Life Sciences: Rong Dong, Bo Zhang, Biqin Tan, Nengming Lin

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Life Sciences 266 (2021) 118895

Contents lists available at ScienceDirect

Life Sciences
journal homepage: www.elsevier.com/locate/lifescie

Review article

Long non-coding RNAs as the regulators and targets of macrophage


M2 polarization
Rong Dong a, Bo Zhang b, Biqin Tan a, Nengming Lin a, b, *
a
Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s
Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
b
Translational Medicine Research Center, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 31006, China

A R T I C L E I N F O A B S T R A C T

Keywords: Macrophages are immune cells with high heterogeneity and plasticity. M2 polarization is one extreme of the
Long non-coding RNAs well-established phenotypes of macrophage polarization, and involves in diverse biological processes. The po­
M2 polarization larization process is initiated at the command of numerous components. Long non-coding RNAs (lncRNAs) are
Macrophages
RNAs longer than 200 nucleotides with limited protein-coding capacity. Recent studies have revealed a newly
Antisense oligonucleotides
found subset of lncRNAs engaged in the M2 polarization and their potent and multifunctional roles in developing
CRISPR interference
diseases. By interfering with specific signaling pathways and altering the active mode, acting as the sponges of
microRNAs or decoys of transcription factors, lncRNAs prompted macrophages to an M2 phenotype. Further,
lncRNAs can bind to the genome to regulate the chromatin dynamics or work as a platform for protein complexes
tether. Exosomal lncRNAs can also orchestrate the polarization in a paracrine way. To make it easier to interpret
the roles of lncRNAs in the M2 polarization, we review the reported lncRNAs according to the underlying
mechanisms. Moreover, we discuss the possibilities of targeting macrophages’ M2 polarization using the oli­
gonucleotides drugs or clustered regularly interspaced palindromic repeats (CRISPR) technologies to provoke
wisdom on the therapeutic strategies.

1. Introduction interleukin-13 (IL-13) may induce alternatively activated macrophages,


also known as M2 macrophages, which are involved in the anti-
Macrophages are crucial antigen-presenting cells with phenotypic inflammatory factors’ generation and tissue repair [7]. Usually, M2
and functional plasticity in the immune system [1]. Versatile polarized macrophage can promote inflammation reduction, and wound healing
macrophages could exert different functions depending on the circum­ [8,9]. When infiltrated into the tumor microenvironment, macrophages
stances [2,3]. Macrophages stimulated by bacterial lipopolysaccharide are often called tumor-associated macrophages (TAMs), which are often
(LPS) or interferon-γ (IFN-γ) are the so-called classically activated considered to be synonymous with M2 like and can promote tumori­
macrophages, or M1 macrophages, which promote the production of genesis, proliferation, metastasis, and angiogenesis [10–12]. Targeting
pro-inflammatory factors [4–6]. However, interleukin-4 (IL-4) or the M2 macrophages or the regulators of M2 polarization is considered a

Abbreviations: LncRNAs, long non-coding RNAs; LPS, lipopolysaccharide; IFN-γ, interferon-γ; IL-4, interleukin-4; IL-13, interleukin-13; TAMs, tumor associated
macrophages; JAK2, Janus Kinase 2; STAT, signal transducer and activator of transcription; SNHG20, small nucleolar RNA host gene 20; EMT, epithelial mesen­
chymal transition; HMGB1, high mobility group box 1 protein; USP11, ubiquitin specific protease 11; TLRs, toll like receptors; FISH, fluorescence in situ hybridi­
zation; MIR155HG, miRNA 155 host gene; COPD, chronic obstructive pulmonary disease; NF-κB, nuclear factor kappa-B; IL-10, interleukin-10; NSCLC, non-small cell
lung cancer; NECAB3, N-terminal EF-hand calcium binding protein 3; CCAT1: HIF-1α, hypoxia inducible factor 1 subunit alpha; PRC2, polycomb-group proteins; T-
UCRs, transcribed ultra-conserved RNAs; β-TrCP, beta-transducin repeat containing protein; GAS5, growth arrest-specific 5; IRF4, interferon regulatory factor 4;
EZH2, enhancer of zeste homolog 2; H3K27me3, trimethylation of lysine 27 on histone 3; SOCS3, suppressor of cytokine signaling 3; HCC, hepatocellular carcinoma;
MALAT1, metastasis associated in lung adenocarcinoma; COX-2, cyclooxygenase-2; ASOs, antisense oligonucleotides; CRISPRs, clustered regularly interspaced
palindromic repeats; crRNA, CRISPR-derived RNA; tracrRNA, trans-activating RNA; sgRNA, singleguide RNA; DETECTR, DNA endonuclease-targeted CRISPR trans
reporter; PAM, protospacer adjacent motif; PPAR-γ, peroxisome proliferators-activated receptor-γ.
* Corresponding author at: Room 207, No. 5 Building, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of
Medicine, Hangzhou, China.
E-mail address: lnm1013@zju.edu.cn (N. Lin).

https://doi.org/10.1016/j.lfs.2020.118895
Received 9 September 2020; Received in revised form 16 November 2020; Accepted 4 December 2020
Available online 10 December 2020
0024-3205/© 2020 Elsevier Inc. All rights reserved.
R. Dong et al. Life Sciences 266 (2021) 118895

