Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Cellular Signalling 83 (2021) 109995

Contents lists available at ScienceDirect

Cellular Signalling
journal homepage: www.elsevier.com/locate/cellsig

Micro-RNA: The darkhorse of cancer


Mridul Budakoti a, Abhay Shikhar Panwar a, Diksha Molpa a, Rahul Kunwar Singh b,
Dietrich Büsselberg d, Abhay Prakash Mishra c, *, Henrique Douglas Melo Coutinho e, *,
Manisha Nigam a, *
a
Department of Biochemistry, H. N. B. Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
b
Department of Microbiology, H. N. B. Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
c
Department of Pharmaceutical Chemistry, H. N. B. Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
d
Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
e
Regional University of Cariri, URCA, Crato, Brazil

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

Keywords: The discovery of micro RNAs (miRNA) in cancer has opened up new vistas for researchers in recent years. Micro
Micro RNAs RNAs area set of small, endogenous, highly conserved, non-coding RNAs that control the expression of about
Cancer biology 30% genes at post-transcriptional levels. Typically, microRNAs impede the translation and stability of messenger
Oncogene
RNAs (mRNA), control genes associated with cellular processes namely inflammation, cell cycle regulation, stress
Tumor suppressors
Therapy
response, differentiation, apoptosis, and migration. Compelling findings revealed that miRNA mutations or
disruption correspond to diverse human cancers and suggest that miRNAs can function as tumor suppressors or
oncogenes. Here we summarize the literature on these master regulators in clinical settings from last three de­
cades as both abrupt cancer therapeutics and as an approach to sensitize tumors to chemotherapy. This review
highlights (I) the prevailing perception of miRNA genomics, biogenesis, as well as function; (II) the significant
advancements in regulatory mechanisms in the expression of carcinogenic genes; and (III) explains, how miRNA
is utilized as a diagnostic and prognostic biomarker for the disease stage indicating survival as well as therapeutic
targets in cancer.

1. Introduction its expression through sequence-specific RNA-RNA interactions with the


3’-untranslated regions of its mRNA [3]. Currently, a total of approxi­
Lin4, the first micro RNA was detected in Caenorhabditis elegans mately 2000 human miRNA annotated precursor genes on miRBase,
[C. elegans] by Victor Ambros and colleagues in 1993, while studying the while several miRNA functions are still unknown, are described. Hence,
gene lin-14 [1]. It is a “small, non-protein-coding RNA, regulating the nowadays, miRNA-oriented gene regulation is an intriguing field of
expression of protein-coding gene” [1]. Today, lin-4 is considered as the research and analysis. The miRNA plays remarkable roles in diverse
fountain of an extended class of small regulatory RNAs, defined as processes, comprising cell proliferation and death, metabolism,
microRNAs (miRNAs). Seven years later, another miRNA, let-7 in C. neuronal patterning, hematopoietic differentiation and immunity [4].
elegans was reported by Reinhart et al. [2], which negatively regulates miRNAs are a family of small, evolutionary conserved, single-

Abbreviations: DGCR8, DiGeorge syndrome critical region 8; RISC, RNA-induced silencing complex; B CLL, B - cell chronic lymphocytic leukemia; ALL, Acute
lymphoblastic leukemia; MLL, Mixed-lineage leukemia; ZEB, Zinc-finger E-box-binding homeobox; dsRBD, dsRNA binding domain; Exp-5, Exportin-5; TRBP,
Transactivation response RNA- binding protein; HCV, Hepatitis C virus; NSCLC, Non-small-cell lung cancer; PDAC, Pancreatic ductal adenocarcinoma; CLL, Coated
cationic lipid; MSC-EVs, Mesenchymal stem cell-derived extracellular vesicles; OSCC, Oral squamous cell cancer; OMPD, Oral potentially malignant disorders; NPs,
NPs, nanoparticles; OXL, Oxaliplatin; HCC, Hepatocellular carcinoma; PACT, Protein- kinase, interferon-inducible double-stranded RNA-dependent activator; UTR,
Untranslated region of mRNA; ORF, Open reading frame; CEA, Carcinoembryonic antigen; CRC, Colorectal cancer; APC, Adenomatous polyposis coli; SERM, Selective
estrogen receptor modulator; ATM, Ataxia-telangiectasia mutated; PTEN, Phosphatase and tensin homology; PPB, Pleuropulmonaryblastoma; THBS1, Thrombo­
spondin 1; CTGF, Connective tissue growth factor; EMT, Epithelial-mesenchymal transition; METTL3, Methyltransferase- like 3.
* Corresponding authors.
E-mail addresses: dib2015@qatar-med.cornell.edu (D. Büsselberg), abhaypharmachemhnbgu@gmail.com (A.P. Mishra), hdmcoutinho@gmail.com
(H.D.M. Coutinho), m.nigam@hnbgu.ac.in (M. Nigam).

https://doi.org/10.1016/j.cellsig.2021.109995
Received 24 January 2021; Received in revised form 25 March 2021; Accepted 25 March 2021
Available online 27 March 2021
0898-6568/© 2021 Elsevier Inc. All rights reserved.
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

stranded, non-coding RNA molecules that prevents the translation of 3.2. microRNA microarray
target mRNA using one of two prominent mechanisms [5]. First mech­
anism involves two-step cleavage of primary miRNA [pri-miRNA] Northern blotting is used for miRNA analysis, but it has certain
generating mature miRNA, which ultimately integrates into the limitations, such as unequal hybridization efficiency of individual
effector complex known as RNA-induced silencing complex (RISC) [5]. probes [8] and difficulties in simultaneously detecting multiple miRNAs.
In second mechanism, the miRNA down-regulates the expression of However, cancer studies require to compare the expression pattern of all
target mRNA by base-pairing with it. The silencing mechanism utilized known miRNAs between cancer cells and healthy cells. Therefore, it is
either cleavage of target messenger RNA [mRNA] with consequent better to have methods that identify all miRNA expression at once. This
degradation or translational inhibition, determined by the extent of limitation was overcome by two-color fluorescence dependent micro­
complementarity amid guide miRNA and its target mRNA [5]. Although array (DNA microarray) technology that commonly used to identify
there is a general understanding of miRNA function, mechanistic detail gene expression simultaneously. With the advancement, DNA micro­
of miRNA biogenesis and gene silencing is still unclear and under array technology was revised to shape miRNA microarray technology
scrutiny [5]. The altered expression profiles of miRNAs in distinct tu­ [9]. A custom dual-channel miRNA microarray was developed and
mors, imply that these molecules may be involved in cancer develop­ employed to monitor expression levels of 124 mammalian miRNA [8].
ment and other malignancies [5]. miRNAs’ expression patterns differ among adult mouse tissue and em­
This review intends to highlight micro RNA biogenesis, its regula­ bryonic stem cells [8]. Lu et al. [10] developed a novel strategy to detect
tion, integral roles and how different elements of microRNA synthetic abundant miRNA expression profiles in different cell types, including
machinery gets disrupted in multiple types of cancer with all the latest several human cancers. To address the questions about the specificity of
findings involved. The topics highlighted further includes the method­ the probes in the miRNA microarray analysis, they first conducted so­
ology employed for studying the role of microRNA in cancer and the lution hybridization and then quantified the polymer heads hybridized
mechanisms by which it gets deregulated. Moreover, how miRNAs are to miRNA species utilizing multicolor flow sorting [10]. This approach is
involved in attributes that are the hallmarks of cancer, either as onco­ used to identify miRNA expression profiles in cancer, even in tumors
genes or tumor suppressors, will also be conferred. Finally, an inspection that are poorly differentiated [10]. miRNA microarray analysis has
has been made that whether miRNA could be utilized as biomarkers for become a robust tool for understanding the relationship between cancer
malignant growth determination, prognosis, and treatment. The article tissues and healthy tissues. Furthermore, miRNA microarray technology
evaluated the literature of last three decades available on the clinical use is commonly used to analyze miRNA’s involvement in cancers [9].
of miRNAs as cancer therapeutics and as an approach to sensitize tumors
to concurrent chemotherapy. 3.3. Quantitative real-time PCR (qRT-PCR)

