncRNA RP11-361F15.2 acts as a ceRNA that binds to miR-30c-5p, blocks the inhibitory action of this microRNA on CPEB4 and thereby increases its expression levels [108]. The upregulation of CPEB4 leads to increased cell motility and migration, proliferation, and invasive ability in OS [108]. Upregulation of RP11-361F15.2 and CPEB4 is responsible for increase in the expression of many polarization markers of M2 such as CD206, Arg1, and Ym1 [108]. All together RP11-361F15.2 fosters tumor progression through CPEB4 and inhibits M2-Like polarization of TAMs using miR-30c-5p in OS [108].
lncRNA TUC339 is an HCC-derived exosomal lncRNA [109] involved in regulating the macrophage polarization of M1/M2 in the TME. This lncRNA has a positive association with IL-4 (M2-type) macrophage polarization. Its overexpression is linked to the downregulation of the FcR-mediated phagocytosis pathway and the actin cytoskeleton pathway, thus concedes phagocytosis. TUC339 overexpression also affects cell motility and migration of macrophage’s progenitors, reduces the pro-inflammatory cytokines production while decreasing co-stimulatory molecules expression by the interaction with THP-1 monocytic cells [110].
lncRNA XIST act as an oncogene by promoting tumor growth and EMT in several cancer types such as colorectal cancer and retinoblastoma where it is demonstrated to be a marker of poor prognosis [111113]. In LCs, it has been demonstrated that lncRNA XIST plays an essential role for invasion, migration and proliferation of LC cells by modulating miR-186-5p [55]. Furthermore, the expression of lncRNA XIST is regulated by TCF-4 (Transcription factor 4) and it positively regulates M2 macrophage polarization making themselves potential targets for gene therapy in LCs [114].

Complexities and contradictions in this field

ncRNAs are functionally categorized as regulatory molecules that are involved in many cellular processes. They can interact with each other and generate a complex regulatory network that can affect target genes [2, 3, 115]. These properties make the ncRNA biology more complex and harder to understand their differential role in cancer cells and TME, as some ncRNAs act as tumor suppressors or oncogenes depending on the target genes, cancer type and the genetic make-up of the cells [116]. Regarding the roles of ncRNAs in regulating M1/M2 polarization, some of them show dual macrophage polarization behavior in different cancer types (Table 2). Let7a-5p is involved in M1 macrophage reprogramming in lung adenocarcinoma cells [119, 120] while let-7a in hypoxic melanoma B16-F0 cells is involved in M2 macrophage polarization and enhances oxidative phosphorylation activity through regulation of mTOR signaling pathway in macrophages [62, 121]. miR-21 is involved in TLR activation, activation of pro-inflammatory cytokine signaling, and NF-kB in NSCLC cells [122] and CRC cells which are M1-type functional characteristics of macrophages [123]. However, in glioma cells, miR-21 has been reported to induce proliferation, tumor promotion, and immunosuppression that shows M2-type functional characteristics [124].
Table 2
Summary of all the ncRNAs that are currently reported to be involved in regulating macrophage polarization in different types of cancer
ncRNA
Cancer type
Macrophage polarization
Targets/signaling pathways
Categorization
Reference
miR-34-a
BC
Promote M1 Polarization, Inhibit M2 Polarization
IL-6R, STAT3
Juggling Tumor Suppressor
[44, 45]
miR-16
BC
Promote M1 Polarization, Inhibit M2 Polarization
IL-6, TGFβ, TNFα
Juggling Tumor Suppressor
[39]
miR-503
BC
Promote M2 Polarization, Inhibit M1 Polarization
STAT3 phosphorylation, NF-kB
Juggling Oncogene
[55]
miR-19a-3p
BC
Promote M2 Polarization
Fra-1 proto-oncogene, TNFα, NF-kB
Oncogene
[40, 41]
lncRNA GNAS-AS1
BC, NSCLC
Promote M2 Polarization
p53
Oncogene
[82, 83]
LincRNA p21
BC
Promote M2 Polarization
miR-1303 sponging, PTBP3
Oncogene
[101104]
lncRNA BCRT1
BC
Promote M2 Polarization
miR-433-3p sponging, IL-10 and Arg-1
Oncogene
[65]
miR-Let-7b
PC
Promote M2 Polarization
IL-12, IL-23, TNFα
Oncogene
[5862]
miR-224
PC
Promote M2 Polarization
TRIB1, IKB-zeta
Oncogene
[52, 53]
lncRNA CCAT1
PC
Promote M1 Polarization, Inhibit M2 Polarization
lncRNA CCAT1/miR-148a/PKCζ axis
Juggling Tumor Suppressor
[6668]
miR-301a-3p
PANC
Promote M2 Polarization
PTEN/PI3Kgamma
Oncogene
[54, 117]
miR-155
PANC, leukemia
Promote M1 Polarization
C/EBPβ
Tumor Suppressor
[4850]
miR-1246
glioma, OC
Promote M2 Polarization
NF-kB-STAT3-TERF2IP axis
Oncogene
[56]
miR-142-3p
glioma
Promote M1 Polarization
TGF-β
Tumor Suppressor
[46, 118]
lncRNA LINC00662
HCC
Promote M2 Polarization
Wnt/β-catenin
Oncogene
[84, 85, 88]
lncRNA MALAT1
HCC
Promote M2 Polarization, Inhibit M1 Polarization
miR-140, VEGF-A
Juggling Oncogene
[92, 93]
lncRNA TUC339
HCC
Promote M2 Polarization
IL-1 β, TNFα
Oncogene
[109, 110]
lncRNA COX-2
HCC
Promote M1 Polarization, Inhibit M2 Polarization
IL-12, iNOS, and TFN-alpha(M1), Arg-1, IL-10, and Fizz-1(M2)
Juggling Tumor Suppressor
[71, 72]
miR-145
CRC
Promote M2 Polarization
IL-12p40, IL-10, HDAC11
Oncogene
[47]
miR-195-5p
CRC
Promote M2 Polarization
miR-195-5p/NOTCH2-GATA3/IL-4 axis
Oncogene
[51]
lncRNA GAS5
CRC
Promote M1 Polarization, Inhibit M2 Polarization
IRF4
Juggling Tumor Suppressor
[75, 76]
lncRNA RPPH1
CRC
Promote M2 Polarization
TUBB3
Oncogene
[105107]
lncRNA MM2P
OS
Promote M2 Polarization
STAT6
Oncogene
[94]
lncRNA RP11-361F15.2
OS
Inhibit M2 Polarization
miR-30c-5p, CPEB4
Tumor Suppressor
[108]
miR-29a-3p
OSCC
Promote M2 Polarization
SOCS1/STAT6 signaling
Oncogene
[43]
miR-21
HNC
Promote M2 Polarization
PDCD4, IL12A
Oncogene
[42]
lncRNA NIFK-AS1N
EC
Promote M1 Polarization, Inhibit M2 Polarization
NIFK-AS1/miR-146a/NOTCH1 axis
Juggling Tumor Suppressor
[99]
lncRNA ANCR
GC
Inhibit M1 Polarization
FOXO1
Oncogene
[63]
lncRNA NEAT1
MM
Promote M2 Polarization
lncRNA NEAT1/miR-214/B7-H3/JAK2-STAT3 axis
Oncogene
[95, 96]
lncRNA XIST
LC
Promote M2 Polarization
IL-4, TCF-4
Oncogene
[111, 114]
Among the lncRNAs involved in M1/ M2 polarization, some have both tumor-suppressive or oncogenic behavior depending on cancer type; for example, lncRNA ANCR regulates M1 polarization in GC by promoting migration and invasion of tumors. However, some of the previous studies demonstrated that ANCR inhibits tumor metastasis by acting as a tumor suppressor in different cancer types as CRC [125], BC [126], and NSCLC [127]. In NSCLC, ANCR acts as an upstream regulator of TGF-β, the downregulation of which normally promotes EMT, suggesting an oncogenic function [127]. Also, lncRNA NEAT1 induces M2 TAM polarization and promotes proliferation of tumor cells in MM [97], while it exerts tumor suppressive behavior in other cancers such as colon, lung and BC where the reduced expression is associated with poor prognosis by transcriptionally regulating p53-dependent pathways [128]. MALAT1 is one of the most studied lncRNA in the last decade and has been shown to promote M2 polarization via VEGF-A and angiogenesis in HCC [93]. It acts as oncogenes also in many other cancer types, including liver cancer [89] and renal carcinoma [90]. However, in colon and BC, tumor-suppressive role of MALAT1 has been discovered [129]. MALAT1 is a downstream target of PTEN, and it is localized in the nucleus and perinuclear space where it is modulated by PTEN-targeting microRNAs such as miR-17, 20a, and 106b [129]. MALAT1 and PTEN are downregulated in BC and CRC, inhibiting their invasiveness and migration by regulating pro-migratory gene expression, as EPCAM and ITGB4 [129]. All these discoveries emphasize the cancer type-specific role of MALAT1 in tumor development [92]. lncRNA CCAT1 is an essential regulator of M2 polarization and tumor cell invasion in PC, where it’s less expressed while its target gene miR-148a is highly expressed and so it inhibits PKCζ [68]. However, CCAT1 is also well known as an oncogenic lncRNA that is overexpressed in different cancer types as CRC, HCC, OC, LC, GC, and bladder cancer where it enhances tumor cell proliferation, invasiveness, and migration [66]. Studies performed on cancer patients of the abovementioned types also demonstrated that an elevated expression of the lncRNA CCAT1 correlated with poor prognosis [130]. Overall, these studies suggest that the role of ncRNAs in macrophage polarization in different cancers are complex and need to be extensively studied in multiple types of cancer that will help in better understanding and in proposing a new systematic categorization.