new approach to conquer the immunological diseases [13]. two protein-coding genes. (5) Intronic lncRNAs are transcribed from the
Precise regulation of the polarization of macrophages is a critical intron region of the genome. As the research goes deepen, we must
step to arouse the specific function of the immune system [14]. The realize that lncRNAs can function independently of their genome loci’s
majority of studies have focused on the coding genes, transcription DNA sequence.
factors, and pathways in these processes. Studies with macrophages Herein we review our current understanding of lncRNA functions.
from the IL-4 receptor-deficient mice were found to lack M2 macrophage Firstly, lncRNAs can interfere with specific signaling pathways and alter
development in mouse models of Th2 cell-mediated inflammation [15]. the active mode [32]. They can also act as the sponges of microRNAs,
The phenotype that defines M2 is characterized by the increased pro­ decoys of transcription factors, or transcriptional coactivators or co­
duction of IL-10, TGF- TGF-β, Arg-1, Fizz1, and CCL18. These charac­ repressors [33]. Third, they can bind to the genome to regulate chro­
teristics are promoted by the dimerization of IL-4Rα and subsequent matin dynamics, histone modifications, methylation or downstream
JAK-mediated tyrosine phosphorylation of STAT6 [16]. On the other gene expression [34–37]. Finally, lncRNAs could alter the localization,
hand, the STAT6 can bind to PPAR-γ, a master regulator of lipid meta­ transportation of proteins, or the protein complex formation.
bolism in macrophages, and is known to inhibit the pro-inflammatory
gene expression and act as a cofactor in PPAR-γ mediated gene regula­ 3. LncRNAs in M2 polarization
tion in metabolic dysfunction and diseases [17]. Overwhelming evi­
dence supports the role of the PI3K-Akt signaling pathway in M2 3.1. Signaling lncRNAs
polarization. PI3K generates phosphatidylinositol-3,4,5-triphosphate,
which activates the downstream protein kinase B- Akt. Akt has three LncRNAs are pivotal regulators in the macrophage polarization
distinct isoforms: Akt1, 2, and 3. Studies utilizing Akt isoforms-specific processes in response to intracellular or extracellular stimuli (Table 1).
deficient mice revealed that Akt1 promotes the polarization of M2 The commonly held view is that M2 polarization is driven by Th2 cy­
macrophages and inhibits the polarization of M1 macrophages, while tokines or chemokines, such as IL-4 or IL-13 [12]. These cues are thought
Akt2 vice versa [18]. MicroRNA-155 (miR-155) and C/EBPβ play a vital to provoke the level change of lncRNAs and activate the JAK2-STAT6
role in regulating Akt-dependent macrophage polarization. Akt2 en­ pathway, MAPK-ERK/MEK/JNK pathway, and PI3K-Akt pathway to
hances the expression of miR-155, which down-regulates the expression promote the M2 polarization [14]. Many lncRNAs have been docu­
of C/EBPβ, and ultimately promotes the polarization of M1 macrophages mented to regulate the polarization in this manner (Fig. 1a).
[19]. One of these lncRNAs is small nucleolar RNA host gene 20
Comparatively, less is known about the non-coding RNAs’ functions (SNHG20), a 2183 bp transcript originated from the chromosome
of, especially the long non-coding RNAs (lncRNAs) [20]. LncRNAs are an 17q25.2. It was first reported in human hepatocellular carcinoma and
array of RNAs longer than 200 nucleotides with limited protein-coding then expanded to various malignancies [38]. SNHG20 could control the
potential [21–23]. Thanks to the advances of the next generation proliferation via the cell cycle-associated genes, e.g., Cyclin A1, to
sequencing technology, a series of lncRNAs are annotated and func­ promote the metastasis via the EMT signaling pathway [39]. Recently,
tionally analyzed well in the differentiation and maturation of various SNHG20 was suggested to be highly expressed in Kupffer cells, the
immune cells [24–26]. LncRNA-Morrbid was found to control the sur­ resident macrophages in the liver, during nonalcoholic fatty liver dis­
vival of short-lived myeloid cells via the cis-regulation of Bcl2l11 ease progression to hepatocellular carcinoma [40]. Gain and loss of
expression [27]. Lnc-DC promotes the dendritic cells by the C terminus function study of SNHG20 in Kupffer cells revealed that M2 polarization
of STAT3 to prevent the dephosphorylation of STAT3 Y705 by SHP1 induced and M1 polarization was deduced in RAW264.7 cells, respec­
[28]. The lnc-MC regulates the differentiation of monocyte to macro­ tively. Increased phosphorylation of STAT6, a classical mediator of M2
phage to some extent [29]. Systematic research has suggested that polarization, was observed when SNHG20 was overexpressed. STAT6
several lncRNAs are dynamically changing during the polarization of inhibitor AS1517499 efficiently decreased the promotion of M2 polari­
macrophages. But the annotation of lncRNAs is still far from clear, and as zation [41].
such, no existing comprehensive study exists has reviewed the role of Our previous work also determined another novel murine lncRNA,
lncRNAs specifically involved in the process of macrophage M2 polari­ 5730422e09Rik, which we referred to as lncRNA MM2P, was involved
zation. Consequentially, here we review the recent advances in eluci­ in the M2 polarization [42]. MM2P is a 1715 bp transcript from chro­
dation of various lncRNAs and their mechanisms within the M2 mosome 5, between gene HMGB1 and USP11 [43,44]. When we started
polarization. In addition, we discuss the possibility of applying antisense the mechanism study, we assumed MM2P might be associated with
nucleic acid drugs to intervene in the process to cure related diseases. HMGB1 protein, since HMGB1 was a key regulator in initiating and
maintaining of chronic inflammation through TLRs signaling pathways
2. Overview of lncRNAs and chromosomal binding. But the FISH assay revealed MM2P was
predominantly localized in the cytosol. Knockdown MM2P affected the
Comprehensive analyses of the human genome have shown that phosphorylation of STAT6 but not the upstream kinase JAK2. When
although large numbers of RNA transcripts are transcribed into proteins, applying the chemical agent Na3VO4, it can partially reverse the
they only take up a very small portion of the transcripts. The rest of the dephosphorylation of STAT6 induced by MM2P. Therefore, we supposed
transcripts are non-coding RNAs, including miRNAs, long non-coding MM2P has participated in the dephosphorylation of STAT6, but the
RNAs, piRNAs, circRNAs, etc. [30]. Long non-coding RNAs are always further remains exclusive. The biological analysis revealed that MM2P
longer than 200 nt with little ability to encode proteins or polypeptides. promoted the tumorigenesis, angiogenesis of osteosarcoma.
Unlike the message RNAs, lncRNAs may have polyadenylic acid tails or MiRNA-155 is a micro non-coding RNA involved in immunity,
not. Compared with the protein-coding genes, they have higher tissue inflammation and tumors [45]. Its host gene was a long non-coding RNA
and organ specificity but less low conserved sequence among species. named MIR155HG [46]. Recently, MIR155HG was found to regulate
There are several ways to classify lncRNAs, one of the simple ways is macrophages’ polarization in chronic obstructive pulmonary disease
based on the genomic level organization [31]. In this aspect, they are (COPD) patients [47]. Overexpression of MIR155HG could increase the
divided into five groups: (1) sense lncRNAs, the transcription direction M1 but decrease the M2 polarization. Luciferase assay revealed that NF-
of this type of lncRNAs is the same as that of the neighborhood mRNAs. κB protein p65 increased the expression of MIR155HG through binding
(2) Antisense lncRNAs are those whose transcription direction is oppo­ to the upstream of the transcription stat site of MIR155HG.
site to that of the protein-coding genes. (3) Bidirectional lncRNAs can be
transcribed from the same or the opposite directions to the adjacent
mRNA transcription. (4) Intergenic lncRNAs are transcribed between

2
R. Dong et al. Life Sciences 266 (2021) 118895

Table 1
Summary of lncRNAs involved in the M2 polarization and mechanisms.
lncRNA Mechanism of action and Disease Cell type Ref.
function
SNHG20 Induce M2 polarization Nonalcoholic RAW264.7, [40]
through STAT6 fatty liver Liver
disease to nonparenchymal
hepatocellular cells
carcinoma
MIR155HG Knockdown of MIR-155HG Chronic PBMCs [47]
(MIR155 inhibits the polarization of obstructive
host gene) M1 macrophages and pulmonary
increase M2 macrophage disease
polarization (COPD)
MM2P Promote M2 polarization Osteosarcoma RAW264.7, [42]
through the BMDMs
dephosphorylation of STAT6
KCNQ1OT1 Function as a decoy of Aseptic RAW264.7, [50]
miR-21a-5p, regulate IL-10 loosening after BMDMs
expression, and ameliorate total joint
particle-induced replacement
NIFK-AS1 Inhibit M2 polarization via Endometrial PBMCs, TAMs, [51]
targeting miR-146a, thereby cancer THP-1
reducing the
estrogen-induced
proliferation, migration an d
invasion of endometrial
cancer cells
CCAT1 CCAT1 knockdown promote Prostate cancer RAW264.7, [52]
M2 polarization by PBMCs
upregulating miR-148a (not
a sponge)
RP11-361F Act as a competing Osteosarcoma RAW264.7 [54]
15.2 endogenous RNA against
miR-30c-5p and upregulate
CPEB4 expression,
promote proliferation,
migration, invasion and
M2-like polarization in
osteosarcoma
GNAS-AS1 Promote M2 polarization NSCLC THP-1, PBMCs [55]
and NSCLC progression via
inhibiting miR-4319, which
could target N-terminal