2. Methodology Quantitative Real-time PCR technology are quite efficient and widely
used in amplifying and quantifying both the precursor and mature
A comprehensive search of the various scientific databases from last microRNA [11]. The most commonly used methods are Taqman probe
three decades was utilized to explore the studies evaluating microRNAs and SYBR Green qRT-PCR. Comparison of these two methods has shown
and their role in cancer. The search strategy included relevant key terms that there is no significant differences in observed miRNA expression
for instance; microRNAs, biogenesis, cancer, oncogenes and tumor profile [12]. Both the methods require specific primers for analysis of
suppressors, biomarkers and therapeutics. The reference lists all eligible mature, pri- and pre- miRNAs. He et al [13] have reported the differ­
research articles and reviews to discern pertinent studies. ential expression of four miRNAs namely, hsa-miR-29a-5p, hsa-miR-
135a-5p, hsa-miR-542-3p, and hsa-miR-4491 in patients with lung
3. Techniques to investigate miRNAs’ function in cancer cancer as compared to control using RT-PCR. Although qRT-PCR is used
in quantifying the expression profile of miRNA, however short length of
The expression of miRNAs has been used as non-invasive marker of mature miRNA and high similarity between miRNA family mambers
cancer for last few decades. There are several techniques to detect the create challenges in expression analysis. Advantage of reverse tran­
expression of miRNAs ranging from nucleic acid amplification to scription step has been taken by many available methods in multiple
sequencing, which are briefly described: ways, so that additional sequences can be incorporated that facilitate the
amplification by PCR and its identification. To avoid challenges in
expression analysis of miRNA, more advance methods has been devel­
3.1. Northern blot analysis oped with imporved sensitivity and specificity which includes stem-loop
RT-PCR and poly(A)-tailed RT-PCR. It is imperative to note that much
Northern blot analysis is one of the strategies for detecting gene more attention has been dedicated to mature miRNA expression
expression at the mRNA level including miRNAs. Due to its practicality profiling, although pri-miRNA and pre-miRNA are important in­
and reliability, the technique is considered as gold standard for RNA termediates during miRNA biogenesis and intrestingly, pre-miRNAs
detection. It was introduced early on to analyze the function of miRNA have also been recognized as valuable disease biomarkers [14].
genes [1] and also been used as an approach to identify miRNA
expression in cancer cells. Hayashita et al. [6] implemented Northern 3.4. NanoString technology
blot in their investigation and found that the miR-17–92 cluster is
significantly over-expressed in lung cancer compared with miRNA The NanoStringn technology is a new and promising technology
expression in healthy cells, particularly with small cell lung cancer [6]. which ganing prevelence in measuring nucleic acid abundances in
With the advancement in molecular biology techniques, high- various clinical studies.This technology is more advance than PCR based
throughput alternatives of Northern blotting have been introduce as it method and sufficent to avoid the constrains associated with PCR. The
has certain drawbacks in comparison to other analytical approaches major advantages of NanoString technology is that amplification step is
being less sensitive, time-consuming and labor-intensive with reduced not required and there is direct sequence examination and digital
efficiency. As the method requires very high starting amount of RNA detection for absolute quantification [15]. This considerably minimize
pool therefore it is not suitable for regular clinical investigations and the chance of contamination or experiment error and efficiently deal
high-performance miRNA research. The susceptibility of the process for with the formalin-fixed paraffin-embedded samples. The key feature of
RNase degradation and disproportionate hybridization efficiency of NanoString technology is that it make use of novel digital color-coded
probes has made this method less fascinating among scientist. [7] barcode technology based on direct multiplexed measurement of gene