Juggling tumor suppressors vs Juggling oncogenic ncRNAs

As described above, various ncRNAs play different roles in TAM-M2 polarization in cancer. We have denoted them as “Oncogenes” for the ncRNAs that promote M2 polarization or inhibit M1 polarization. This category includes miR-19a-3p, miR-21, miR-29a-3p, miR-145, miR-195-5p, miR-224, miR-301a-3p, miR-1246, miR-let-7, lncRNA BCRT1, lncRNA GNAS-AS1, LINC00662, LincRNA-p21, lncRNA MM2P, lncRNA NEAT1, lncRNA RPPH1, and lncRNA TUC339 for M2 promotion and lncRNA ANCR for M1 inhibition. We denoted another ncRNA category as “Tumor Suppressor” for all those ncRNAs that promote M1 polarization or inhibit M2 polarization such as miR-142-3p and miR-155 for M1 promotion and lncRNA RP11-361F15.2 for M2 inhibition. The other two categories of ncRNAs are designated as “Juggling Tumor Suppressors” or “Juggling Oncogenes”; the juggling tumor suppressors are ncRNAs that promote M1 and inhibit M2 polarization while the juggling oncogenes promote M2 and inhibit M1 polarization (Fig. 2) because the juggling attribute describes the multitasking abilities of these ncRNAs. The juggling tumor suppressor ncRNA category includes miR-16, miR-34a, miR-142-3p, lncRNA CCAT1, lncRNA COX2, lncRNA GAS5, and lncRNA NIFK-AS1 while the category of juggling oncogene includes miR-503 and lncRNA MALAT1. We should also consider the fact that a juggling oncogene can be a tumor suppressor and vice versa in malignant cells (Fig. 3).
The above classification can be influenced by the increased knowledge of each ncRNA and so further studies about ncRNAs role in macrophage polarization in cancer could lead to more additions to our classification. For example, discoveries could convert a simple tumor suppressor into a juggling tumor suppressor depending on the new findings about the functional role of ncRNAs. However, no reported studies were found yet in the literature that demonstrated the role of other ncRNAs (excluding miRNAs and lncRNAs) in regulating macrophagic polarization in different cancers, although few ones are related to non-cancer diseases [131].

Conclusion and future directions on ncRNA-therapeutics

In the past decade, the therapeutic applications of ncRNAs not only in cancer but also in other deadly human diseases have significantly increased in numbers and first-in-human miRNA-therapeutics were initiated and tested in phase I clinical trials and recently moved into Phase II clinical trials for advanced tumors, including Cobomarsen (locked nucleic acid based oligo-inhibitor of miR-155) in T-cell Leukemia/ Lymphoma [132], TargomiR (miR-16 mimic based therapy) in mesothelioma [133] and Miravirsen (anti-miR-122) in individuals infected with hepatitis C virus (HCV) [134]. ncRNAs can have the potential to be used as regulators of macrophage polarization and reprogramme TME as well as modulate the immune response as several studies have reported that ncRNAs regulating the gene expression are involved in immune cell function and immune checkpoint inhibitors, thereby mediating the immune responses in various cancers [135]. In fact, currently miRNA mimics and antagonists capable of reprogramming TME are being tested in clinical trials in humans as a novel class of promising therapeutic strategies [136]. The miRNA mimics would act as replacements for downregulated endogenous miRNAs. For example, miR-138 mimic can specifically target and bind to mRNA of PDL1 and CTLA4 and suppress their expressions, mimicking the roles of anti-PD-1 and anti-CTLA-4 antibodies [137].
Manipulation of exosomal lncRNAs to target tumors and TME is another interesting therapeutic strategy owing to the exceptional biocompatibility and biodistribution of extracellular vesicles which can be used for the delivery of therapeutic cargo or modulator ncRNA delivery into tumors [138]. It is also interesting to underline that in addition to miRNAs, lncRNAs can also be thrown into the extracellular space and have potential to get detected in the body fluids. This can be exploited further to develop potential circulating ncRNAs biomarkers that can reflect the expression of secreted body fluids in normal versus malignant cells [37].
Therefore, there is a clear need to broaden the ncRNA horizon which demand more investigations for the proper elucidation of novel ncRNAs that are likely to emerge as important regulators of immunological processes including macrophage polarization and potential molecular targets and therapeutic tools related to their oncogenic or tumor suppressor functions in various human cancers.

Acknowledgements

We have used Biorender.com to prepare the figure illustrations.
Not applicable
Not applicable

Competing interests

Authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74.CrossRef Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74.CrossRef
2.
Zurück zum Zitat Fabbri M, Girnita L, Varani G, Calin GA. Decrypting noncoding RNA interactions, structures, and functional networks. Genome Res. 2019;29(9):1377–88.PubMedPubMedCentralCrossRef Fabbri M, Girnita L, Varani G, Calin GA. Decrypting noncoding RNA interactions, structures, and functional networks. Genome Res. 2019;29(9):1377–88.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018;18(1):5–18.PubMedCrossRef Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018;18(1):5–18.PubMedCrossRef
4.
Zurück zum Zitat Tani H, Numajiri A, Aoki M, Umemura T, Nakazato T. Short-lived long noncoding RNAs as surrogate indicators for chemical stress in HepG2 cells and their degradation by nuclear RNases. Sci Rep. 2019;9(1):20299.PubMedPubMedCentralCrossRef Tani H, Numajiri A, Aoki M, Umemura T, Nakazato T. Short-lived long noncoding RNAs as surrogate indicators for chemical stress in HepG2 cells and their degradation by nuclear RNases. Sci Rep. 2019;9(1):20299.PubMedPubMedCentralCrossRef
5.
6.
Zurück zum Zitat Rigoutsos I, Lee SK, Nam SY, Anfossi S, Pasculli B, Pichler M, et al. N-BLR, a primate-specific non-coding transcript leads to colorectal cancer invasion and migration. Genome Biol. 2017;18(1):98.PubMedPubMedCentralCrossRef Rigoutsos I, Lee SK, Nam SY, Anfossi S, Pasculli B, Pichler M, et al. N-BLR, a primate-specific non-coding transcript leads to colorectal cancer invasion and migration. Genome Biol. 2017;18(1):98.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Redis RS, Calin GA. SnapShot: non-coding RNAs and metabolism. Cell Metab. 2017;25(1):220–e1.PubMedCrossRef Redis RS, Calin GA. SnapShot: non-coding RNAs and metabolism. Cell Metab. 2017;25(1):220–e1.PubMedCrossRef
8.
Zurück zum Zitat Ma L, Bajic VB, Zhang Z. On the classification of long non-coding RNAs. RNA Biol. 2013;10(6):925–33.PubMedCrossRef Ma L, Bajic VB, Zhang Z. On the classification of long non-coding RNAs. RNA Biol. 2013;10(6):925–33.PubMedCrossRef
9.
Zurück zum Zitat Ferdin J, Nishida N, Wu X, Nicoloso MS, Shah MY, Devlin C, et al. HINCUTs in cancer: hypoxia-induced noncoding ultraconserved transcripts. Cell Death Differ. 2013;20(12):1675–87.PubMedPubMedCentralCrossRef Ferdin J, Nishida N, Wu X, Nicoloso MS, Shah MY, Devlin C, et al. HINCUTs in cancer: hypoxia-induced noncoding ultraconserved transcripts. Cell Death Differ. 2013;20(12):1675–87.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Tacke F, Ginhoux F, Jakubzick C, van Rooijen N, Merad M, Randolph GJ. Immature monocytes acquire antigens from other cells in the bone marrow and present them to T cells after maturing in the periphery. J Exp Med. 2006;203(3):583–97.PubMedPubMedCentralCrossRef Tacke F, Ginhoux F, Jakubzick C, van Rooijen N, Merad M, Randolph GJ. Immature monocytes acquire antigens from other cells in the bone marrow and present them to T cells after maturing in the periphery. J Exp Med. 2006;203(3):583–97.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Boyette LB, Macedo C, Hadi K, Elinoff BD, Walters JT, Ramaswami B, et al. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS One. 2017;12(4):e0176460.PubMedPubMedCentralCrossRef Boyette LB, Macedo C, Hadi K, Elinoff BD, Walters JT, Ramaswami B, et al. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS One. 2017;12(4):e0176460.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Allen M, Louise JJ. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J Pathol. 2011;223(2):162–76.PubMedCrossRef Allen M, Louise JJ. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J Pathol. 2011;223(2):162–76.PubMedCrossRef
15.