3
R. Dong et al. Life Sciences 266 (2021) 118895

EF-hand calcium binding


protein 3
linc-p21 Knockdown of lincRNA-p21 Breast cancer TAMs [56]
promote MDM2 to
antagonize p53 function,
tumor cell apoptosis,
migration, and invasion
uc.306 May target gene BTRC and Hepatocellular U937 [57]
involved in Wnt signaling carcinoma
pathway
GAS5 Promote the M1 Diabetic PBMCs [59]
polarization by upregulating wounds
STAT1, reduction of GAS5 RAW264.7
enhance the transition of Childhood Microglia from
M1 macrophages to M2 pneumonia mice
macrophages Multiple
act as a sponge for sclerosis
miR-455-5p to facilitate
SOCS3 expression
Suppress transcription of
TRF4 by recruiting the
PRC2 to inhibit microglial
M2 polarization
MALAT1 Promote the expression of Pre-eclampsia MSCs, THP-1 [66]
VEGF not only through
affecting the promoter but
also the miRNAs and Atherosclerosis
regulate the proliferation,
angiogenesis and
immunosuppressive
properties of MSCs
Exosomal MALAT1 derived
from oxLDL-treated
endothelial cells promoted
M2 polarization
LINC00662 Physically bind miR-15a, Hepatocellular THP-1 [65]
miR-16, miR-107, activate carcinoma
Wnt/β-catenin signaling in a
paracrine manner
CASC2c Interact with and repress Glioblastoma THP-1, BMDMs [68]
miR-338-3P multiforme
COX-2 Inhibit immune evasion and Hepatocellular RAW264.7 [69]
tumor growth by inhibiting carcinoma
the polarization of M2
macrophages
RPPH1 CRC cells-derived Colorectal PBMCs [71]
exosomes transport RPPH1 cancer
into macrophages and
mediate M2 polarization
TUC339 Required for IL-4 Hepatocellular THP-1, [70]
macrophages polarization carcinoma

RAW264.7: murine macrophage/monocyte like cell linage, originating from Abelson leukemia virus
transformed cell linage derived from BALB/c mice; PBMCs: peripheral blood mononuclear cells;
BMDMs: bone marrow-derived macrophages; THP-1: human acute monocytic leukemia cell line;
TAMs: tumor associated macrophages; U937: human histiocytic lymphoma cells; MSCs: mesen­
chymal stem cells.
Each of the color shading represents a subgroup of lncRNAs classified according to the lncRNA’
function. The lncRNAs in blue shading are signaling lncRNAs. The lncRNAs in pink shading are

4
R. Dong et al. Life Sciences 266 (2021) 118895

decoy lncRNAs. The lncRNAs in yellow shading are guide lncRNAs. The lncRNAs in pink shading are
scoffold lncRNAs. The last part are exosomal lncRNAs.

3.2. Decoy lncRNAs direct them to target genomic location and gene promoter.
LncRNA-GAS5 was suggested as a 630 bp long tumor suppressor
Another way for lncRNAs to participate in macrophage polarization lncRNA locus on the chromosome 1q25. It participated in the prolifer­
is to compete with other endogenous molecules [48,49]. They can ation, apoptosis, cell migration and cellular metabolism to promote the
sponge miRNAs or decoy transcription factors to change protein tran­ malignancy [58]. As research continues to deepen, the role of GAS5 in
scription or degradation. This kind of interference alters the polarization the immune system has become clear. Zhao and colleagues suggested
state and networks of macrophages as well (Fig. 1b). that GAS5 could suppress IRF4 transcription by binding PRC2 to inhibit
LncRNA KCNQ1OT1 is a bidirectional antisense long non-coding M2 polarization in the autoimmune encephalomyelitis animal model of
transcript locus on chromosome 12p15.5. It was reported to be a multiple sclerosis [59]. Binding between EZH2 and IRF4 promoter,
sponge of miR-370 in glioma tissues, a competing endogenous RNA of H3K27me3 and IRF4 promoter was attenuated by GAS5 (Fig. 1d).
miR-701-3p in osteoblast. The report showed that KCNQ1OT1 decoyed Although we considered GAS5 a scaffold lncRNA, it was reported to
the miR-21a-5p in macrophages to regulate the IL-10 expression, further promote M1 polarization by upregulating STAT1 as a signaling lncRNA
promoted M2 polarization, and ameliorated osteolysis [50]. in the diabetic wounds healing and acted as a sponge for miR-455-5p to
Zhou et al. reported that lncRNA NIFK-AS1, a 1267 bp-long antisense facilitate SOCS3 expression in the childhood pneumonia [60,61].
RNA locus on the chromosome 2q14.3, could regulate the M2 polari­
zation in endometrial cancer recently [51]. Bioinformatics analysis 3.5. Exosomal lncRNAs
revealed that NIFK-AS1 could target miR-146a and thus promote the
expression of Notch1 protein. LncRNA GNAS-AS1 is another antisense As previously described, lncRNAs might parent from macrophages’
long non-coding RNA from the chromosome 20q13.32. It was reported genome and they manipulated the polarization process. As the research
to increase in the TAMs from NSCLC patients and promote the NSCLC continues to deepen, lncRNAs from the nearby cells also can interfere
cell proliferation, migration and invasion by directly competitively with the macrophage polarization in a paracrine way [62].
binding of miR-431 with protein NECAB3. Exosomes are small extracellular vesicles released from cell mem­
Studies have reported other lncRNAs that could promote M2 polar­ branes with a size of about 30–100 nm. They can be biological depots
ization through miRNAs, such as lncRNA CCAT1 [52,53]. However, it carrying proteins, mRNAs or lncRNAs in various fluids and help
did not confirm the sponge or decoy role of CCAT1; therefore, the communicate between different cells [63]. Exosomal lncRNAs are
mechanism may need further work. Rp11-361F15.2 was also a lncRNA implicated in the M2 polarization as well (Fig. 1e).
that reported to promote the M2 polarization, as it was a competing Linc00662 was a 2085 bp long transcript originated from chromo­
ceRNA against miR-30c-5p in the osteosarcoma [54]. In NSCLC speci­ some 19q11. It could physically bind miR-15a, miR-16, and miR-107,
mens, GNAS-AS1 was found to promote M2 polarization via inhibiting which target WNT3A [64]. Tian and colleagues found that linc00662
miR-4319, which could target the N-terminal EF-hand calcium-binding could promote the HCC progression in an autocrine manner and pro­
protein3 [55]. mote the TAMs to a M2 polarization via the exosomal linc00662 in a
paracrine manner. Both biological effects were promoted by activating
3.3. Guide lncRNAs the Wnt/β-catenin signaling pathway [65].
Other reported exosomal lncRNAs included MALAT1 in pre-
Guide lncRNAs could bind to enzymes or protein complexes to eclampsia and atherosclerosis [66,67], CASC2c in glioblastoma multi­
activate downstream cascades. This kind of lncRNAs is seldom reported forme [68], COX-2 and TUC339 in hepatocellular carcinoma [69,70],
in macrophage polarization since the precise biological system associ­ and RPPH1 in colorectal cancer [71]. They were all reported to promote
ated with macrophage polarization is still being studied. Only a few the M2 polarization by secretory exosomes between different cell
were supposed to regulate the polarization in this way (Fig. 1c). populations.
LincRNA-p21 was about 3000 bp in length locus on the upstream of
protein p21, a cell-cycle and proliferation regulator, and regulated the 4. Therapeutic strategies targeting the macrophage polarization
p21 level. Other reports suggested that linc-p21 could interact with
proteins like MDM2, p53, HIF-1α, PRC2. A recent study reported that the Macrophages are the first line of the immune system. Abundant
linc-p21-MDM2-p53 axis was involved in the TAMs. LincRNA-p21 pro­ macrophages infiltrated in the milieu may elicit extensive responses
moted the function of TAMs by preventing the combination of MDM2 dependent on the polarization state. Non-coding RNAs take up about a
and p53. Once lincRNA-p21 was knockdown, MDM2 stayed in the nu­ significant portion of the whole genome. They decline the exponentially
cleus and NF-κB-STAT3 was activated to reeducate the TAMs to an M1 important role in the macrophage polarization processes and are rapidly
phenotype [56]. emerging as novel therapeutic targets because of their abundantly
Transcribed ultra-conserved RNAs (T-UCRs) are a novel subset of expressed properties and specific expression patterns [72–75]. We
long non-coding RNAs that are completely conserved across disparate would like to discuss two novel therapeutic strategies other than the
taxa. Increasing evidence showed that T-UCRs also regulate the traditional chemical agents targeting the macrophage polarization
macrophage polarization in disease development. T-UCR uc.306 was pathways.
one of the top differentially expressed T-UCRs in the M2 polarization
status [57]. Target gene prediction revised that uc.306 might be the E3 4.1. Antisense oligonucleotides interference
ligase of β-TrCP, an E3 ligase of β-catenin. β-Catenin is one of the key
regulators in the Wnt signaling pathway activation. Wnt signaling Small interference RNA technologies are classical means to study the
pathway can manipulate the activation status of macrophages. Luo and gene function according to the lab’s base complementary principle in
colleagues found that uc.306 might be upregulated in M1 macrophages the lab [76,77]. Now patients have the opportunities to benefit from
and lay a low profile in HBV-related HCC. these technologies as the applications go from bench to bedside [78–80].
RNA interference-mediated TMPRSS6 mRNA degradation improved the
3.4. Scaffold lncRNAs erythropoiesis and anemia in animal models, and later SLN124, a
GalNAc-siRNA conjugate targeting TMPRSS6, has been granted as an
Scaffold lncRNAs can interact with various protein complexes and orphan drug by the European medicines agency [81,82]. However,