2
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

expression [16].Which is responsible for enhanced sensitivity and pre­ 4. Micro RNA biogenesis in animals
cision of this tool. Basically there is a binding of each color-code barcode
to a single target-specifie probe that correspond to a single gene The biogenesis of microRNA begins in the nucleus with the tran­
consequently count individually without amplification. The molecular scription of the miRNA gene to primary miRNA (pri-miRNA) which is
counting through NanoString able to measure mRNA transcripts directly then processed by a protein complex called microprocessor complex to
and in a single reaction ~800 colored probes can be run simultaneously ~85 nucleotide stem-loop structure called precursor miRNA (pre-
[17]. Recently NanoString technology has been employed by cancer miRNA) [3]. The microprocessor complex consists of a protein namely
researchers to study the miRNA expression profiling in cancer such as DGCR8 (DiGeorge syndrome critical region gene 8) [4,5] which is
lung cancer, prostate cancer etc [18,19]. capable of binding double-stranded RNA and a class 2 RNase III endo­
nuclease, Drosha. The pre-mRNA is capped at 5’ and polyadenylated at
3’ [3]. Then this pre-miRNA is transferred from the nucleus to cytoplasm
3.5. miRNA sequencing through Ran/GTP/ exportin-5 (EXP-5). Further processing to form ~20-
22 nucleotide mature miRNA [miRNA/miRNA*] duplex (*denotes pas­
With the rapid progression of next-generation sequencing (NGS) senger strand and the other complementary strand is the guide or
techniques, trend of miRNA profiling has been boosted up with miRNA mature stand) occurs in the cytoplasm through Dicer a RNase III type
sequencing. Sequencing of complete set of miRNAs present in an RNA protein [3]. Mature miRNA is ultimately loaded upon the Argonaute
sample is possible through NGS technology. Being high-throughput and (Ago) protein to generate the effector RNA- induced silencing complex
high-resolution tools it is able to sequence small amount of RNA samples (RISC). Subsequently, the miRNA duplex is unwound and guides RISC to
with accuracy.NGS is preferred method for miRNA profiling because of target mRNA. Fig. 1 illustrates the processes involved [5].
its high sensistivity, single-nucleotide resolution and ability to parallelly In 2000 a new domain of miRNA genomics emerged with the
profile the significant number of samples [20]. It has high multiplexing disclosure of the let-7 RNA in Caenorhabditis elegans [25], followed by its
potential, and display excellent reproducibility.The major steps detection in organisms including humans [26]. Ever since a multitude of
involved in miRNA-seq data analysis are as following- mapping the miRNAs have been reported in humans and other multicellular organ­
reads to miRBase, followed by considering mismatches during the isms using various techniques. miRNAs are phylogenetically conserved
hairpin alignment (windowing),and finally counting the reads (quanti­ in bilateral animals [4]. Almost half of all known miRNAs are found
fication) [21]. The data generated by this NGS experiments is helpful to close to other miRNAs and are uttered as poly-cistronic primary tran­
figure out the expression and function of miRNA, sequence isoforms scripts. Based on their genomic location, miRNA genes are categorized
identification, novel miRNAs prediction, and prediction of potential as exonic miRNAs in coding transcription units, exonic miRNAs in non-
mRNA target molecules [22]. NGS for miRNA expression profiling has coding transcription units, intronic miRNAs in coding transcription units
become the prefered tool and extensively used in a range of biological and intronic miRNAs in non-coding transcription units [4]. The exact
studies including cancer research [23,24]. mechanism of miRNA expression regulation is still unknown. However,
The strengths and weaknesses of these approaches for studying the primary miRNA transcripts produced by Pol II are presumed to be
functions of miRNAs in cancer pathogenesis are summarized in Table 1.

Table 1
miRNA screening techniques.
Technique Agent Definition Mechanism of action Strengths Weaknesses References

Northern blot Northern blotting is a The most reliable No direct correlation between [1,6,7]
procedure extensively used to techniques employed to the levels of mRNA expression
analyze the molecular size and study miRNA expression in and if the up- or down-
abundance of mRNA. cancers. regulation of specific miRNAs
is the cause of cancer or a
downstream consequence of
the disease.
miRNA Microarray technology is a powerful The expression of multiple No direct connection between [8–10]
microarray high-throughput tool capable of miRNAs in cancers can be the levels of mRNA expression
monitoring the expression of detected, might become a and if the up- or down-
thousands of small non-coding technique in cancer regulation of certain miRNAs is
RNAs in tens of parallel samples in a epidemiology and early the cause of cancer or a
single experiment at once cancer detection. downstream consequence of
the disease.
Quantitative To measure the time course of miRNA expression No direct correlation between [11–14]
real-time precursor and mature miR-155 detected rapidly, especially the levels of mRNA expression
PCR (qRT- expression in monocytes stimulated pri-miRNA expression. and whether the up- or down-
PCR) by lipopolysaccharide, real-time regulation of certain miRNAs is
PCR assays were used. the cause of cancer or a
downstream effect of the
disease
NanoString It employs a novel digital Hundreds of unique To account for the sample [15–19]
Technology color-coded barcode transcripts are identified variations, the data from the
technology that provides a and counted in a single NanoString nCounter requires
high degree of accuracy and reaction using "molecular additional processing,
sensitivity based on direct barcodes" and single normalization and analysis.
multiplexed measurement of molecule imaging.
gene expression
miRNA A high-throughput deep- High precision, in Low quantities of specimens [20–24]
Sequencing sequencing technology widely distinguishing miRNAs such as FFPE specimens pose a
used in studying which are very similar in major concern to RNA-Seq
transcriptome. sequence as well as efficiency.
isomiRs.

nt – nucleotide; AMO – anti-microRNA antisense oligodeoxyribonucleotide; LNA – locked nucleic acids; SMIRs: small-molecule inhibitors of miRNAs (SMIRs).

3
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Other evidence suggests miRNA regulation in response to hypoxia, di­


etary, and hormonal changes [4,5].