Zurück zum Zitat Tarique AA, Logan J, Thomas E, Holt PG, Sly PD, Fantino E. Phenotypic, functional, and plasticity features of classical and alternatively activated human macrophages. Am J Respir Cell Mol Biol. 2015;53(5):676–88.PubMedCrossRef Tarique AA, Logan J, Thomas E, Holt PG, Sly PD, Fantino E. Phenotypic, functional, and plasticity features of classical and alternatively activated human macrophages. Am J Respir Cell Mol Biol. 2015;53(5):676–88.PubMedCrossRef
16.
Zurück zum Zitat Soysal SD, Tzankov A, Muenst SE. Role of the tumor microenvironment in breast cancer. Pathobiology. 2015;82(3–4):142–52.PubMedCrossRef Soysal SD, Tzankov A, Muenst SE. Role of the tumor microenvironment in breast cancer. Pathobiology. 2015;82(3–4):142–52.PubMedCrossRef
17.
Zurück zum Zitat Essandoh K, Li Y, Huo J, Fan GC. MiRNA-mediated macrophage polarization and its potential role in the regulation of inflammatory response. Shock. 2016;46(2):122–31.PubMedPubMedCentralCrossRef Essandoh K, Li Y, Huo J, Fan GC. MiRNA-mediated macrophage polarization and its potential role in the regulation of inflammatory response. Shock. 2016;46(2):122–31.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Weng YS, Tseng HY, Chen YA, Shen PC, Al Haq AT, Chen LM, et al. MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol Cancer. 2019;18(1):42.PubMedPubMedCentralCrossRef Weng YS, Tseng HY, Chen YA, Shen PC, Al Haq AT, Chen LM, et al. MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol Cancer. 2019;18(1):42.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61.PubMedCrossRef Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61.PubMedCrossRef
21.
Zurück zum Zitat Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–86.CrossRefPubMed Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–86.CrossRefPubMed
22.
Zurück zum Zitat Arora S, Dev K, Agarwal B, Das P, Syed MA. Macrophages: their role, activation and polarization in pulmonary diseases. Immunobiology. 2018;223(4–5):383–96.PubMedCrossRef Arora S, Dev K, Agarwal B, Das P, Syed MA. Macrophages: their role, activation and polarization in pulmonary diseases. Immunobiology. 2018;223(4–5):383–96.PubMedCrossRef
23.
Zurück zum Zitat Anderson CF, Mosser DM. A novel phenotype for an activated macrophage: the type 2 activated macrophage. J Leukoc Biol. 2002;72(1):101–6.PubMed Anderson CF, Mosser DM. A novel phenotype for an activated macrophage: the type 2 activated macrophage. J Leukoc Biol. 2002;72(1):101–6.PubMed
24.
Zurück zum Zitat Wang HW, Joyce JA. Alternative activation of tumor-associated macrophages by IL-4: priming for protumoral functions. Cell Cycle. 2010;9(24):4824–35.PubMedPubMedCentralCrossRef Wang HW, Joyce JA. Alternative activation of tumor-associated macrophages by IL-4: priming for protumoral functions. Cell Cycle. 2010;9(24):4824–35.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Silverman DA, Martinez VK, Dougherty PM, Myers JN, Calin GA, Amit M. Cancer-associated neurogenesis and nerve-cancer crosstalk. Cancer Res. 2020. Silverman DA, Martinez VK, Dougherty PM, Myers JN, Calin GA, Amit M. Cancer-associated neurogenesis and nerve-cancer crosstalk. Cancer Res. 2020.
26.
Zurück zum Zitat Dragomir MP, Moisoiu V, Manaila R, Pardini B, Knutsen E, Anfossi S, et al. A holistic perspective: exosomes shuttle between nerves and immune cells in the tumor microenvironment. J Clin Med. 2020;9(11). Dragomir MP, Moisoiu V, Manaila R, Pardini B, Knutsen E, Anfossi S, et al. A holistic perspective: exosomes shuttle between nerves and immune cells in the tumor microenvironment. J Clin Med. 2020;9(11).
27.
Zurück zum Zitat Lin YH, Wu MH, Yeh CT, Lin KH. Long Non-Coding RNAs as Mediators of Tumor Microenvironment and Liver Cancer Cell Communication. Int J Mol Sci. 2018;19(12). Lin YH, Wu MH, Yeh CT, Lin KH. Long Non-Coding RNAs as Mediators of Tumor Microenvironment and Liver Cancer Cell Communication. Int J Mol Sci. 2018;19(12).
28.
29.
Zurück zum Zitat Amit M, Takahashi H, Dragomir MP, Lindemann A, Gleber-Netto FO, Pickering CR, et al. Loss of p53 drives neuron reprogramming in head and neck cancer. Nature. 2020;578(7795):449–54.PubMedCrossRef Amit M, Takahashi H, Dragomir MP, Lindemann A, Gleber-Netto FO, Pickering CR, et al. Loss of p53 drives neuron reprogramming in head and neck cancer. Nature. 2020;578(7795):449–54.PubMedCrossRef
30.
Zurück zum Zitat Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol. 2009;86(5):1065–73.PubMedCrossRef Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol. 2009;86(5):1065–73.PubMedCrossRef
32.
Zurück zum Zitat Pello OM, De Pizzol M, Mirolo M, Soucek L, Zammataro L, Amabile A, et al. Role of c-MYC in alternative activation of human macrophages and tumor-associated macrophage biology. Blood. 2012;119(2):411–21.PubMedCrossRef Pello OM, De Pizzol M, Mirolo M, Soucek L, Zammataro L, Amabile A, et al. Role of c-MYC in alternative activation of human macrophages and tumor-associated macrophage biology. Blood. 2012;119(2):411–21.PubMedCrossRef
33.
Zurück zum Zitat Mahabeleshwar GH, Kawanami D, Sharma N, Takami Y, Zhou G, Shi H, et al. The myeloid transcription factor KLF2 regulates the host response to polymicrobial infection and endotoxic shock. Immunity. 2011;34(5):715–28.PubMedPubMedCentralCrossRef Mahabeleshwar GH, Kawanami D, Sharma N, Takami Y, Zhou G, Shi H, et al. The myeloid transcription factor KLF2 regulates the host response to polymicrobial infection and endotoxic shock. Immunity. 2011;34(5):715–28.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Biswas SK, Gangi L, Paul S, Schioppa T, Saccani A, Sironi M, et al. A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation). Blood. 2006;107(5):2112–22.PubMedCrossRef Biswas SK, Gangi L, Paul S, Schioppa T, Saccani A, Sironi M, et al. A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation). Blood. 2006;107(5):2112–22.PubMedCrossRef
35.
Zurück zum Zitat Mitrofanova I, Zavyalova M, Telegina N, Buldakov M, Riabov V, Cherdyntseva N, et al. Tumor-associated macrophages in human breast cancer parenchyma negatively correlate with lymphatic metastasis after neoadjuvant chemotherapy. Immunobiology. 2017;222(1):101–9.PubMedCrossRef Mitrofanova I, Zavyalova M, Telegina N, Buldakov M, Riabov V, Cherdyntseva N, et al. Tumor-associated macrophages in human breast cancer parenchyma negatively correlate with lymphatic metastasis after neoadjuvant chemotherapy. Immunobiology. 2017;222(1):101–9.PubMedCrossRef
36.
Zurück zum Zitat Larionova I, Cherdyntseva N, Liu T, Patysheva M, Rakina M, Kzhyshkowska J. Interaction of tumor-associated macrophages and cancer chemotherapy. Oncoimmunology. 2019;8(7):1596004.PubMedPubMedCentralCrossRef Larionova I, Cherdyntseva N, Liu T, Patysheva M, Rakina M, Kzhyshkowska J. Interaction of tumor-associated macrophages and cancer chemotherapy. Oncoimmunology. 2019;8(7):1596004.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Anfossi S, Babayan A, Pantel K, Calin GA. Clinical utility of circulating non-coding RNAs - an update. Nat Rev Clin Oncol. 2018;15(9):541–63.PubMedCrossRef Anfossi S, Babayan A, Pantel K, Calin GA. Clinical utility of circulating non-coding RNAs - an update. Nat Rev Clin Oncol. 2018;15(9):541–63.PubMedCrossRef
39.
Zurück zum Zitat Jang JY, Lee JK, Jeon YK, 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. 2013;13:421.PubMedPubMedCentralCrossRef Jang JY, Lee JK, Jeon YK, 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. 2013;13:421.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wang T, Xu X, Xu Q, Ren J, Shen S, Fan C, et al. miR-19a promotes colitis-associated colorectal cancer by regulating tumor necrosis factor alpha-induced protein 3-NF-kappaB feedback loops. Oncogene. 2017;36(23):3240–51.PubMedCrossRef Wang T, Xu X, Xu Q, Ren J, Shen S, Fan C, et al. miR-19a promotes colitis-associated colorectal cancer by regulating tumor necrosis factor alpha-induced protein 3-NF-kappaB feedback loops. Oncogene. 2017;36(23):3240–51.PubMedCrossRef
41.
Zurück zum Zitat 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. 2014;33(23):3014–23.PubMedCrossRef 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. 2014;33(23):3014–23.PubMedCrossRef
42.