5
R. Dong et al. Life Sciences 266 (2021) 118895

Fig. 1. Involvement and regulatory mechanisms of lncRNAs in M2 macrophage polarization. (a) lncRNAs can interfere with specific signaling pathways and alter the
active mode. (b) lncRNAs can act as the sponges of microRNAs, decoys of transcription factors or transcriptional coactivators or corepressors. (c) lncRNAs can bind to
the genome to regulate the chromatin dynamics, histone modifications, methylation or downstream gene expression. (d) lncRNAs could alter the localization,
transportation of proteins or the formation of the protein complex. (e) lncRNAs from the nearby cells also can interfere with the macrophage polarization in a
paracrine way.

6
R. Dong et al. Life Sciences 266 (2021) 118895

unlike protein-coding genes, lncRNAs may locate in the nucleus, which have been discovered. Although currently available technologies such as
is not easy to reach for siRNAs. In addition, the knockdown efficiency of small RNA interference and CRISPR-based libraries screen make it easier
siRNAs may not last long since the siRNAs are vulnerable to the RNases. to discover and target the lncRNAs, there are seldom oligonucleotide
Antisense oligonucleotides (ASOs) are synthetic single-stranded drugs specifically targeting lncRNAs modulating M2 polarization. While
nucleic acids complementary to their target and trigger RNase H- some researches based on oligonucleotide therapeutics may shed light
mediated degradation [83,84]. To avoid the disadvantages of siRNAs, on this problem.
ASOs must meet two criteria [85]. First, they should not be degraded by AZD9150 is a 16-nucleotide STAT3 ASO. It is designed to target and
endogenous nucleases to be sent to target cells to produce the most indirectly downregulate the expression of STAT3 mRNA and has pre­
significant effect. Presently, viral vectors are used to transfer to cells sented its efficacy in refractory/relapsed lymphoma and DLBCL [103].
[86,87]. But the side effects of viral vectors prompted researchers to As the JAK/STAT signaling pathway is a critical regulator in the
explore and develop non-viral transfer systems considering the safety macrophage polarization, studies with the connection between anti-
and clinical application [88]. In addition, the antisense nucleic acid cancer efficacy and the states of peripheral blood mononuclear cells
molecules must be able to approach the target tissue at a specific con­ would be further investigated. Other ASO drugs include an antisense
centration and penetrate the cell membrane into the cell to produce a oligonucleotide drug on lung fibrosis. NF-κB is a crucial nuclear tran­
therapeutic effect [89]. Thus, various chemical modifications and scription factor. It participates in the inflammatory response, immune
structural modifications to ASOs have been done to ensure their efficacy response, and cell apoptosis. The over-activation of NF-κB is found in
[90]. The first generation is to modify the backbone with phosphate; many human diseases such as rheumatoid arthritis, inflammatory dis­
thus, the phosphorothioates are the most widely used of this kind. Later, eases in the heart and brain. H. Saitoh et al. have revealed the effect of an
diverse chemical groups, like alkyl modifications at the 2′ position of the antisense oligonucleotide drug to the p65 subunit of NF-κB in the ARDS
ribose and locked nucleic acids, were introduced into the structure to mouse model [104,105]. The suppression of NF-κB in the alveolar
improve the binding affinity, enzyme resistance and pharmacokinetics macrophages resulted in the decreased secretion of IL-8. Yasunori et al.
profile. also reported the successful delivery of locked nucleic acid-containing
Gong et al. constructed specific ASOs targeted lncRNA MALAT1 to antisense oligonucleotides in the alveolar macrophages [106].
control breast cancer’s metastasis [91]. The ASOs were packed with a Genome-wide CRISPR screen has been used to identify functional
nucleus-targeting TAT peptide in Au nanoparticles to downregulate the non-coding RNAs in cell differentiation and development. Phagocytosis
expression of MALAT1. The TAT peptide could lead the ASOs into the is a significant skill for the macrophage to neutralize and terminate
nuclear, and Au nanoparticles could defeat the endonucleases and foreign pathogens. CRISPR-based library screen has revealed multiple
exonucleases. These kinds of modifications of ASOs reveal the possibil­ novel genes in the phagocytic processes. F. Imperatore et al. revealed
ities to target lncRNAs to cure diseases. that CRISPR/Cas9 mediated deletion of SIRT1 impacts the steady-state
and self-renewal in alveolar and peritoneal macrophages [107].
4.2. Clustered regularly interspaced palindromic repeats interference Knockout or the CRISPRi in-vivo models of lncRNA-Cox2 revealed that
the altered expression of inflammatory genes in lncRNA-Cox2 deficient
Gene-editing technology has reached its peak by unmasking the role macrophages and murine tissues confirmed its trans- and cis- manipu­
of the clustered regularly interspaced palindromic repeats (CRISPRs) lation of the neighbor gene Ptgs2 and its drastic influence in the immune
and its Cas9 nuclease enzyme in the bacteria. CRISPR-Cas9 system works signaling axes [108]. Experimental studies also confirmed that lncRNA-
as a prokaryotic adaptive immune system to purge phages and foreign ANRIL is capable of forming circANRIL. CRISPR-based deletion of cir­
plasmids [92]. The crRNA (CRISPR-derived RNA) can bind to the cANRIL isoforms controls the cell proliferation and apoptosis in mac­
tracrRNA (trans-activating RNA) through base pairing to form the rophages in atherosclerosis [109].
tracrRNA/crRNA complex, which guides the nuclease cas9 protein to the
target site to cut double-stranded DNA [93]. The cas-mediated double- 5. Conclusions
strain break will next activate the homology-directed repair or non-
homologous end joining pathway. By artificially designing these two Macrophages are in constant changes with two extreme polarization
types of RNAs, these can be transformed into a single-guide RNA states, respectively M1 and M2 macrophages. The M2 macrophages
(sgRNA), which is enough to guide cas9 dependent cleavage of DNA were promoters in various diseases. Distinguishing the pivotal regula­
[94]. Now it has been used as a potent gene manipulation tool to cure tors of macrophage polarization may be a significant step to cure dis­
diseases [95]. eases. Though the studies of lncRNAs are still in its infancy, increasing
CRISPR interference could help screen genome-wide coding and non- numbers of lncRNAs were investigated in the macrophages’ M2 polari­
coding genes functions, including lncRNAs [96]. S. John et al. recently zation process [110,111]. Their multifunctional capabilities and het­
used CRISPRi technology to screen the potential targets of lncRNAs in erogeneous working mechanisms make them potential targets for the
the glioblastoma cells, and nine candidates were found to be relevant to manipulation of diseases [112].
the sensitivity of glioma cells to radiation therapy [97]. ASOs were Gene therapy has been an attractive and alternative approach to cure
applied to this study to reveal the role of lncGRS-1, which could selec­ diseases, especially the monogenic human genetic diseases, due to gene-
tively inhibit the glioma cells’ growth and resistance to radiation. editing technologies’ breakthrough. Discrepancies between the RNAi-
Systematically studies continue, more cas-nucleases were found to and CRISPRi-based therapies make it comparatively easy to screen and
expand the precise gene editing tools. Cas12a was found to cleave single- target the lncRNAs in the complicated biological reactions in versatile
stranded DNA as well as double-stranded DNA [98]. Combining cas12a, modality. Oligonucleotide drugs can be used to modulate the non-
a method named DETECTR (DNA endonuclease-targeted CRISPR trans coding gene expression, as well as the protein-coding genes [83,113].
reporter) was developed to detect the human papillomavirus in patient They work through the complementary Watson-Crick base pairing
samples [99]. Cas13a can also be engineered for RNA knockdown and principle and so interrogate the presented target. Though the cross-
binding [100,101]. Cas14 can bind and cleave single-stranded DNA in­ species conservation of lncRNAs is relatively low, the tissue-specific
dependent of PAM sequences [102]. All these gene editing technologies lncRNA expression shows strong conservation across species, and
are revolutionary weapons which can be applied to target the lncRNAs. neighborhood gene locus is quite conserved. The length of ASOs
partially improves their specificity because 16 to 20 nucleotides can
4.3. Oligonucleotide drugs in macrophages uniquely bind to the target. After binding to the target RNA, they can
regulate the RNA by promoting the fragmentation or degradation of
To date, an increasing number of lncRNAs in the M2 polarization target RNA or sterically blocking the target [114,115]. As of January