4.1. Transcription of Pri-miRNA in cancer

Numerous human cancers disrupt the first step of miRNA biogenesis


namely the transcription of pri-miRNA [4] as considerable number of
human miRNA genes reside at fragile sites or in genomic regions that are
deleted, amplified or translocated in cancer [30]. These genomic vari­
ations, alterations are frequently observed in pri-miRNA transcription
and miRNA expression which results in the abnormal expression of
downstream target mRNAs that can enhance cancer initiation and pro­
gression [30,31].
For instance:
• the sides of miR-15 and miR-16 (microRNA coding genes) on
chromosome 13q14 (usually regulate apoptosis by targeting BCL-2
mRNA) [32], are recurrently deleted in B-cell chronic lymphocytic
leukemia (CLL), leading to the loss or diminished expression of both of
these miRNAs in ~70% of B-CLLs [33]. It is pertinent to note that the
first evidence of involvement of microRNAs in cancer was reported by
Calin et al., who observed knockdown or knockout of miR-15a and miR-
16-1 in approximatively 69% of CLL patients [33].
• the processing of pri-miR-128b is blocked due to a point mu­
tation in the miR-128b (also called miR-128-2) gene, resulting in
reduced levels of mature miR-128b, thus resulting glucocorticoid resis­
tance in acute lymphoblastic leukemia (ALL) cells with mixed-lineage
leukemia (MLL)–AF4 (also called as KMT2A–AFF1) translocation [34].
Apart from genomic alterations, disrupted miRNA expression could
emerge from alterations in tumor suppressor or oncogenic factors that
function as transcriptional activators or repressors to regulate pri-
miRNA transcription.
For instance:
• Myc, a proto-oncoprotein, comprising the miR-17~92 cluster
induces expression of oncogenic miRNAs in cancer. Such MYC-targeted
miRNAs facilitate cancer progression by regulating the expression of
E2F1, thrombospondin 1 (THBS1), connective tissue growth factor
(CTGF) and other target mRNAs to regulate cell cycle progression and
angiogenesis [35,36].
• expression of the miR-200 family (miR-200a, miR-200b, and
miR-200c) is recurrently silenced in human tumors. mRNAs encoding
the zinc-finger E-box-binding homeobox (ZEB) transcription factors,
ZEB1 and ZEB2 are direct targets of these miRNAs, which suppress the
expression of epithelial genes to promote the epithelial-mesenchymal
Fig. 1. Canonical miRNA biogenesis and processing. A primary microRNA(pri-
transition (EMT). Paradoxically, ZEB1 and ZEB2 specifically bind with
miRNA) is generated after the transcription of the miRNA gene. Then, the
miR-200 promoter to a regulatory factor to suppress miR-200 tran­
primary microRNA undergoes nuclear cleavage by a microprocessor complex,
composed of DGCR8 which is an RNA binding protein and Drosha, a type 3 scription as part of a negative regulatory feedback loop that facilitates
RNase to generate ~85- nucleotide stem-loop structure called precursor EMT [37].
microRNA (pre-miRNA), which is transported from nucleus to cytoplasm by Other cancer-related transcription factors often aberrantly control
Ran/GTP/Exportin 5 complex, where it is further cleaved generating ~20-22 the transcription of miRNAs in cancer. Transcriptional dysregulation,
nucleotide microRNA duplex (miRNA: miRNA*) (asterisk designates passenger either by genetic degradation of miRNA genes or through aberrant
strand whereas the other strand is mature miRNA). The mature miRNA is transcription factor activity, is a significant cause for altered miRNA
assembled into a protein complex called RISC after the duplex unwinds. The expression in cancer.
recently discovered Methyltransferase-like 3 (METTL3) has a role in methyl­
ation of pri-miRNAs, which marks them to be recognized and processed by
4.2. Nuclear processing
DGCR8 to yield mature miRNA. The mature miRNA directs gene silencing of
target mRNA either by translation repression or gene silencing, which is
determined by the degree of complementarity between the miRNA and the
Within the nucleus, pri-miRNA is transformed to pre-miRNA through
mRNA target. Drosha, an “RNase III enzyme [4,38–44] that also contains a dsRNA-
binding protein, DGCR8 (DiGeorge syndrome chromosomal region 8;
also known as Pasha in Caenorhabditis elegans and Drosophila). Usually,
regulated like protein-coding transcripts [5]. Proteins like HnRNPA1,
pri-miRNAs consist of a stem-loop of nearly 33 base segments – pairs
SMAD1, and SMAD5 interact with miRNA precursors and control their
(bp) and a terminal loop along with single-stranded RNA (ss RNA)
subsequent processing into mature miRNA [4,5,27]. Besides, mature
flanking segments. DGCR8 corresponds with ss RNA and directs Drosha
miRNA is impaired by certain regulatory proteins and thus restricting its
to process pri-miRNA [4]. “RNA duplexes are cleaved about 11bp
expression. An example of such a regulatory protein is Lin-28, which
beyond the ss RNA stem-loop junction by Drosha and consequently
binds to let-7 miRNA and targets its degradation [5,28]. It is predicted
processes pri-miRNA to pre-miRNA with a 5’- phosphate and about 2
that DNA methylation controls about 10% of miRNA expression [29].
nucleotide 3’ overhang” [4,40,45,46]. The pre-miRNA has one end of

4
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

the mature miRNA [4] identified by Drosha’s cut and includes mature spliced proteins are disabled to cooperate with DGCR8 and thus defec­
miRNA in either the 3’ or 5’ arm [4,47]. tive in the in-vitro processing of pri-miRNA.
• There is dysregulation of the expression DGCR8 in cancer.
Furthermore, mutations of DGCR8 were claimed in Wilms tumors: a
4.3. Defective cancer microprocessor frequent mutation (E518K) in dsRBD1 consequences in the diminished
expression of critical miRNAs in the tumors. As with Drosha’s knock­
The function and expression of the microprocessor components are down, DGCR8’s knockdown also facilitates cell transformation and
frequently disrupted in cancer (Table 2). tumor growth, further reinforcing the microprocessor’s imperative role
For instance: in cancer. Besides, in many other cancers downregulation of Drosha has
• The Drosha expression levels are upregulated in various cancer been reported [56–59].
types. In over 50 percent of advanced cervical squamous cell carci­
nomas, Drosha is overexpressed [48]. Its enhanced expression acceler­
ates cell proliferation, migration, and invasion eventually leading to 4.4. Exportin 5 (XPO5)
cancer advancement [49–51].
• Alternatively, expression level of Drosha is down-regulated in The subsequent step in miRNA biogenesis is the transportation of
many other types of cancer resulted in reduced miRNA expression [52] pre-miRNA into the cytoplasm to synthesize mature miRNA for further
and is linked with metastasis, invasion [53] and low survival rate in processing. The pre-miRNA shipment occurs via nuclear pore com­
subjects [54]. plexes, which are massive protein channels incorporated in the nuclear
• In lung adenocarcinoma cells knockdown of Drosha leads to membrane [4]. Free Exp5 undergoes extensive opening motion, and
augmented proliferation and tumor growth in vitro and in vivo, indi­ therefore facilitates the RanGTP binding [66]. Pre-miRNA assembles the
cating its involvement as a tumor suppressor to impede cancer nucleocytoplasmic transport factor Exportin-5 and Ran-GTP into a
progression. complex that prevents nuclear degradation and promotes cytoplasm
• In melanoma and teratocarcinoma cells, Drosha mRNA was translocation [4].One proposed miRNA transport model indicates that
alternatively spliced [55]. Consequently, the splice variants encode pre-miRNA export is triggered when the XPO5 identifies the >14-bp
carboxy-terminal-truncated Drosha proteins that partially lack the double-stranded RNA (ds-RNA) stem-loop with a 3’overhang accompa­
RNase III b domain and the dsRNA binding domain (dsRBD). Such nied by collective binding to both pre-miRNA along with GTP-bound

Table 2
Summary of dysregulated components of miRNA biogenesis machinery in cancers.
Protein Type of dysregulation Type of cancer Clinical alteration References