Zurück zum Zitat Hsieh CH, Tai SK, Yang MH. Snail-overexpressing cancer cells promote M2-like polarization of tumor-associated macrophages by delivering MiR-21-abundant exosomes. Neoplasia. 2018;20(8):775–88.PubMedPubMedCentralCrossRef Hsieh CH, Tai SK, Yang MH. Snail-overexpressing cancer cells promote M2-like polarization of tumor-associated macrophages by delivering MiR-21-abundant exosomes. Neoplasia. 2018;20(8):775–88.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Cai J, Qiao B, Gao N, Lin N, He W. Oral squamous cell carcinoma-derived exosomes promote M2 subtype macrophage polarization mediated by exosome-enclosed miR-29a-3p. Am J Phys Cell Phys. 2019;316(5):C731–C40.CrossRef Cai J, Qiao B, Gao N, Lin N, He W. Oral squamous cell carcinoma-derived exosomes promote M2 subtype macrophage polarization mediated by exosome-enclosed miR-29a-3p. Am J Phys Cell Phys. 2019;316(5):C731–C40.CrossRef
44.
Zurück zum Zitat Zhang L, Liao Y, Tang L. MicroRNA-34 family: a potential tumor suppressor and therapeutic candidate in cancer. J Exp Clin Cancer Res. 2019;38(1):53.PubMedPubMedCentralCrossRef Zhang L, Liao Y, Tang L. MicroRNA-34 family: a potential tumor suppressor and therapeutic candidate in cancer. J Exp Clin Cancer Res. 2019;38(1):53.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Rokavec M, Oner MG, Li H, Jackstadt R, Jiang L, Lodygin D, et al. IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer invasion and metastasis. J Clin Invest. 2014;124(4):1853–67.PubMedPubMedCentralCrossRef Rokavec M, Oner MG, Li H, Jackstadt R, Jiang L, Lodygin D, et al. IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer invasion and metastasis. J Clin Invest. 2014;124(4):1853–67.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Xu S, Wei J, Wang F, Kong LY, Ling XY, Nduom E, et al. Effect of miR-142-3p on the M2 macrophage and therapeutic efficacy against murine glioblastoma. J Natl Cancer Inst. 2014;106(8). Xu S, Wei J, Wang F, Kong LY, Ling XY, Nduom E, et al. Effect of miR-142-3p on the M2 macrophage and therapeutic efficacy against murine glioblastoma. J Natl Cancer Inst. 2014;106(8).
47.
Zurück zum Zitat Shinohara H, Kuranaga Y, Kumazaki M, Sugito N, Yoshikawa Y, Takai T, et al. Regulated polarization of tumor-associated macrophages by miR-145 via colorectal cancer-derived extracellular vesicles. J Immunol. 2017;199(4):1505–15.PubMedCrossRef Shinohara H, Kuranaga Y, Kumazaki M, Sugito N, Yoshikawa Y, Takai T, et al. Regulated polarization of tumor-associated macrophages by miR-145 via colorectal cancer-derived extracellular vesicles. J Immunol. 2017;199(4):1505–15.PubMedCrossRef
48.
Zurück zum Zitat Sethupathy P, Borel C, Gagnebin M, Grant GR, Deutsch S, Elton TS, et al. Human microRNA-155 on chromosome 21 differentially interacts with its polymorphic target in the AGTR1 3′ untranslated region: a mechanism for functional single-nucleotide polymorphisms related to phenotypes. Am J Hum Genet. 2007;81(2):405–13.PubMedPubMedCentralCrossRef Sethupathy P, Borel C, Gagnebin M, Grant GR, Deutsch S, Elton TS, et al. Human microRNA-155 on chromosome 21 differentially interacts with its polymorphic target in the AGTR1 3′ untranslated region: a mechanism for functional single-nucleotide polymorphisms related to phenotypes. Am J Hum Genet. 2007;81(2):405–13.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat 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(5):341–3.PubMedCrossRef 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(5):341–3.PubMedCrossRef
50.
Zurück zum Zitat Su MJ, Aldawsari H, Amiji M. Pancreatic cancer cell exosome-mediated macrophage reprogramming and the role of MicroRNAs 155 and 125b2 transfection using nanoparticle delivery systems. Sci Rep. 2016;6:30110.PubMedPubMedCentralCrossRef Su MJ, Aldawsari H, Amiji M. Pancreatic cancer cell exosome-mediated macrophage reprogramming and the role of MicroRNAs 155 and 125b2 transfection using nanoparticle delivery systems. Sci Rep. 2016;6:30110.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Lin X, Wang S, Sun M, Zhang C, Wei C, Yang C, et al. miR-195-5p/NOTCH2-mediated EMT modulates IL-4 secretion in colorectal cancer to affect M2-like TAM polarization. J Hematol Oncol. 2019;12(1):20.PubMedPubMedCentralCrossRef Lin X, Wang S, Sun M, Zhang C, Wei C, Yang C, et al. miR-195-5p/NOTCH2-mediated EMT modulates IL-4 secretion in colorectal cancer to affect M2-like TAM polarization. J Hematol Oncol. 2019;12(1):20.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Lin ZY, Huang YQ, Zhang YQ, Han ZD, He HC, Ling XH, et al. MicroRNA-224 inhibits progression of human prostate cancer by downregulating TRIB1. Int J Cancer. 2014;135(3):541–50.PubMedCrossRef Lin ZY, Huang YQ, Zhang YQ, Han ZD, He HC, Ling XH, et al. MicroRNA-224 inhibits progression of human prostate cancer by downregulating TRIB1. Int J Cancer. 2014;135(3):541–50.PubMedCrossRef
53.
Zurück zum Zitat Liu ZZ, Han ZD, Liang YK, Chen JX, Wan S, Zhuo YJ, et al. TRIB1 induces macrophages to M2 phenotype by inhibiting IKB-zeta in prostate cancer. Cell Signal. 2019;59:152–62.PubMedCrossRef Liu ZZ, Han ZD, Liang YK, Chen JX, Wan S, Zhuo YJ, et al. TRIB1 induces macrophages to M2 phenotype by inhibiting IKB-zeta in prostate cancer. Cell Signal. 2019;59:152–62.PubMedCrossRef
54.
Zurück zum Zitat Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, et al. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kgamma to promote pancreatic cancer metastasis. Cancer Res. 2018;78(16):4586–98.PubMedCrossRef Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, et al. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kgamma to promote pancreatic cancer metastasis. Cancer Res. 2018;78(16):4586–98.PubMedCrossRef
55.
Zurück zum Zitat Xing F, Liu Y, Wu SY, Wu K, Sharma S, Mo YY, et al. Loss of XIST in breast cancer activates MSN-c-met and reprograms microglia via exosomal miRNA to promote brain metastasis. Cancer Res. 2018;78(15):4316–30.PubMedPubMedCentralCrossRef Xing F, Liu Y, Wu SY, Wu K, Sharma S, Mo YY, et al. Loss of XIST in breast cancer activates MSN-c-met and reprograms microglia via exosomal miRNA to promote brain metastasis. Cancer Res. 2018;78(15):4316–30.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Qian M, Wang S, Guo X, Wang J, Zhang Z, Qiu W, et al. Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-kappaB pathways. Oncogene. 2020;39(2):428–42.PubMedCrossRef Qian M, Wang S, Guo X, Wang J, Zhang Z, Qiu W, et al. Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-kappaB pathways. Oncogene. 2020;39(2):428–42.PubMedCrossRef
57.
Zurück zum Zitat Kanlikilicer P, Bayraktar R, Denizli M, Rashed MH, Ivan C, Aslan B, et al. Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine. 2018;38:100–12.PubMedPubMedCentralCrossRef Kanlikilicer P, Bayraktar R, Denizli M, Rashed MH, Ivan C, Aslan B, et al. Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine. 2018;38:100–12.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Wang Z, Xu L, Hu Y, Huang Y, Zhang Y, Zheng X, et al. miRNA let-7b modulates macrophage polarization and enhances tumor-associated macrophages to promote angiogenesis and mobility in prostate cancer. Sci Rep. 2016;6:25602.PubMedPubMedCentralCrossRef Wang Z, Xu L, Hu Y, Huang Y, Zhang Y, Zheng X, et al. miRNA let-7b modulates macrophage polarization and enhances tumor-associated macrophages to promote angiogenesis and mobility in prostate cancer. Sci Rep. 2016;6:25602.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Guo L, Chen C, Shi M, Wang F, Chen X, Diao D, et al. Stat3-coordinated Lin-28-let-7-HMGA2 and miR-200-ZEB1 circuits initiate and maintain oncostatin M-driven epithelial-mesenchymal transition. Oncogene. 2013;32(45):5272–82.PubMedCrossRef Guo L, Chen C, Shi M, Wang F, Chen X, Diao D, et al. Stat3-coordinated Lin-28-let-7-HMGA2 and miR-200-ZEB1 circuits initiate and maintain oncostatin M-driven epithelial-mesenchymal transition. Oncogene. 2013;32(45):5272–82.PubMedCrossRef
60.