7
R. Dong et al. Life Sciences 266 (2021) 118895

2020, ten oligonucleotide drugs have received approval from the FDA, [19] F. Ma, F. Liu, L. Ding, et al., Anti-inflammatory effects of curcumin are associated
with down regulating microRNA-155 in LPS-treated macrophages and mice,
and over 100 oligonucleotides drugs are in clinical trials [116]. Five of
Pharm. Biol. 55 (2017) 1263–1273.
the ten approved drugs target the liver organ since it is pivotal in drug [20] S. Roy, S. Schmeier, E. Arner, et al., Redefining the transcriptional regulatory
metabolism and detoxification [117]. Kupffer cells are resident macro­ dynamics of classically and alternatively activated macrophages by deepCAGE
phages in the liver and account for most uptake of the ASO drugs. The transcriptomics, Nucleic Acids Res. 43 (2015) 6969–6982.
[21] K.R. Sigdel, A. Cheng, Y. Wang, et al., The emerging functions of long noncoding
knockout of scavenger receptor A in Kupffer cells results in about 25% RNA in immune cells: autoimmune diseases, J Immunol Res 2015 (2015),
reduction of the accumulation and a 50% decrease of ASO drugs [118]. 848790.
Targeting macrophages and targeting the lncRNAs using the nucleic [22] A. Bhan, M. Soleimani, S.S. Mandal, Long noncoding RNA and cancer: a new
paradigm, Cancer Res. 77 (2017) 3965–3981.
acid-based therapeutics may hold therapeutic potential. [23] J.D. Ransohoff, Y. Wei, P.A. Khavari, The functions and unique features of long
In conclusion, lncRNAs exhibit tremendous effects on the macro­ intergenic non-coding RNA, Nat. Rev. Mol. Cell Biol. 19 (2018) 143–157.
phage differentiation and polarization, providing other druggable tar­ [24] F. Kopp, J.T. Mendell, Functional classification and experimental dissection of
long noncoding RNAs, Cell 172 (2018) 393–407.
gets for the interference of macrophage engaged disorders. [25] P.J. Batista, H.Y. Chang, Long noncoding RNAs: cellular address codes in
development and disease, Cell 152 (2013) 1298–1307.
Funding [26] K.C. Wang, H.Y. Chang, Molecular mechanisms of long noncoding RNAs, Mol.
Cell 43 (2011) 904–914.
[27] J.J. Kotzin, S.P. Spencer, S.J. McCright, et al., The long non-coding RNA Morrbid
This work was supported by the Zhejiang Provincial Natural Science regulates Bim and short-lived myeloid cell lifespan, Nature 537 (2016) 239–243.
Foundation (grant number LQ19H160012 to Rong Dong). [28] P. Wang, Y. Xue, Y. Han, et al., The STAT3-binding long noncoding RNA lnc-DC
controls human dendritic cell differentiation, Science 344 (2014) 310–313.
[29] M.T. Chen, H.S. Lin, C. Shen, et al., PU.1-regulated long noncoding RNA lnc-MC
CRediT authorship contribution statement controls human monocyte/macrophage differentiation through interaction with
microRNA 199a-5p, Mol. Cell. Biol. 35 (2015) 3212–3224.
[30] S. Djebali, C.A. Davis, A. Merkel, et al., Landscape of transcription in human cells,
Conceptualization, Rong Dong. and Biqin Tan.; methodology, Rong
Nature 489 (2012) 101–108.
Dong.; investigation, Rong Dong.; resources, Biqin Tan.; wri­ [31] I. Ulitsky, D.P. Bartel, lincRNAs: genomics, evolution, and mechanisms, Cell 154
ting—original draft preparation, Rong Dong.; writing—review and (2013) 26–46.
editing, Biqin Tan and Bo Zhang.; visualization, Rong Dong.; supervi­ [32] D. Zhou, C. Huang, Z. Lin, et al., Macrophage polarization and function with
emphasis on the evolving roles of coordinated regulation of cellular signaling
sion, Nengming Lin.; project administration, Nengming Lin.; funding pathways, Cell. Signal. 26 (2014) 192–197.
acquisition, Rong Dong. All authors have read and agreed to the pub­ [33] R.W. Yao, Y. Wang, L.L. Chen, Cellular functions of long noncoding RNAs, Nat.
lished version of the manuscript. Cell Biol. 21 (2019) 542–551.
[34] Y.G. Chen, A.T. Satpathy, H.Y. Chang, Gene regulation in the immune system by
long noncoding RNAs, Nat. Immunol. 18 (2017) 962–972.
Declaration of competing interest [35] K. Mishra, C. Kanduri, Understanding long noncoding RNA and chromatin
interactions: what we know so far, Non-coding RNA 5 (2019).
[36] L. Nair, H. Chung, U. Basu, Regulation of long non-coding RNAs and genome
The authors declare that they have no competing interests. dynamics by the RNA surveillance machinery, Nat. Rev. Mol. Cell Biol. 21 (2020)
123–136.
References [37] F. Zhang, L. Zhang, C. Zhang, Long noncoding RNAs and tumorigenesis: genetic
associations, molecular mechanisms, and therapeutic strategies, Tumour biology:
the journal of the International Society for Oncodevelopmental Biology and
[1] C. Varol, A. Mildner, S. Jung, Macrophages: development and tissue
Medicine 37 (2016) 163–175.
specialization, Annu. Rev. Immunol. 33 (2015) 643–675.
[38] D. Zhang, C. Cao, L. Liu, et al., Up-regulation of LncRNA SNHG20 predicts poor
[2] S.C. Funes, M. Rios, J. Escobar-Vera, et al., Implications of macrophage
prognosis in hepatocellular carcinoma, J. Cancer 7 (2016) 608–617.
polarization in autoimmunity, Immunology 154 (2018) 186–195.
[39] J. Liu, C. Lu, M. Xiao, et al., Long non-coding RNA SNHG20 predicts a poor