DROSHA Upregulation Cervical SCC Altered miRNA profile; associated with neoplastic progression [48,49]
Esophageal cancer Regulates cell proliferation; associated with poor patient survival [50]
Smooth muscle tumors Associated with tumor progression [51]
Gastric cancer Associated with pathological characteristics and patient survival [56]
Serous ovarian carcinoma Associated with advanced tumor stages [57]
Down-regulation Bladder cancer Altered miRNA profile [58]
Ovarian cancer Associated with poor patient survival [51]
Endometrial cancer Correlated with histological grade [51]
Nasopharyngeal carcinoma Correlated with shorter patient survival [58]
Breast cancer Not determined [59]
Gallbladder adenocarcinoma Correlated with metastasis, invasion and poor prognosis [50]
Neuroblastoma Correlated with global downregulation of miRNAs and poor outcome [52]
Cutaneous melanoma Associated with cancer progression and poor survival [54, ]
DGCR8 Upregulation Esophageal cancer Associated with poor patient survival [50]
Bladder cancer Altered miRNA profile [58]
SCC and BCC Not determined [60]
Prostate cancer Associated with dysregulated miRNA [61]
Colorectal carcinoma Not associated with any clinical parameters [62]
Ovarian cancer Required for cell proliferation, migration, and invasion [63]
DICER1 Upregulation Smooth muscle tumors Associated with high-grade disease and tumor progression [51]
Gastric cancer Correlated with gastric tumor subtype [56]
Serous ovarian carcinoma Associated with advanced tumor stages [57]
Prostate cancer Dysregulated miRNA expression; correlated with tumor stage [54]
Oral cancer Required for proliferation [54]
Colorectal cancer Correlated with tumor stage and associated with poor survival [54]
Cutaneous melanoma Correlated with clinical stage [54]
Down-regulation Triple-negative breast cancer No clinical correlation [54]
Bladder cancer Altered miRNA profile [54]
BCC Not determined [54]
Ovarian cancer Associated with advanced tumor stage and poor patient survival [54]
Endometrial cancer No association with histological grade detected [64]
Nasopharyngeal carcinoma Correlated with shorter patient survival [54]
Neuroblastoma Associated with global downregulation of miRNAs and poor outcome [52]
Breast cancer Associated with cancer progression and recurrence [54]
Gallbladder adenocarcinoma Correlated with metastasis, invasion and poor prognosis [54]
Non-small cell lung cancer Low levels of DICER1 expression correlate with shortened survival [54]
Chronic lymphocytic leukemia Associated with progression and prognosis [65]
PACT Upregulation AK, SCC, and BCC Not determined [60]
XPO5 Downregulation Bladder cancer Associated with altered miRNA profile [54]
AGO1 Upregulation AK, SCC, and BCC Not determined [60]
Serous ovarian carcinoma Associated with advanced tumor stages [57]
AGO2 Upregulation AK, SCC, and BCC Not determined [60]
Serous ovarian carcinoma Correlated with advanced tumor stages and associated with shorter survival [57]

5
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

cofactor Ran within the nucleus [4].The pre-miRNA attached EXP5 suggests that the stable end of the miRNA duplex binds to interacting
transport from the nucleus, where GTP hydrolysis contributes to pre- proteins in the RLC after the processed miRNA duplexes are liberated
miRNA release [4]. Being an important transportation protein of pre- from Dicer and the unstable end links with the Ago protein [76,77].
miRNA from nucleus to cytoplasm, any alterations in XPO5 owing to The endo-nucleolytic enzyme activity of the Ago protein is respon­
genetic mutation, epigenetic change, abnormal expression level or sible for removing the miRNA passenger strand [78]. RNA Helicase A
posttranslational modification, have an effect on miRNA expression (RHA), also referred to as DHX9 OR NDH11, is responsible for RNA
consequently influence tumorigenesis [67]. It has been noticed in he­ duplex unwinding associated with RISC activation in the human miRNA
patocellular carcinoma that ERK kinase phosphorylation of exportin-5 pathway [78]. RHA is capable of unwinding RNA: RNA duplexes with 3 ’
(XPO5) at T345/S416/S497 and hinder the pre-miRNA export from overhangs, but the efficacy of RHA is uncertain and is assumed to rely on
the nucleus moreover prolyl isomerase Pin1 alter the conformation of the interaction of RHA with the associated proteins [78–80]. In a spatial
XPO5 after phosphorylation by ERK kinase and as a result there is turn arrangement optimal for unwinding RNA:RNA duplexes, RHA interacts
down the loading of pre-miRNA [67]. with Dicer, Ago2, and TRBP. Upon loading, miRNA directs the RISC to
its target mRNA, which is silenced by degradation or translation
4.5. Pre-miRNA export in cancer repression [4]. Helicase MOV10 (Moloney leukemia virus 10 homolog)
was correlated with some Ago2 RISC complexes but further studies are
In the rare colon, gastric and endometrial tumors with microsatellite required to determine if it is associated with miRNA duplex unwinding.
instability, three recurring heterozygous XPO5-inactivating mutations
were established [68]. Such XPO5 mutations hinder the export of pre- 4.7. Mutations in Dicer 1 in cancer
miRNA and induce aggregation of pre-miRNAs within the nucleus,
resulting in defects in miRNA biogenesis. Genetic and epigenetic asso­ Dicer1 depletion in cancer cells and mouse models facilitate cell
ciation researches indicated that XPO5 genetic variation and degree of growth along with tumorigenesis, suggesting Dicer1’s essential function
expression correlate with breast cancer risk [69]. XPO5 dysregulation in oncogenesis [81,82]. Dicer is believed to be a haplo-insufficient tumor
thus results in miRNA processing defects and tumorigenesis. It has been suppressant gene since the attenuation of a single Dicer1 allele decreases
noticed in hepatocellular carcinoma that ERK kinase phosphorylation of lung cancer’s subsistence in a mouse model [83].
exportin-5 (XPO5) at Thr345, Ser416, Ser497, and hinder the pre- • Mutations of heterozygous Dicer1 are accountable for pleuro­
miRNA export from the nucleus moreover prolyl isomerase Pin1 alter pulmonary blastoma (PPB), an unusual pediatric lung tumor that
the conformation of XPO5 after phosphorylation by ERK kinase and as a emerges during the advancement of the fetal lung and is frequently an
result there is turn down the loading of pre-miRNA [67,70]. Therefore element of a hereditary cancer syndrome [84].
XPO5 phosphorylation is associated with a global downregulation of • The the metal-binding residues (E1705, D1709, G1809, D1810,
miRNAs and poor prognosis in hepatocellular carcinoma patients [71]. and E1813) in RNase IIIb domain are hot spots for the mutations [85].
Such mutations cannot alter the expression of Dicer1 protein but cause
4.6. Cytoplasmic processing defects in the function of the RNase IIIb domain. Consequently, the
maturation of 5p miRNAs is specifically blocked while the processing of
miRNA maturation in cytosol spins around RNase III endonuclease 3p miRNAs (miRNAs originating from the 3’ side of the pre-miRNA
Dicer, identified in all eukaryotes studied to date except budding yeast hairpin) stays unaffected, resulting in a global downfall of 5p miRNAs
[5,72]. Dicer, is a multi-domain protein located in the cytoplasm and/or in cancer [85,86].
the rough endoplasmic reticulum. It is composed of an N-terminal • Dicer1 RNase IIIb mutations significantly diminish the expression
ATPase / helicase domain, DUF283 (a domain of unknown function), of the let-7 tumor-suppressive miRNA family (all of which are 5’
PAZ (Piwi/Argonaute/Zwilli) domain and two tandem RNase3 nuclease derived), which helps to understand the selective pressures that occur in
domains (RNase 3a and RNase3b) positioned at the C-terminal followed this particular cancer mutation spectrum.
by a dsRNA-binding domain (dsRBD) [5,73]. Dicer works juxtaposed • In different types of inherited tumors such as PPB, non-
with other proteins, including R2D2, fragile X mental retardation 1 epithelial ovarian cancer, Wilms tumor, pituitary blastoma, cystic
(FMR1) in D. melanogaster, RNAi deficient – 4 (RDE-4) in C. elegans and nephroma, rhabdomyosarcoma, and others [54], Dicer1 mutations are
the Argonaute family proteins (Ago family protein) in various other frequently observed. Patients harboring these Dicer1 mutations have
organisms [4]. Lately, it was revealed that D. Melanogaster Dicer -1 de­ curtailed Dicer1 expression and/or disabled Dicer1 function, which
mands Loquacious (LOQS; often referred to as R3D1), which contains causes aberrant expression of miRNAs and contributes to the patho­
three binding dsRNA motifs for pre-miRNA processing [4,7,74,75]. Two genesis of cancer Consequently, the mutation of Dicer1 has deemed a
closely related proteins associated with human Dicer are; interferon- condition with tumor predisposition known as Dicer1 syndrome
inducible double-stranded RNA-dependent activator (PRKRA, also [87,88].
called PACT) [4] and transactivation response RNA- binding protein
(TRBP) [4]. Dicer-associated proteins are not explicitly necessary for 4.8. Mutations of TRBP in cancer
miRNA processing but work in the development of RNA-induced
silencing complexes. Although, research determining specific roles of Impaired TRBP activity illustrates the dysregulation of miRNA in
these proteins is still going on. Upon the development by Dicer cleavage cancer, e.g.:
of an approximately 22 nucleotide miRNA duplexes, an effector complex • Gene sequences, which encodes the miRNA transcription ma­
is generated composed of miRNA duplex incorporated into a complex of chinery, disclosed two frameshift mutations of TRBP in sporadic as well
the Ago family protein. Out of two strands, guide strand or miRNA re­ as hereditary carcinomas along with microsatellite instability [68]. The
mains bound to Ago as mature miRNA while the other passenger or consequence of these mutations was diminished TRBP and Dicer1
miRNA* strand is degraded. After being loaded into RISC in a few cases, expression together with the defective processing of pre-miRNAs. When
miRNA (passenger strand) stays functional. The critical determinant of wild type TRBP was reintroduced into the mutated cell lines, TRBP and
which strand will be degraded and which one will be loaded on Ago Dicer 1 expression was rescued, restoring miRNA processing and sup­
protein to form RISC complex eventually is the thermodynamic stability pressing repressed cancer cell growth in vitro and in vivo [87].
of two strands. Dicer leads to RISC assembly by creating a RISC loading
complex (RLC) in tandem with other interacting proteins (TRBP and/or 5. Mechanism of action of miRNA in gene silencing
PACT in humans and LOQS in fly) and Ago family proteins [4]. RLC’s
function in loading RNA to Ago is still undisclosed. However, evidence Contrary to the perfect complementarity of sequence amid siRNA