Zurück zum Zitat Shin JI, Brusselle GG. Mechanistic links between COPD and lung cancer: a role of microRNA let7? Nat Rev Cancer. 2014;14(1):70.PubMedCrossRef Shin JI, Brusselle GG. Mechanistic links between COPD and lung cancer: a role of microRNA let7? Nat Rev Cancer. 2014;14(1):70.PubMedCrossRef
61.
Zurück zum Zitat Guo L, Cheng X, Chen H, Chen C, Xie S, Zhao M, et al. Induction of breast cancer stem cells by M1 macrophages through Lin-28B-let-7-HMGA2 axis. Cancer Lett. 2019;452:213–25.PubMedCrossRef Guo L, Cheng X, Chen H, Chen C, Xie S, Zhao M, et al. Induction of breast cancer stem cells by M1 macrophages through Lin-28B-let-7-HMGA2 axis. Cancer Lett. 2019;452:213–25.PubMedCrossRef
63.
Zurück zum Zitat Xie C, Guo Y, Lou S. LncRNA ANCR promotes invasion and migration of gastric cancer by regulating FoxO1 expression to inhibit macrophage M1 polarization. Dig Dis Sci. 2020. Xie C, Guo Y, Lou S. LncRNA ANCR promotes invasion and migration of gastric cancer by regulating FoxO1 expression to inhibit macrophage M1 polarization. Dig Dis Sci. 2020.
64.
Zurück zum Zitat Yang JB, Zhao ZB, Liu QZ, Hu TD, Long J, Yan K, et al. FoxO1 is a regulator of MHC-II expression and anti-tumor effect of tumor-associated macrophages. Oncogene. 2018;37(9):1192–204.PubMedCrossRef Yang JB, Zhao ZB, Liu QZ, Hu TD, Long J, Yan K, et al. FoxO1 is a regulator of MHC-II expression and anti-tumor effect of tumor-associated macrophages. Oncogene. 2018;37(9):1192–204.PubMedCrossRef
65.
Zurück zum Zitat Liang Y, Song X, Li Y, Chen B, Zhao W, Wang L, et al. LncRNA BCRT1 promotes breast cancer progression by targeting miR-1303/PTBP3 axis. Mol Cancer. 2020;19(1):85.PubMedPubMedCentralCrossRef Liang Y, Song X, Li Y, Chen B, Zhao W, Wang L, et al. LncRNA BCRT1 promotes breast cancer progression by targeting miR-1303/PTBP3 axis. Mol Cancer. 2020;19(1):85.PubMedPubMedCentralCrossRef
66.
67.
Zurück zum Zitat Zhao J, Cheng L. Long non-coding RNA CCAT1/miR-148a axis promotes osteosarcoma proliferation and migration through regulating PIK3IP1. Acta Biochim Biophys Sin Shanghai. 2017;49(6):503–12.PubMedCrossRef Zhao J, Cheng L. Long non-coding RNA CCAT1/miR-148a axis promotes osteosarcoma proliferation and migration through regulating PIK3IP1. Acta Biochim Biophys Sin Shanghai. 2017;49(6):503–12.PubMedCrossRef
68.
Zurück zum Zitat Liu J, Ding D, Jiang Z, Du T, Liu J, Kong Z. Long non-coding RNA CCAT1/miR-148a/PKCzeta prevents cell migration of prostate cancer by altering macrophage polarization. Prostate. 2019;79(1):105–12.PubMedCrossRef Liu J, Ding D, Jiang Z, Du T, Liu J, Kong Z. Long non-coding RNA CCAT1/miR-148a/PKCzeta prevents cell migration of prostate cancer by altering macrophage polarization. Prostate. 2019;79(1):105–12.PubMedCrossRef
69.
Zurück zum Zitat Al-Qatati A, Akrong C, Stevic I, Pantel K, Awe J, Saranchuk J, et al. Plasma microRNA signature is associated with risk stratification in prostate cancer patients. Int J Cancer. 2017;141(6):1231–9.PubMedCrossRef Al-Qatati A, Akrong C, Stevic I, Pantel K, Awe J, Saranchuk J, et al. Plasma microRNA signature is associated with risk stratification in prostate cancer patients. Int J Cancer. 2017;141(6):1231–9.PubMedCrossRef
70.
Zurück zum Zitat Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science. 2013;341(6147):789–92.PubMedPubMedCentralCrossRef Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science. 2013;341(6147):789–92.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Krawczyk M, Emerson BM. p50-associated COX-2 extragenic RNA (PACER) activates COX-2 gene expression by occluding repressive NF-kappaB complexes. Elife. 2014;3:e01776.PubMedPubMedCentralCrossRef Krawczyk M, Emerson BM. p50-associated COX-2 extragenic RNA (PACER) activates COX-2 gene expression by occluding repressive NF-kappaB complexes. Elife. 2014;3:e01776.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Ye Y, Xu Y, Lai Y, He W, Li Y, Wang R, et al. Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. J Cell Biochem. 2018;119(3):2951–63.PubMedCrossRef Ye Y, Xu Y, Lai Y, He W, Li Y, Wang R, et al. Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. J Cell Biochem. 2018;119(3):2951–63.PubMedCrossRef
73.
Zurück zum Zitat Muthian G, Raikwar HP, Johnson C, Rajasingh J, Kalgutkar A, Marnett LJ, et al. COX-2 inhibitors modulate IL-12 signaling through JAK-STAT pathway leading to Th1 response in experimental allergic encephalomyelitis. J Clin Immunol. 2006;26(1):73–85.PubMedCrossRef Muthian G, Raikwar HP, Johnson C, Rajasingh J, Kalgutkar A, Marnett LJ, et al. COX-2 inhibitors modulate IL-12 signaling through JAK-STAT pathway leading to Th1 response in experimental allergic encephalomyelitis. J Clin Immunol. 2006;26(1):73–85.PubMedCrossRef
74.
Zurück zum Zitat He T, Xing YQ, Zhao XH, Ai M. Interaction between iNOS and COX-2 in hypoxia-induced retinal neovascularization in mice. Arch Med Res. 2007;38(8):807–15.PubMedCrossRef He T, Xing YQ, Zhao XH, Ai M. Interaction between iNOS and COX-2 in hypoxia-induced retinal neovascularization in mice. Arch Med Res. 2007;38(8):807–15.PubMedCrossRef
75.
Zurück zum Zitat Cheng K, Zhao Z, Wang G, Wang J, Zhu W. lncRNA GAS5 inhibits colorectal cancer cell proliferation via the miR1825p/FOXO3a axis. Oncol Rep. 2018;40(4):2371–80.PubMed Cheng K, Zhao Z, Wang G, Wang J, Zhu W. lncRNA GAS5 inhibits colorectal cancer cell proliferation via the miR1825p/FOXO3a axis. Oncol Rep. 2018;40(4):2371–80.PubMed
76.
Zurück zum Zitat Tang L, Li Q, Ge X, Miao L. LncRNA GAS5 inhibits progression of colorectal cancer by regulating M1/M2 macrophages polarization. AIP Conference Proceedings. 2019;2110(1):020013.CrossRef Tang L, Li Q, Ge X, Miao L. LncRNA GAS5 inhibits progression of colorectal cancer by regulating M1/M2 macrophages polarization. AIP Conference Proceedings. 2019;2110(1):020013.CrossRef
77.
Zurück zum Zitat Shi X, Sun M, Liu H, Yao Y, Kong R, Chen F, et al. A critical role for the long non-coding RNA GAS5 in proliferation and apoptosis in non-small-cell lung cancer. Mol Carcinog. 2015;54(Suppl 1):E1–E12.PubMedCrossRef Shi X, Sun M, Liu H, Yao Y, Kong R, Chen F, et al. A critical role for the long non-coding RNA GAS5 in proliferation and apoptosis in non-small-cell lung cancer. Mol Carcinog. 2015;54(Suppl 1):E1–E12.PubMedCrossRef
78.
Zurück zum Zitat Sun M, Jin FY, Xia R, Kong R, Li JH, Xu TP, et al. Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer. BMC Cancer. 2014;14:319.PubMedPubMedCentralCrossRef Sun M, Jin FY, Xia R, Kong R, Li JH, Xu TP, et al. Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer. BMC Cancer. 2014;14:319.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Shen J, Hodges TR, Song R, Gong Y, Calin GA, Heimberger AB, et al. Serum HOTAIR and GAS5 levels as predictors of survival in patients with glioblastoma. Mol Carcinog. 2018;57(1):137–41.PubMedCrossRef Shen J, Hodges TR, Song R, Gong Y, Calin GA, Heimberger AB, et al. Serum HOTAIR and GAS5 levels as predictors of survival in patients with glioblastoma. Mol Carcinog. 2018;57(1):137–41.PubMedCrossRef
80.
Zurück zum Zitat Zhang XQ, Sun S, Lam KF, Kiang KM, Pu JK, Ho AS, et al. A long non-coding RNA signature in glioblastoma multiforme predicts survival. Neurobiol Dis. 2013;58:123–31.PubMedCrossRef Zhang XQ, Sun S, Lam KF, Kiang KM, Pu JK, Ho AS, et al. A long non-coding RNA signature in glioblastoma multiforme predicts survival. Neurobiol Dis. 2013;58:123–31.PubMedCrossRef
81.