[3] L.C. Davies, S.J. Jenkins, J.E. Allen, et al., Tissue-resident macrophages, Nat.
prognosis for HCC and promotes cell invasion by regulating the epithelial-to-
Immunol. 14 (2013) 986–995.
mesenchymal transition, Biomedicine & pharmacotherapy = Biomedecine &
[4] A. Shapouri-Moghaddam, S. Mohammadian, H. Vazini, et al., Macrophage
pharmacotherapie 89 (2017) 857–863.
plasticity, polarization, and function in health and disease, J. Cell. Physiol. 233
[40] B. Wang, X. Li, W. Hu, et al., Silencing of lncRNA SNHG20 delays the progression
(2018) 6425–6440.
of nonalcoholic fatty liver disease to hepatocellular carcinoma via regulating liver
[5] C.H. Lu, C.Y. Lai, D.W. Yeh, et al., Involvement of M1 macrophage polarization in
Kupffer cells polarization, IUBMB Life 71 (2019) 1952–1961.
endosomal toll-like receptors activated psoriatic inflammation, Mediat. Inflamm.
[41] K. Binnemars-Postma, R. Bansal, G. Storm, et al., Targeting the Stat6 pathway in
2018 (2018), 3523642.
tumor-associated macrophages reduces tumor growth and metastatic niche
[6] J.L. Mege, V. Mehraj, C. Capo, Macrophage polarization and bacterial infections,
formation in breast cancer, FASEB journal: official publication of the Federation
Curr. Opin. Infect. Dis. 24 (2011) 230–234.
of American Societies for Experimental Biology 32 (2018) 969–978.
[7] P.J. Murray, Macrophage polarization, Annu. Rev. Physiol. 79 (2017) 541–566.
[42] J. Cao, R. Dong, L. Jiang, et al., LncRNA-MM2P identified as a modulator of
[8] T.A. Wynn, K.M. Vannella, Macrophages in tissue repair, regeneration, and
macrophage M2 polarization, Cancer immunology research 7 (2019) 292–305.
fibrosis, Immunity 44 (2016) 450–462.
[43] H. Xiang, J. Zhang, C. Lin, et al., Targeting autophagy-related protein kinases for
[9] B. Chazaud, Macrophages: supportive cells for tissue repair and regeneration,
potential therapeutic purpose, Acta Pharm. Sin. B 10 (2020) 569–581.
Immunobiology 219 (2014) 172–178.
[44] C. Vlasschaert, X. Xia, J. Coulombe, et al., Evolution of the highly networked
[10] Q. Zhou, M. Xian, S. Xiang, et al., All-trans retinoic acid prevents osteosarcoma
deubiquitinating enzymes USP4, USP15, and USP11, BMC Evol. Biol. 15 (2015)
metastasis by inhibiting M2 polarization of tumor-associated macrophages,
230.
Cancer immunology research 5 (2017) 547–559.
[45] X. Zang, X. Zhang, X. Zhao, et al., Targeted delivery of miRNA 155 to tumor
[11] R. Dong, Y. Gong, W. Meng, et al., The involvement of M2 macrophage
associated macrophages for tumor immunotherapy, Mol. Pharm. 16 (2019)
polarization inhibition in fenretinide-mediated chemopreventive effects on colon
1714–1722.
cancer, Cancer Lett. 388 (2017) 43–53.
[46] X.Y. Ren, Y.D. Han, Q. Lin, Long non-coding RNA MIR155HG knockdown
[12] A. Mantovani, S. Sozzani, M. Locati, et al., Macrophage polarization: tumor-
suppresses cell proliferation, migration and invasion in NSCLC by upregulating
associated macrophages as a paradigm for polarized M2 mononuclear
TP53INP1 directly targeted by miR-155-3p and miR-155-5p, Eur. Rev. Med.
phagocytes, Trends Immunol. 23 (2002) 549–555.
Pharmacol. Sci. 24 (2020) 4822–4835.
[13] A. Mantovani, F. Marchesi, A. Malesci, et al., Tumour-associated macrophages as
[47] N. Li, Y. Liu, J. Cai, LncRNA MIR155HG regulates M1/M2 macrophage
treatment targets in oncology, Nature Reviews, Clin. Oncol. 14 (2017) 399–416.
polarization in chronic obstructive pulmonary disease, Biomedicine &
[14] T. Lawrence, G. Natoli, Transcriptional regulation of macrophage polarization:
pharmacotherapy = Biomedecine & pharmacotherapie 117 (2019), 109015.
enabling diversity with identity, Nat. Rev. Immunol. 11 (2011) 750–761.
[48] F. Li, C. Huang, Q. Li, et al., Construction and comprehensive analysis for
[15] F. Brombacher, B. Arendse, R. Peterson, et al., Analyzing classical and alternative
dysregulated long non-coding RNA (lncRNA)-associated competing endogenous
macrophage activation in macrophage/neutrophil-specific IL-4 receptor-alpha-
RNA (ceRNA) network in gastric cancer, Medical science monitor: international
deficient mice, Methods Mol. Biol. 531 (2009) 225–252.
medical journal of experimental and clinical research 24 (2018) 37–49.
[16] K. Takeda, T. Tanaka, W. Shi, et al., Essential role of Stat6 in IL-4 signalling,
[49] Z. Liu, X. Wang, G. Yang, et al., Construction of lncRNA-associated ceRNA
Nature 380 (1996) 627–630.
networks to identify prognostic lncRNA biomarkers for glioblastoma, J. Cell.
[17] R.R. Ricardo-Gonzalez, A. Red Eagle, J.I. Odegaard, et al., IL-4/STAT6 immune
Biochem. 121 (2020) 3502–3515.
axis regulates peripheral nutrient metabolism and insulin sensitivity, Proc. Natl.
[50] X. Gao, J. Ge, W. Li, et al., LncRNA KCNQ1OT1 ameliorates particle-induced
Acad. Sci. U. S. A. 107 (2010) 22617–22622.
osteolysis through inducing macrophage polarization by inhibiting miR-21a-5p,
[18] E. Vergadi, E. Ieronymaki, K. Lyroni, et al., Akt signaling pathway in macrophage
Biol. Chem. 399 (2018) 375–386.
activation and M1/M2 polarization, J. Immunol. 198 (2017) 1006–1014.