6
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

and mRNA, in the majority of miRNA target sites, there are mismatches Table 3
and bulges. In earlier reports, a distinction between siRNA and miRNA Animal miRNAs and their biological functions.
proposed that siRNA destabilizes mRNA, whereas miRNA inhibits MicroRNAs Gene Functionality Species References
mRNA translation without affecting the mRNA level [89]. The degree of targeted
complementarity between short RNA and target, therefore, was thought SV miRNAs SV40 viral Susceptibility to [96]
to be a significant determinant distinguishing between the two mecha­ mRNAs cytotoxic T cells
nisms [90]. miR-375 Myotrophin Insulin secretions M. musculus [97]
miRNA degradation differentiated from siRNA-mediated mRNA miR-273 DIE-1 Neuronal cell fate C. elegans [98]
and
cleavage can be interpreted by mRNA-processing bodies (P-bodies), developmental
which are RNA degradation sites [4]. Conclusively, miRNAs prevent the timing
translation of target mRNAs that are then sequestered into P-bodies and miR-223 NFI-A, Regulation of H. sapiens [99]
degraded. miRNAs also contribute to the degradation of the target Mef2c granulocytic
maturation
mRNAs without sequestration to P-bodies [90,91]. The specificity of
miR-196 HOXA7, Development? M. musculus [100]
miRNA – mRNA interaction is bestowed mainly by a miRNA’s first eight HOXB8,
nucleotides (known as seed sequence) [92]. Not only the seed pairing HOXC8,
but other factors such as the number of target sites, the context of sur­ HOXD8
rounding sequence in mRNA, and the occlusion of target sites by RNA- miR-155 PU-1, c- Maf T-cell Mouse [4]
development and
binding proteins decides whether a predicted target is a genuine target in innate
or not [93]. When miRNAs have a high degree of sequence comple­ immunity
mentarity, then target mRNA degradation processes are facilitated by miR-146 c-Myc, Development and H. sapiens [4]
Ago protein slicer activity. The Ago-catalyzed mRNA degradation pro­ ROCK1 function of the
immune system
cess is responsible for the degradation of mRNA, but other mechanisms
miR-143, Unknown Downregulated in H. sapiens [101]
such as mRNA de-adenylation, de-capping, and exonucleolytic digestion miR-145 colonic
are also involved [4]. Degradation of mRNA by miRNA requires Ago, adenocarcinoma
GW182, and the cellular de-capping and de-adenylation machinery H. sapiens
[89]. miR-143 ERK5 Adipocyte [4]
differentiation
miR-32 Retrovirus Antiviral defense H. sapiens [101]
6. Integral roles of miRNA PFV1
miR-17-92 c-Myc, E2F1 Upregulated in B- H. sapiens [35,102]
miRNAs play significant roles in cell fate determination, prolifera­ cell lymphoma
miR-16 Several AU-rich element H. sapiens [103]
tion, and cell death [94] (Table 3). Besides these essential processes,
mediated mRNA
miRNAs are implicated in several cellular functions, including immune instability
response, insulin secretion, neurotransmitter synthesis, circadian miR-15a, Bcl2 Down-regulated in [32,33]
rhythm, and viral replication [89]. miRNAs are firmly established as miR-16-1 B cell chronic
critical molecular components of cells in both standard and pathologic lymphocyte
leukemia
states in last few years. miR-14 Caspase? Cell death and D. melanogaster [104]
Since past decade, microRNA research has been an area of significant proliferation
focus in cancer therapeutics. Plethora of studies have reported the sig­ miR-7 Notch Notch signaling D. melanogaster [4]
nificance of microRNAs in cancer biology to promote tumor growth, targets
miR-1 HAND 2 Cardiomyocyte Mus musculus [104]
invasion, angiogenesis and immune invasion by regulating the expres­
differentiation
sion of their target mRNAs. The tumor microRNA profiles are also linked and proliferation
to specific subtypes, patient survival and responsiveness to treatment. lsy-6 COG-1 Neuronal cell fate C. elegans [4]
Notably, microRNA biomarkers associated with cancer can be identified and
in biological fluids to enable less invasive monitoring [95]. developmental
timing C. elegans
lin-4 lin-14, lin- Physiological C. elegans [105,106]
7. Cancer correspondence 28 condition and
developmental
miRNA may serve as a tumor suppressor like the protein coding timing
let-7 lin-41, HBL- Regulation of C. elegans [4]
genes in such a manner that any failure of its function could initiate or
1 developmental
contribute to a normal cell’s malignant transformation [34]. miRNA’s timing
functional impairment may be ascribed to several causes, including
genomic deletion, mutation, epigenetic silencing, and/or alterations in
miRNA processing [107]. About 50% of annotated human miRNAs are including PTEN [109,110] and could also facilitate the migration of
clustered in regions of the genome, regarded as fragile sites inferring the tumors by targeting pro-invasion genes. It is important to mention that
crucial role of miRNAs in cancer development [24]. For instance, the miRNA’s over-expression in vivo promotes tumor growth, maintenance,
homolog of C. elegans lin-4, mir-125b-1, is located in a fragile site on and survival [111].
chromosome 11q24, which is deleted in a subset of patients with
ovarian, cervical, breast, lung cancers [23]. In 2002, the first report on 8. The mechanism for MicroRNA deregulation in cancer
miRNAs’ function in cancer showed that in most people with chronic
lymphocytic leukemia (CLL), the gene cluster consisting of the miRNAs miRNAs control the expression of their target mRNAs and over- or
miR-15 and miR-16 are deleted [33]. Additional findings revealed that under expression of miRNAs is expected to lead to down- or upregula­
miR-15 and miR-16 function as tumor suppressors by targeting the tion of the target mRNAs protein product, respectively. It is possible to
oncogene BCL2 encoding a cell survival protein [3]. miR-21 is an link a miRNA in cancer when an oncogene or tumor suppressor gene is a
excellent example of oncogenic miRNA (a so-called oncomir) and is direct target of a miRNA [111]. Tumor tissues and tumor cells often have
overexpressed in most tumors [108]. miR-21 increases proliferation and significantly lower expression of mature miRNAs [10]. Diverse mecha­
decreases apoptosis by targeting several tumor suppressor genes, nisms have been described for the aberrant expression of miRNA. Three