Zurück zum Zitat Wang Y, Xin S, Zhang K, Shi R, Bao X. Low GAS5 levels as a predictor of poor survival in patients with lower-grade gliomas. J Oncol. 2019;2019:1785042.PubMedPubMedCentral Wang Y, Xin S, Zhang K, Shi R, Bao X. Low GAS5 levels as a predictor of poor survival in patients with lower-grade gliomas. J Oncol. 2019;2019:1785042.PubMedPubMedCentral
82.
Zurück zum Zitat Li Z, Feng C, Guo J, Hu X, Xie D. GNAS-AS1/miR-4319/NECAB3 axis promotes migration and invasion of non-small cell lung cancer cells by altering macrophage polarization. Funct Integr Genomics. 2020;20(1):17–28.PubMedCrossRef Li Z, Feng C, Guo J, Hu X, Xie D. GNAS-AS1/miR-4319/NECAB3 axis promotes migration and invasion of non-small cell lung cancer cells by altering macrophage polarization. Funct Integr Genomics. 2020;20(1):17–28.PubMedCrossRef
83.
Zurück zum Zitat Liu SQ, Zhou ZY, Dong X, Guo L, Zhang KJ. LncRNA GNAS-AS1 facilitates ER+ breast cancer cells progression by promoting M2 macrophage polarization via regulating miR-433-3p/GATA3 axis. Biosci Rep. 2020;40(7). Liu SQ, Zhou ZY, Dong X, Guo L, Zhang KJ. LncRNA GNAS-AS1 facilitates ER+ breast cancer cells progression by promoting M2 macrophage polarization via regulating miR-433-3p/GATA3 axis. Biosci Rep. 2020;40(7).
84.
Zurück zum Zitat Li N, Zhang LY, Qiao YH, Song RJ. Long noncoding RNA LINC00662 functions as miRNA sponge to promote the prostate cancer tumorigenesis through targeting miR-34a. Eur Rev Med Pharmacol Sci. 2019;23(9):3688–98.PubMed Li N, Zhang LY, Qiao YH, Song RJ. Long noncoding RNA LINC00662 functions as miRNA sponge to promote the prostate cancer tumorigenesis through targeting miR-34a. Eur Rev Med Pharmacol Sci. 2019;23(9):3688–98.PubMed
85.
Zurück zum Zitat Gong W, Su Y, Liu Y, Sun P, Wang X. Long non-coding RNA Linc00662 promotes cell invasion and contributes to cancer stem cell-like phenotypes in lung cancer cells. J Biochem. 2018;164(6):461–9.PubMedCrossRef Gong W, Su Y, Liu Y, Sun P, Wang X. Long non-coding RNA Linc00662 promotes cell invasion and contributes to cancer stem cell-like phenotypes in lung cancer cells. J Biochem. 2018;164(6):461–9.PubMedCrossRef
86.
Zurück zum Zitat Liu LZ, Zhang Z, Zheng BH, Shi Y, Duan M, Ma LJ, et al. CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology. 2019;69(1):143–59.PubMedCrossRef Liu LZ, Zhang Z, Zheng BH, Shi Y, Duan M, Ma LJ, et al. CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology. 2019;69(1):143–59.PubMedCrossRef
87.
Zurück zum Zitat Liu F, Yuan JH, Huang JF, Yang F, Wang TT, Ma JZ, et al. Long noncoding RNA FTX inhibits hepatocellular carcinoma proliferation and metastasis by binding MCM2 and miR-374a. Oncogene. 2016;35(41):5422–34.PubMedCrossRef Liu F, Yuan JH, Huang JF, Yang F, Wang TT, Ma JZ, et al. Long noncoding RNA FTX inhibits hepatocellular carcinoma proliferation and metastasis by binding MCM2 and miR-374a. Oncogene. 2016;35(41):5422–34.PubMedCrossRef
88.
Zurück zum Zitat Tian X, Wu Y, Yang Y, Wang J, Niu M, Gao S, et al. Long noncoding RNA LINC00662 promotes M2 macrophage polarization and hepatocellular carcinoma progression via activating Wnt/beta-catenin signaling. Mol Oncol. 2020;14(2):462–83.PubMedCrossRef Tian X, Wu Y, Yang Y, Wang J, Niu M, Gao S, et al. Long noncoding RNA LINC00662 promotes M2 macrophage polarization and hepatocellular carcinoma progression via activating Wnt/beta-catenin signaling. Mol Oncol. 2020;14(2):462–83.PubMedCrossRef
89.
Zurück zum Zitat Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013;73(3):1180–9.PubMedCrossRef Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013;73(3):1180–9.PubMedCrossRef
90.
Zurück zum Zitat Hirata H, Hinoda Y, Shahryari V, Deng G, Nakajima K, Tabatabai ZL, et al. Long noncoding RNA MALAT1 promotes aggressive renal cell carcinoma through Ezh2 and interacts with miR-205. Cancer Res. 2015;75(7):1322–31.PubMedPubMedCentralCrossRef Hirata H, Hinoda Y, Shahryari V, Deng G, Nakajima K, Tabatabai ZL, et al. Long noncoding RNA MALAT1 promotes aggressive renal cell carcinoma through Ezh2 and interacts with miR-205. Cancer Res. 2015;75(7):1322–31.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Tee AE, Liu B, Song R, Li J, Pasquier E, Cheung BB, et al. The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression. Oncotarget. 2016;7(8):8663–75.PubMedPubMedCentralCrossRef Tee AE, Liu B, Song R, Li J, Pasquier E, Cheung BB, et al. The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression. Oncotarget. 2016;7(8):8663–75.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Huang JK, Ma L, Song WH, Lu BY, Huang YB, Dong HM, et al. LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion. J Cell Biochem. 2017;118(12):4821–30.PubMedCrossRef Huang JK, Ma L, Song WH, Lu BY, Huang YB, Dong HM, et al. LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion. J Cell Biochem. 2017;118(12):4821–30.PubMedCrossRef
93.
Zurück zum Zitat Hou ZH, Xu XW, Fu XY, Zhou LD, Liu SP, Tan DM. Long non-coding RNA MALAT1 promotes angiogenesis and immunosuppressive properties of HCC cells by sponging miR-140. Am J Phys Cell Phys. 2020;318(3):C649–C63.CrossRef Hou ZH, Xu XW, Fu XY, Zhou LD, Liu SP, Tan DM. Long non-coding RNA MALAT1 promotes angiogenesis and immunosuppressive properties of HCC cells by sponging miR-140. Am J Phys Cell Phys. 2020;318(3):C649–C63.CrossRef
94.
Zurück zum Zitat Cao J, Dong R, Jiang L, Gong Y, Yuan M, You J, et al. LncRNA-MM2P identified as a modulator of macrophage M2 polarization. Cancer Immunol Res. 2019;7(2):292–305.PubMedCrossRef Cao J, Dong R, Jiang L, Gong Y, Yuan M, You J, et al. LncRNA-MM2P identified as a modulator of macrophage M2 polarization. Cancer Immunol Res. 2019;7(2):292–305.PubMedCrossRef
95.
Zurück zum Zitat Fang J, Qiao F, Tu J, Xu J, Ding F, Liu Y, et al. High expression of long non-coding RNA NEAT1 indicates poor prognosis of human cancer. Oncotarget. 2017;8(28):45918–27.PubMedPubMedCentralCrossRef Fang J, Qiao F, Tu J, Xu J, Ding F, Liu Y, et al. High expression of long non-coding RNA NEAT1 indicates poor prognosis of human cancer. Oncotarget. 2017;8(28):45918–27.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Wang J, Zhao X, Guo Z, Ma X, Song Y, Guo Y. Regulation of NEAT1/miR-214-3p on the growth, migration and invasion of endometrial carcinoma cells. Arch Gynecol Obstet. 2017;295(6):1469–75.PubMedCrossRef Wang J, Zhao X, Guo Z, Ma X, Song Y, Guo Y. Regulation of NEAT1/miR-214-3p on the growth, migration and invasion of endometrial carcinoma cells. Arch Gynecol Obstet. 2017;295(6):1469–75.PubMedCrossRef
97.
Zurück zum Zitat Gao Y, Fang P, Li WJ, Zhang J, Wang GP, Jiang DF, et al. LncRNA NEAT1 sponges miR-214 to regulate M2 macrophage polarization by regulation of B7-H3 in multiple myeloma. Mol Immunol. 2020;117:20–8.PubMedCrossRef Gao Y, Fang P, Li WJ, Zhang J, Wang GP, Jiang DF, et al. LncRNA NEAT1 sponges miR-214 to regulate M2 macrophage polarization by regulation of B7-H3 in multiple myeloma. Mol Immunol. 2020;117:20–8.PubMedCrossRef
98.
Zurück zum Zitat Liu F, Zhang T, Zou S, Jiang B, Hua D. B7H3 promotes cell migration and invasion through the Jak2/Stat3/MMP9 signaling pathway in colorectal cancer. Mol Med Rep. 2015;12(4):5455–60.PubMedCrossRef Liu F, Zhang T, Zou S, Jiang B, Hua D. B7H3 promotes cell migration and invasion through the Jak2/Stat3/MMP9 signaling pathway in colorectal cancer. Mol Med Rep. 2015;12(4):5455–60.PubMedCrossRef
99.