8
R. Dong et al. Life Sciences 266 (2021) 118895

[51] Y.X. Zhou, W. Zhao, L.W. Mao, et al., Long non-coding RNA NIFK-AS1 inhibits M2 [82] K.E. Finberg, R.L. Whittlesey, N.C. Andrews, Tmprss6 is a genetic modifier of the
polarization of macrophages in endometrial cancer through targeting miR-146a, Hfe-hemochromatosis phenotype in mice, Blood 117 (2011) 4590–4599.
Int. J. Biochem. Cell Biol. 104 (2018) 25–33. [83] C.F. Bennett, Therapeutic antisense oligonucleotides are coming of age, Annual
[52] J. Liu, D. Ding, Z. Jiang, et al., Long non-coding RNA CCAT1/miR-148a/PKCzeta Review of Medicine, Vol 70 (70) (2019) 307–321.
prevents cell migration of prostate cancer by altering macrophage polarization, [84] H.J. Wu, W.F. Lima, H. Zhang, et al., Determination of the role of the human
Prostate 79 (2019) 105–112. RNase H1 in the pharmacology of DNA-like antisense drugs, J. Biol. Chem. 279
[53] H. Chen, Y. He, Y.S. Hou, et al., Long non-coding RNA CCAT1 promotes the (2004) 17181–17189.
migration and invasion of prostate cancer PC-3 cells, Eur. Rev. Med. Pharmacol. [85] S.T. Crooke, S.Y. Wang, T.A. Vickers, et al., Cellular uptake and trafficking of
Sci. 22 (2018) 2991–2996. antisense oligonucleotides, Nat. Biotechnol. 35 (2017) 230–237.
[54] D. Yang, K. Liu, L. Fan, et al., LncRNA RP11-361F15.2 promotes osteosarcoma [86] W.S. Wold, K. Toth, Adenovirus vectors for gene therapy, vaccination and cancer
tumorigenesis by inhibiting M2-like polarization of tumor-associated gene therapy, Current gene therapy 13 (2013) 421–433.
macrophages of CPEB4, Cancer Lett. 473 (2020) 33–49. [87] S.R. Choudhury, E. Hudry, C.A. Maguire, et al., Viral vectors for therapy of
[55] Z. Li, C. Feng, J. Guo, et al., GNAS-AS1/miR-4319/NECAB3 axis promotes neurologic diseases, Neuropharmacology 120 (2017) 63–80.
migration and invasion of non-small cell lung cancer cells by altering macrophage [88] Y. Wang, F. Yuan, Delivery of viral vectors to tumor cells: extracellular transport,
polarization, Functional & integrative genomics 20 (2020) 17–28. systemic distribution, and strategies for improvement, Ann. Biomed. Eng. 34
[56] L. Zhou, Y. Tian, F. Guo, et al., LincRNA-p21 knockdown reversed tumor- (2006) 114–127.
associated macrophages function by promoting MDM2 to antagonize* p53 [89] A. Khvorova, J.K. Watts, The chemical evolution of oligonucleotide therapies of
activation and alleviate breast cancer development, Cancer immunology, clinical utility, Nat. Biotechnol. 35 (2017) 238–248.
immunotherapy : CII 69 (2020) 835–846. [90] W. Shen, C.L. De Hoyos, M.T. Migawa, et al., Chemical modification of PS-ASO
[57] H.L. Luo, J. Chen, T. Luo, et al., Downregulation of macrophage-derived T-UCR therapeutics reduces cellular protein-binding and improves the therapeutic index,
uc.306 associates with poor prognosis in hepatocellular carcinoma, Cellular Nat. Biotechnol. 37 (2019) 640–650.
physiology and biochemistry: international journal of experimental cellular [91] N. Gong, X. Teng, J. Li, et al., Antisense oligonucleotide-conjugated
physiology, biochemistry, and pharmacology 42 (2017) 1526–1539. nanostructure-targeting lncRNA MALAT1 inhibits cancer metastasis, ACS Appl.
[58] Li Li, Xiaozong Lin, Peng Xu, et al., LncRNA GAS5 sponges miR-362-5p to Mater. Interfaces 11 (2019) 37–42.
promote sensitivity of thyroid cancer cells to 131 I by upregulating SMG1, IUBMB [92] L. Xiao-Jie, X. Hui-Ying, K. Zun-Ping, et al., CRISPR-Cas9: a new and promising
Life (2020) 2420–2431, https://doi.org/10.1002/iub.2365. player in gene therapy, J. Med. Genet. 52 (2015) 289–296.
[59] D. Sun, Z. Yu, X. Fang, et al., LncRNA GAS5 inhibits microglial M2 polarization [93] F. Jiang, J.A. Doudna, CRISPR-Cas9 structures and mechanisms, Annu. Rev.
and exacerbates demyelination, EMBO Rep. 18 (2017) 1801–1816. Biophys. 46 (2017) 505–529.
[60] J. Hu, L. Zhang, C. Liechty, et al., Long noncoding RNA GAS5 regulates [94] J.G. Doench, E. Hartenian, D.B. Graham, et al., Rational design of highly active
macrophage polarization and diabetic wound healing, The Journal of sgRNAs for CRISPR-Cas9-mediated gene inactivation, Nat. Biotechnol. 32 (2014)
investigative dermatology 140 (8) (2020) 1629–1638. 1262–1267.
[61] X. Chi, B. Ding, L. Zhang, et al., lncRNA GAS5 promotes M1 macrophage [95] S.S. Wu, Q.C. Li, C.Q. Yin, et al., Advances in CRISPR/Cas-based gene therapy in
polarization via miR-455-5p/SOCS3 pathway in childhood pneumonia, J. Cell. human genetic diseases, Theranostics 10 (2020) 4374–4382.
Physiol. 234 (2019) 13242–13251. [96] A. Goyal, K. Myacheva, M. Gross, et al., Challenges of CRISPR/Cas9 applications
[62] J. Cheng, J. Meng, L. Zhu, et al., Exosomal noncoding RNAs in glioma: biological for long non-coding RNA genes, Nucleic Acids Res. 45 (2017) e12.
functions and potential clinical applications, Mol. Cancer 19 (2020) 66. [97] S.J. Liu, M. Malatesta, B.V. Lien, et al., CRISPRi-based radiation modifier screen
[63] R. Kalluri, V.S. LeBleu, The biology, function, and biomedical applications of identifies long non-coding RNA therapeutic targets in glioma, Genome Biol. 21
exosomes, Science (2020) 367. (2020) 83.
[64] N. Li, L.Y. Zhang, Y.H. Qiao, et al., Long noncoding RNA LINC00662 functions as [98] S.Y. Li, Q.X. Cheng, J.K. Liu, et al., CRISPR-Cas12a has both cis- and trans-
miRNA sponge to promote the prostate cancer tumorigenesis through targeting cleavage activities on single-stranded DNA, Cell Res. 28 (2018) 491–493.
miR-34a, Eur. Rev. Med. Pharmacol. Sci. 23 (2019) 3688–3698. [99] K. Petri, V. Pattanayak, SHERLOCK and DETECTR open a new frontier in
[65] X. Tian, Y. Wu, Y. Yang, et al., Long noncoding RNA LINC00662 promotes M2 molecular diagnostics, The CRISPR journal 1 (2018) 209–211.
macrophage polarization and hepatocellular carcinoma progression via activating [100] P. Lin, S. Qin, Q. Pu, et al., CRISPR-Cas13 inhibitors block RNA editing in bacteria
Wnt/beta-catenin signaling, Mol. Oncol. 14 (2020) 462–483. and mammalian cells, Mol. Cell 78 (5) (2020) 850–861.