7
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

of them are discussed in detail (i) Genetic alterations and single nucle­ transcription factors relationships were established in cancers [111]
otide polymorphism (SNP), (ii) Epigenetic silencing, and (iii) Tran­ (Table 4). For instance, transcription factor p53 is mutated in 50% of
scriptional regulation [112]. human cancers. Several miRNAs are transcriptionally regulated with
p53 [107]. Profiling of miRNA levels of expression after induction of p53
8.1. Genetic alterations and single nucleotide polymorphism (SNP) by genotoxic stress suggested that the miR-34 family members -a, -b and
-c were the most strongly upregulated miRNAs (Fig. 2), with the tran­
In 2004, it was reported that around 50 percent of miRNAs are scription of pri-miRNAs from both miR-34a and miR-34b/c loci acti­
located in fragile sites and cancer susceptibility loci [30]. Since then, vated in a p53-dependent manner [107]. The miR-34 family’s mRNA
more comprehensive mapping of human miRNA genes on fragile sites, targets comprise cyclins D and E2, cyclin-dependent kinases 4 and 6
cancer-specific translocation breakpoints, repetitive sequences and CpG (CDK4 and CDK6), CDC25c, Myc, and BCL2 [107]. Considering the roles
islands was carried out and demonstrate that miRNA genes are associ­ of these different target genes in promoting cell proliferation and
ated with fragile sites [113]. miRNAs express themselves aberrantly in a inhibiting cell cycle arrest and apoptosis, miR-34 induction by p53
variety of cancers. First found in miR-15a and -16-1, which are clustered strongly complements its role in negative cell growth regulation [107].
in chromosome 13q14, a commonly deleted region of B cell chronic
lymphocytic leukemia (CLL)and other cancers [111]. These two miRNAs 9. MicroRNAs as oncogenes and tumor suppressors
were reduced in cancer samples compared to the healthy tissues. In
addition to structural genetic changes, somatic translocations of the Although the available expression profiling data have documented
miRNA target site resulting in target mRNA escape from specific miRNA almost omnipresent dysregulation of miRNA expression in patterns of
[114]. Mutations that alter a miRNA seed sequence could potentially human cancers [109,129,130], miRNA expression is highly specific for
ablate tumor-suppressive miRNAs or allow altered target selection that cell type and cellular differentiation status. Much of the aberrant miRNA
could contribute to oncogenesis. Although natural sequence variants, expression observed in tumors is, therefore, likely a secondary conse­
such as SNPs, were demonstrated to influence miRNA targeting in quence of the loss of healthy cellular identity that accompanies malig­
cancer-related pathways [115]. Cancer cell sequencing revealed that nant transformation. Moreover, up- or down-regulation of a miRNA is
while mutations contained in primary transcripts of miRNA, there was not necessarily indicative of a causative role in tumorigenesis in a given
no proof of mutations that altered the mature miRNA sequence [77]. tumor type. At least four types of evidence reinforce the case that a
miRNA works as a tumor suppressant or oncogene: 1) Data showing
8.2. Epigenetic silencing widespread dysregulation in the various cancers; 2) Gain or loss of
miRNA function owing to deletion, amplification or mutation in tumors;
A conventional peculiarity of cancer cells is aberrant epigenetic 3) Precise monitoring of tumor suppression or tumor development using
changes for instance DNA hypermethylation of tumor suppressor genes, animal models; 4) Identifying and verifying cancer-relevant targets that
extensive hypomethylation of genomic DNA, and transition of histone enlighten mechanisms by which miRNA participates in oncogenesis
modification patterns [116]. Also, miRNA genes have epigenetic [129]. The miRNAs that satisfy some or all of these parameters are
changes in cancer in a similar fashion as protein-coding genes. A sig­ known to be strong candidates as tumor suppressors and oncogenes are
nificant proportion of miRNA loci are associated with CpG islands, listed below Table 5.
which provide a strong basis for their DNA methylation regulation. Most
studies have used chromatin remodeling drug treatment to reveal 9.1. Tumor suppressor miRNAs
epigenetically silenced miRNAs. For example, hypermethylated tumor-
suppressing miRNAs; miR-127 [117], miR-9-1 [118] and the miR-34b/ 9.1.1. miR-17-92 cluster
c cluster [119] was upregulated by 5-aza-20 deoxycytidine treatment. miR-17-92 cluster, found in humans at chromosome 13q31.3, con­
Likewise, differential miR-124a expression was observed in miRNA sists of six miRNAs (mir-17, -18a, -19a, -20a, -19b-1, and-92a-1). A
profiles of DNMT1- and DNMT3b-enzyme deficient colorectal cancer similar cluster exists at chromosome 10 known as miR106a-363, which
cells [120]. By contrast, potentially oncogenic miRNAs can be upregu­ also includes six miRNAs (mir-106a, -18b, -20b, -19b-2, -92a-2, and
lated by the hypomethylation of DNA [121]. Epigenetic silencing of a -363). The miR-17-92 cluster includes the first miRNAs proven to be
miRNA may also be a result of the specificity of the tissue. miR-124a, for oncogenic [89]. Owing to the forced expression of the miR-17-92 cluster
example, is typically highly expressed in neuronal tissues, so its epige­ in transgenic mice overexpressing the c-myc oncogene, the development
netic suppression in colorectal tumors is not unexpected [111]. Addi­ of B cell lymphoma is significantly accelerated [102]. The miR17-92
tional epigenetic phenomena disrupted in cancer comprehends histone cluster is upregulated in a variety of cancers, including lymphomas
acetylation and DNA methylation [4]. A reduction of the number of [102,131], lung cancer [6] and others [132], consistent with its onco­
acetylated histones will decrease the expression of anti-oncogenic genic role. Two mechanisms are apparent for the upregulation of this
miRNAs, as illustrated by studies utilizing histone deacetylase in­
hibitors where an alteration in the miRNA levels was observed post-
Table 4
treatment [117,122]. Specific miRNAs regulate components of the
MicroRNAs (miRNAs) regulated by transcription factors.
epigenetic machinery. For example, the miR-29 expression can inhibit
Transcription miRNA targeted Cancer provoked References
the expression of DNMT3A and DNMT3B [123], thus counteracting
factor
aberrant DNA methylation. miR-29 expression restoration in non-small-
cell lung cancer cells led to the de-repression of tumor suppressor genes Myc miR-17-92 cluster Angiogenesis in colon [36]
cancer model
silenced by CpG island methylation [123]. Furthermore, miR-101 tar­
E2F3 miR-17-92 cluster [89]
gets histone methyltransferase EZH2 which endows the epigenetic MYCN miR-17-5p, -92, Neuroblastoma [89]
silencing of target genes and regulates cancer cell survival and metas­ -320
tasis [124]. Twist miR-10b Breast cancer [125]
p53 miR-34a, miR-34b- Cell lines [126]
34c cluster
8.3. Transcriptional regulation p53 miR-34a Cell lines [127]
p53 miR-34a, miR-34b- Cell lines and in vivo [128]
Several transcription factors may induce miRNAs by activating the 34c cluster
transcription of pri-miRNAs; consequently, it is also true that oncogenes Stat3 miR-21 Myeloma cell lines [89]
E2F1,2,3 miR-17-92 cluster [127]
or tumor suppressors are transcription factors. Several miRNAs –