Zurück zum Zitat Zhou YX, Zhao W, Mao LW, Wang YL, Xia LQ, Cao M, et al. Long non-coding RNA NIFK-AS1 inhibits M2 polarization of macrophages in endometrial cancer through targeting miR-146a. Int J Biochem Cell Biol. 2018;104:25–33.PubMedCrossRef Zhou YX, Zhao W, Mao LW, Wang YL, Xia LQ, Cao M, et al. Long non-coding RNA NIFK-AS1 inhibits M2 polarization of macrophages in endometrial cancer through targeting miR-146a. Int J Biochem Cell Biol. 2018;104:25–33.PubMedCrossRef
100.
Zurück zum Zitat Li Y, Zhao L, Shi B, Ma S, Xu Z, Ge Y, et al. Functions of miR-146a and miR-222 in tumor-associated macrophages in breast cancer. Sci Rep. 2015;5:18648.PubMedPubMedCentralCrossRef Li Y, Zhao L, Shi B, Ma S, Xu Z, Ge Y, et al. Functions of miR-146a and miR-222 in tumor-associated macrophages in breast cancer. Sci Rep. 2015;5:18648.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Castellano JJ, Navarro A, Vinolas N, Marrades RM, Moises J, Cordeiro A, et al. LincRNA-p21 impacts prognosis in resected non-small cell lung cancer patients through angiogenesis regulation. J Thorac Oncol. 2016;11(12):2173–82.PubMedCrossRef Castellano JJ, Navarro A, Vinolas N, Marrades RM, Moises J, Cordeiro A, et al. LincRNA-p21 impacts prognosis in resected non-small cell lung cancer patients through angiogenesis regulation. J Thorac Oncol. 2016;11(12):2173–82.PubMedCrossRef
102.
Zurück zum Zitat Wu G, Cai J, Han Y, Chen J, Huang ZP, Chen C, et al. LincRNA-p21 regulates neointima formation, vascular smooth muscle cell proliferation, apoptosis, and atherosclerosis by enhancing p53 activity. Circulation. 2014;130(17):1452–65.PubMedPubMedCentralCrossRef Wu G, Cai J, Han Y, Chen J, Huang ZP, Chen C, et al. LincRNA-p21 regulates neointima formation, vascular smooth muscle cell proliferation, apoptosis, and atherosclerosis by enhancing p53 activity. Circulation. 2014;130(17):1452–65.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Li L, Ng DS, Mah WC, Almeida FF, Rahmat SA, Rao VK, et al. A unique role for p53 in the regulation of M2 macrophage polarization. Cell Death Differ. 2015;22(7):1081–93.PubMedCrossRef Li L, Ng DS, Mah WC, Almeida FF, Rahmat SA, Rao VK, et al. A unique role for p53 in the regulation of M2 macrophage polarization. Cell Death Differ. 2015;22(7):1081–93.PubMedCrossRef
104.
Zurück zum Zitat Zhou L, Tian Y, Guo F, Yu B, Li J, Xu H, et al. LincRNA-p21 knockdown reversed tumor-associated macrophages function by promoting MDM2 to antagonize* p53 activation and alleviate breast cancer development. Cancer Immunol Immunother. 2020;69(5):835–46.PubMedCrossRef Zhou L, Tian Y, Guo F, Yu B, Li J, Xu H, et al. LincRNA-p21 knockdown reversed tumor-associated macrophages function by promoting MDM2 to antagonize* p53 activation and alleviate breast cancer development. Cancer Immunol Immunother. 2020;69(5):835–46.PubMedCrossRef
105.
Zurück zum Zitat Liang ZX, Liu HS, Wang FW, Xiong L, Zhou C, Hu T, et al. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death Dis. 2019;10(11):829.PubMedPubMedCentralCrossRef Liang ZX, Liu HS, Wang FW, Xiong L, Zhou C, Hu T, et al. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death Dis. 2019;10(11):829.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Zhang Y, Tang L. Inhibition of breast cancer cell proliferation and tumorigenesis by long non-coding RNA RPPH1 down-regulation of miR-122 expression. Cancer Cell Int. 2017;17:109.PubMedPubMedCentralCrossRef Zhang Y, Tang L. Inhibition of breast cancer cell proliferation and tumorigenesis by long non-coding RNA RPPH1 down-regulation of miR-122 expression. Cancer Cell Int. 2017;17:109.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Lei B, He A, Chen Y, Cao X, Zhang P, Liu J, et al. Long non-coding RNA RPPH1 promotes the proliferation, invasion and migration of human acute myeloid leukemia cells through down-regulating miR-330-5p expression. EXCLI J. 2019;18:824–37.PubMedPubMedCentral Lei B, He A, Chen Y, Cao X, Zhang P, Liu J, et al. Long non-coding RNA RPPH1 promotes the proliferation, invasion and migration of human acute myeloid leukemia cells through down-regulating miR-330-5p expression. EXCLI J. 2019;18:824–37.PubMedPubMedCentral
108.
Zurück zum Zitat Yang D, Liu K, Fan L, Liang W, Xu T, Jiang W, et al. LncRNA RP11-361F15.2 promotes osteosarcoma tumorigenesis by inhibiting M2-Like polarization of tumor-associated macrophages of CPEB4. Cancer Lett. 2020;473:33–49.PubMedCrossRef Yang D, Liu K, Fan L, Liang W, Xu T, Jiang W, et al. LncRNA RP11-361F15.2 promotes osteosarcoma tumorigenesis by inhibiting M2-Like polarization of tumor-associated macrophages of CPEB4. Cancer Lett. 2020;473:33–49.PubMedCrossRef
109.
Zurück zum Zitat Kogure T, Yan IK, Lin WL, Patel T. Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: a mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer. 2013;4(7–8):261–72.PubMedPubMedCentralCrossRef Kogure T, Yan IK, Lin WL, Patel T. Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: a mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer. 2013;4(7–8):261–72.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Li X, Lei Y, Wu M, Li N. Regulation of macrophage activation and polarization by HCC-derived exosomal lncRNA TUC339. Int J Mol Sci. 2018;19(10). Li X, Lei Y, Wu M, Li N. Regulation of macrophage activation and polarization by HCC-derived exosomal lncRNA TUC339. Int J Mol Sci. 2018;19(10).
111.
Zurück zum Zitat Liu A, Liu L, Lu H. LncRNA XIST facilitates proliferation and epithelial-mesenchymal transition of colorectal cancer cells through targeting miR-486-5p and promoting neuropilin-2. J Cell Physiol. 2019;234(8):13747–61.PubMedCrossRef Liu A, Liu L, Lu H. LncRNA XIST facilitates proliferation and epithelial-mesenchymal transition of colorectal cancer cells through targeting miR-486-5p and promoting neuropilin-2. J Cell Physiol. 2019;234(8):13747–61.PubMedCrossRef
112.
Zurück zum Zitat Cheng Y, Chang Q, Zheng B, Xu J, Li H, Wang R. LncRNA XIST promotes the epithelial to mesenchymal transition of retinoblastoma via sponging miR-101. Eur J Pharmacol. 2019;843:210–6.PubMedCrossRef Cheng Y, Chang Q, Zheng B, Xu J, Li H, Wang R. LncRNA XIST promotes the epithelial to mesenchymal transition of retinoblastoma via sponging miR-101. Eur J Pharmacol. 2019;843:210–6.PubMedCrossRef
113.
Zurück zum Zitat Liu JL, Zhang WQ, Zhao M, Huang MY. Upregulation of long noncoding RNA XIST is associated with poor prognosis in human cancers. J Cell Physiol. 2019;234(5):6594–600.PubMedCrossRef Liu JL, Zhang WQ, Zhao M, Huang MY. Upregulation of long noncoding RNA XIST is associated with poor prognosis in human cancers. J Cell Physiol. 2019;234(5):6594–600.PubMedCrossRef
114.
Zurück zum Zitat Sun Y, Xu J. TCF-4 regulated lncRNA-XIST promotes M2 polarization of macrophages and is associated with lung cancer. Onco Targets Ther. 2019;12:8055–62.PubMedPubMedCentralCrossRef Sun Y, Xu J. TCF-4 regulated lncRNA-XIST promotes M2 polarization of macrophages and is associated with lung cancer. Onco Targets Ther. 2019;12:8055–62.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Ling H, Girnita L, Buda O, Calin GA. Non-coding RNAs: the cancer genome dark matter that matters! Clin Chem Lab Med. 2017;55(5):705–14.PubMedCrossRef Ling H, Girnita L, Buda O, Calin GA. Non-coding RNAs: the cancer genome dark matter that matters! Clin Chem Lab Med. 2017;55(5):705–14.PubMedCrossRef
116.
Zurück zum Zitat Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6(11):857–66.PubMedCrossRef Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6(11):857–66.PubMedCrossRef
117.