[66] C. Huang, J. Han, Y. Wu, et al., Exosomal MALAT1 derived from oxidized low- [101] J.S. Gootenberg, O.O. Abudayyeh, J.W. Lee, et al., Nucleic acid detection with
density lipoprotein-treated endothelial cells promotes M2 macrophage CRISPR-Cas13a/C2c2, Science 356 (2017) 438–442.
polarization, Mol. Med. Rep. 18 (2018) 509–515. [102] L.B. Harrington, D. Burstein, J.S. Chen, et al., Programmed DNA destruction by
[67] Z. Li, C. Xu, B. Ding, et al., Long non-coding RNA MALAT1 promotes proliferation miniature CRISPR-Cas14 enzymes, Science 362 (2018) 839–842.
and suppresses apoptosis of glioma cells through derepressing Rap1B by sponging [103] Y.K. Lau, M. Ramaiyer, D.E. Johnson, et al., Targeting STAT3 in cancer with
miR-101, J. Neuro-Oncol. 134 (2017) 19–28. nucleotide therapeutics, Cancers (2019) 11.
[68] Y. Zhang, J. Feng, H. Fu, et al., Coagulation factor X regulated by CASC2c [104] I.W. Choi, D.K. Kim, H.M. Ko, et al., Administration of antisense phosphorothioate
recruited macrophages and induced M2 polarization in glioblastoma multiforme, oligonucleotide to the p65 subunit of NF-kappaB inhibits established asthmatic
Front. Immunol. 9 (2018) 1557. reaction in mice, Int. Immunopharmacol. 4 (2004) 1817–1828.
[69] Y. Ye, Y. Xu, Y. Lai, et al., Long non-coding RNA cox-2 prevents immune evasion [105] L.J.A. Toonen, J. Casaca-Carreira, M. Pellise-Tintore, et al.,
and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage Intracerebroventricular administration of a 2′ -o-methyl phosphorothioate
polarization, J. Cell. Biochem. 119 (2018) 2951–2963. antisense oligonucleotide results in activation of the innate immune system in
[70] X. Li, Y. Lei, M. Wu, et al., Regulation of macrophage activation and polarization mouse brain, Nucleic acid therapeutics 28 (2018) 63–73.
by HCC-derived exosomal lncRNA TUC339, Int. J. Mol. Sci. (2018) 19. [106] Y. Uemura, K. Kobayashi, Targeting murine alveolar macrophages by the
[71] Z.X. Liang, H.S. Liu, F.W. Wang, et al., LncRNA RPPH1 promotes colorectal intratracheal administration of locked nucleic acid containing antisense
cancer metastasis by interacting with TUBB3 and by promoting exosomes- oligonucleotides, Drug delivery 26 (2019) 803–811.
mediated macrophage M2 polarization, Cell Death Dis. 10 (2019), 829. [107] F. Imperatore, J. Maurizio, S. Vargas Aguilar, et al., SIRT1 regulates macrophage
[72] R.A. Boon, N. Jae, L. Holdt, et al., Long noncoding RNAs: from clinical genetics to self-renewal, EMBO J. 36 (2017) 2353–2372.
therapeutic targets? J. Am. Coll. Cardiol. 67 (2016) 1214–1226. [108] R. Elling, E.K. Robinson, B. Shapleigh, et al., Genetic models reveal cis and trans
[73] M.C. Jiang, J.J. Ni, W.Y. Cui, et al., Emerging roles of lncRNA in cancer and immune-regulatory activities for lincRNA-Cox2, Cell reports, 25 e6 (2018)
therapeutic opportunities, Am. J. Cancer Res. 9 (2019) 1354–1366. 1511–1524.
[74] C. Wahlestedt, Targeting long non-coding RNA to therapeutically upregulate gene [109] L.M. Holdt, A. Stahringer, K. Sass, et al., Circular non-coding RNA ANRIL
expression, Nat. Rev. Drug Discov. 12 (2013) 433–446. modulates ribosomal RNA maturation and atherosclerosis in humans, Nat.
[75] C.K. Huang, S. Kafert-Kasting, T. Thum, Preclinical and clinical development of Commun. 7 (2016), 12429.
noncoding RNA therapeutics for cardiovascular disease, Circ. Res. 126 (2020) [110] X. Wang, F.Y. Li, W. Zhao, et al., Long non-coding RNA GAS5 overexpression
663–678. inhibits M2-like polarization of tumour-associated macrophages in SMCC-7721
[76] R.R. Nikam, K.R. Gore, Journey of siRNA: clinical developments and targeted cells by promoting PTEN expression, Int. J. Exp. Pathol. 101 (6) (2020) 215–222.
delivery, Nucleic acid therapeutics 28 (2018) 209–224. [111] Y.K. Kim, Y.S. Kim, S. Kim, et al., Comprehensive evaluation of differentially
[77] R. Palchaudhuri, P.J. Hergenrother, Transcript profiling and RNA interference as expressed non-coding RNAs identified during macrophage activation, Mol.
tools to identify small molecule mechanisms and therapeutic potential, ACS Immunol. 128 (2020) 98–105.
Chem. Biol. 6 (2011) 21–33. [112] C. Yunna, H. Mengru, W. Lei, et al., Macrophage M1/M2 polarization, Eur. J.
[78] J. Chery, RNA therapeutics: RNAi and antisense mechanisms and clinical Pharmacol. 877 (2020), 173090.
applications, Postdoc journal: a journal of postdoctoral research and postdoctoral [113] K. Xue, R.E. MacLaren, Antisense oligonucleotide therapeutics in clinical trials for
affairs 4 (2016) 35–50. the treatment of inherited retinal diseases, Expert Opin. Investig. Drugs 29 (2020)
[79] A.M. Quemener, L. Bachelot, A. Forestier, et al., The powerful world of antisense 1163–1170.
oligonucleotides: from bench to bedside, Wiley interdisciplinary reviews, RNA [114] R.I. Hara, K. Yoshioka, T. Yokota, DNA-RNA heteroduplex oligonucleotide for
(2020) e1594. highly efficient gene silencing, Methods Mol. Biol. 2176 (2020) 113–119.
[80] T. Lucas, A. Bonauer, S. Dimmeler, RNA therapeutics in cardiovascular disease, [115] K. Nishina, W. Piao, K. Yoshida-Tanaka, et al., DNA/RNA heteroduplex
Circ. Res. 123 (2018) 205–220. oligonucleotide for highly efficient gene silencing, Nat. Commun. 6 (2015), 7969.
[81] S. Altamura, U. Schaeper, S. Dames, et al., SLN124, a GalNAc-siRNA conjugate
targeting TMPRSS6, efficiently prevents Iron overload in hereditary
haemochromatosis type 1, HemaSphere 3 (2019), e301.

9
R. Dong et al. Life Sciences 266 (2021) 118895

[116] K. Dhuri, C. Bechtold, E. Quijano, et al., Antisense oligonucleotides: an emerging disease: a multicentre, double-blind, randomised, placebo-controlled phase 2
area in drug discovery and development, J. Clin. Med. 9 (2020). trial, The lancet. Gastroenterology & hepatology 5 (2020) 829–838.
[117] R. Loomba, E. Morgan, L. Watts, et al., Novel antisense inhibition of [118] C.M. Miller, M. Tanowitz, A.J. Donner, et al., Receptor-mediated uptake of
diacylglycerol O-acyltransferase 2 for treatment of non-alcoholic fatty liver phosphorothioate antisense oligonucleotides in different cell types of the liver,
Nucleic acid therapeutics 28 (2018) 119–127.

10

You might also like