Zurück zum Zitat Xia X, Zhang K, Cen G, Jiang T, Cao J, Huang K, et al. MicroRNA-301a-3p promotes pancreatic cancer progression via negative regulation of SMAD4. Oncotarget. 2015;6(25):21046–63.PubMedPubMedCentralCrossRef Xia X, Zhang K, Cen G, Jiang T, Cao J, Huang K, et al. MicroRNA-301a-3p promotes pancreatic cancer progression via negative regulation of SMAD4. Oncotarget. 2015;6(25):21046–63.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Lu X, Li X, He Q, Gao J, Gao Y, Liu B, et al. miR-142-3p regulates the formation and differentiation of hematopoietic stem cells in vertebrates. Cell Res. 2013;23(12):1356–68.PubMedPubMedCentralCrossRef Lu X, Li X, He Q, Gao J, Gao Y, Liu B, et al. miR-142-3p regulates the formation and differentiation of hematopoietic stem cells in vertebrates. Cell Res. 2013;23(12):1356–68.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Trivedi M, Talekar M, Shah P, Ouyang Q, Amiji M. Modification of tumor cell exosome content by transfection with wt-p53 and microRNA-125b expressing plasmid DNA and its effect on macrophage polarization. Oncogenesis. 2016;5(8):e250.PubMedPubMedCentralCrossRef Trivedi M, Talekar M, Shah P, Ouyang Q, Amiji M. Modification of tumor cell exosome content by transfection with wt-p53 and microRNA-125b expressing plasmid DNA and its effect on macrophage polarization. Oncogenesis. 2016;5(8):e250.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Syed SN, Frank AC, Raue R, Brune B. MicroRNA-A tumor trojan horse for tumor-associated macrophages. Cells. 2019;8(12). Syed SN, Frank AC, Raue R, Brune B. MicroRNA-A tumor trojan horse for tumor-associated macrophages. Cells. 2019;8(12).
121.
Zurück zum Zitat Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, et al. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019;38(26):5158–73.PubMedCrossRef Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, et al. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene. 2019;38(26):5158–73.PubMedCrossRef
122.
Zurück zum Zitat 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(31):E2110–6.PubMedPubMedCentralCrossRef 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(31):E2110–6.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Patel SA, Gooderham NJ. IL6 mediates immune and colorectal cancer cell cross-talk via miR-21 and miR-29b. Mol Cancer Res. 2015;13(11):1502–8.PubMedCrossRef Patel SA, Gooderham NJ. IL6 mediates immune and colorectal cancer cell cross-talk via miR-21 and miR-29b. Mol Cancer Res. 2015;13(11):1502–8.PubMedCrossRef
124.
Zurück zum Zitat van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro-Oncology. 2016;18(1):58–69.PubMedCrossRef van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro-Oncology. 2016;18(1):58–69.PubMedCrossRef
125.
Zurück zum Zitat Yang ZY, Yang F, Zhang YL, Liu B, Wang M, Hong X, et al. LncRNA-ANCR down-regulation suppresses invasion and migration of colorectal cancer cells by regulating EZH2 expression. Cancer Biomark. 2017;18(1):95–104.PubMedCrossRef Yang ZY, Yang F, Zhang YL, Liu B, Wang M, Hong X, et al. LncRNA-ANCR down-regulation suppresses invasion and migration of colorectal cancer cells by regulating EZH2 expression. Cancer Biomark. 2017;18(1):95–104.PubMedCrossRef
126.
Zurück zum Zitat Li Z, Hou P, Fan D, Dong M, Ma M, Li H, et al. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer. Cell Death Differ. 2017;24(1):59–71.PubMedCrossRef Li Z, Hou P, Fan D, Dong M, Ma M, Li H, et al. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer. Cell Death Differ. 2017;24(1):59–71.PubMedCrossRef
127.
Zurück zum Zitat Wang S, Lan F, Xia Y. lncRA ANCR inhibits non-small cell lung cancer cell migration and invasion by inactivating TGF-beta pathway. Med Sci Monit. 2018;24:6002–9.PubMedPubMedCentralCrossRef Wang S, Lan F, Xia Y. lncRA ANCR inhibits non-small cell lung cancer cell migration and invasion by inactivating TGF-beta pathway. Med Sci Monit. 2018;24:6002–9.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Idogawa M, Ohashi T, Sasaki Y, Nakase H, Tokino T. Long non-coding RNA NEAT1 is a transcriptional target of p53 and modulates p53-induced transactivation and tumor-suppressor function. Int J Cancer. 2017;140(12):2785–91.PubMedCrossRef Idogawa M, Ohashi T, Sasaki Y, Nakase H, Tokino T. Long non-coding RNA NEAT1 is a transcriptional target of p53 and modulates p53-induced transactivation and tumor-suppressor function. Int J Cancer. 2017;140(12):2785–91.PubMedCrossRef
129.
Zurück zum Zitat Kwok ZH, Roche V, Chew XH, Fadieieva A, Tay Y. A non-canonical tumor suppressive role for the long non-coding RNA MALAT1 in colon and breast cancers. Int J Cancer. 2018;143(3):668–78.PubMedCrossRef Kwok ZH, Roche V, Chew XH, Fadieieva A, Tay Y. A non-canonical tumor suppressive role for the long non-coding RNA MALAT1 in colon and breast cancers. Int J Cancer. 2018;143(3):668–78.PubMedCrossRef
130.
Zurück zum Zitat Shi D, Wu F, Gao F, Qing X, Shao Z. Prognostic value of long non-coding RNA CCAT1 expression in patients with cancer: a meta-analysis. PLoS One. 2017;12(6):e0179346.PubMedPubMedCentralCrossRef Shi D, Wu F, Gao F, Qing X, Shao Z. Prognostic value of long non-coding RNA CCAT1 expression in patients with cancer: a meta-analysis. PLoS One. 2017;12(6):e0179346.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Zhang C, Han X, Yang L, Fu J, Sun C, Huang S, et al. Circular RNA circPPM1F modulates M1 macrophage activation and pancreatic islet inflammation in type 1 diabetes mellitus. Theranostics. 2020;10(24):10908–24.PubMedPubMedCentralCrossRef Zhang C, Han X, Yang L, Fu J, Sun C, Huang S, et al. Circular RNA circPPM1F modulates M1 macrophage activation and pancreatic islet inflammation in type 1 diabetes mellitus. Theranostics. 2020;10(24):10908–24.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Foss FM, Querfeld C, Kim YH, Pinter-Brown LC, William BM, Porcu P, et al. Ph 1 study of MRG-106, an inhibitor of miR-155, in CTCL. J Clin Oncol. 2018;36(15_suppl):2511.CrossRef Foss FM, Querfeld C, Kim YH, Pinter-Brown LC, William BM, Porcu P, et al. Ph 1 study of MRG-106, an inhibitor of miR-155, in CTCL. J Clin Oncol. 2018;36(15_suppl):2511.CrossRef
133.
Zurück zum Zitat van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386–96.PubMedCrossRef van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386–96.PubMedCrossRef
134.
Zurück zum Zitat Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368(18):1685–94.PubMedCrossRef Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368(18):1685–94.PubMedCrossRef
135.
Zurück zum Zitat Cortez MA, Anfossi S, Ramapriyan R, Menon H, Atalar SC, Aliru M, et al. Role of miRNAs in immune responses and immunotherapy in cancer. Genes Chromosom Cancer. 2019;58(4):244–53.PubMedCrossRef Cortez MA, Anfossi S, Ramapriyan R, Menon H, Atalar SC, Aliru M, et al. Role of miRNAs in immune responses and immunotherapy in cancer. Genes Chromosom Cancer. 2019;58(4):244–53.PubMedCrossRef
136.
Zurück zum Zitat Shah MY, Ferrajoli A, Sood AK, Lopez-Berestein G, Calin GA. microRNA therapeutics in cancer - an emerging concept. EBioMedicine. 2016;12:34–42.PubMedPubMedCentralCrossRef Shah MY, Ferrajoli A, Sood AK, Lopez-Berestein G, Calin GA. microRNA therapeutics in cancer - an emerging concept. EBioMedicine. 2016;12:34–42.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Smolle MA, Calin HN, Pichler M, Calin GA. Noncoding RNAs and immune checkpoints-clinical implications as cancer therapeutics. FEBS J. 2017;284(13):1952–66.PubMedCrossRef Smolle MA, Calin HN, Pichler M, Calin GA. Noncoding RNAs and immune checkpoints-clinical implications as cancer therapeutics. FEBS J. 2017;284(13):1952–66.PubMedCrossRef
138.
Zurück zum Zitat Dragomir M, Chen B, Calin GA. Exosomal lncRNAs as new players in cell-to-cell communication. Transl Cancer Res. 2018;7(Suppl 2):S243–S52.PubMedCrossRef Dragomir M, Chen B, Calin GA. Exosomal lncRNAs as new players in cell-to-cell communication. Transl Cancer Res. 2018;7(Suppl 2):S243–S52.PubMedCrossRef
Metadaten
Titel
Non-coding RNAs regulation of macrophage polarization in cancer
verfasst von
Swati Mohapatra
Carlotta Pioppini
Bulent Ozpolat
George A. Calin
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2021
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-021-01313-x

Weitere Artikel der Ausgabe 1/2021

Molecular Cancer 1/2021 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.