8
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Fig. 2. miRNA biogenesis in animals. The microRNA


(miRNA) gene is transcribed by RNA polymerase II
(pol II) to yield primary microRNA (pri-miRNA)
which is then processed within the nucleus by a
protein complex called ‘Microprocessor complex’
composed of DGCR8 (DiGeorge syndrome critical
region 8) (Pasha in flies) and Drosha to yield 85-
nucleotide long precursor microRNA (pre-miRNA).
pre-miRNA is recognized by a nucleocytoplasmic
transporter comprised of exportin 5 (XPO5) and Ran-
GTP and transports it to the cytoplasm. In the cyto­
plasm, pre-miRNA is further processed by RNase III
DICER and it’s two closely related proteins PACT
(protein kinase, interferon-inducible double-stranded
RNA dependent activator) and TRBP (Transactivation
response RNA-binding protein) which forms RISC
(RNA induced silencing complex) to generate an
approximately 22 nucleotide miRNA duplex, ‘effector
complex’ composed of miRNA duplex (miRNA:
miRNA*) incorporated into Ago-family protein. Out
of the two strands, ‘guide strand ‘or miRNA remains
bound to Ago as mature miRNA while the other
‘passenger strand’ is degraded. Ago proteins’ endo­
nucleolytic enzymatic activity is responsible for the
removal of the passenger strand. Mature 22 nucleotide miRNA recognizes complementary sequence in the 3’- untranslated region of target mRNA and represses its
expression either by its degradation or translational repression.

cluster in cancers (Fig. 3). regulated by let-7 through 3’UTR binding, which inhibits translation
The first is amplification of the chromosome locus 13q31 across [139]. Let-7 behaves as a tumor suppressor and mediates Ras repression
several lymphomas and other cancers [11,131,133,134]. Furthermore, and modulates the cellular processes [5,111,139].
the other one is transcriptional activation of the pri-miRNA, where c- A non-histone architectural transcription factor HMG2A alters DNA
Myc (an oncogenic transcription factor) binds the genomic locus up­ conformation to direct transcriptional activation of a variety of genes
stream of the miR-17-92 cluster and activates its expression [35]. MYCN, that influence cell growth, differentiation, proliferation, and survival
a protein highly homologous to c-Myc, appears to activate this cluster in [140]. Though HMG2A is undetectable in healthy adult tissue, it is
neuroblastoma cells instead of c-Myc [135]. E2Fs activate the cluster as highly expressed in embryonic tissues, lung cancer, and uterine leio­
well [136,127]. Since the E2Fs are direct targets of miR-17and-20a, the myomas [5]. let-7s regulate HMG2A by destabilizing its mRNA through
miR-17-92 cluster establishes a complex regulatory network with c- 3’UTR binding [111,114,141]. Loss of let-7 regulation of HMG2A
Mycs and E2Fs. E2F1 and c-Myc activate each other to establish a pos­ stimulates cell growth and proliferation [107,136]. Recent data suggest
itive feedback loop. Both E2Fs (E2F1, -2 and -3) are capable of triggering that let-7s play a more significant role in regulating cell proliferation
the activation of the miR-17-92 cluster and are susceptible to repression than initially thought, as multiple cell cycle regulators including c-myc,
from this cluster by miR-17 and-20a. Though degrees of activation and CDC25A, CDK6, and cyclin D2 are functionally inhibited by let-7 [5,81].
repression vary between individual miRNA pairs and members of the
E2F family. In general, E2Fs are thought to be proliferative, but E2F1is 9.1.3. miR-15a and miR-16-1
also proapoptotic. c-Myc and E2Fs switch on the miR-17-92 cluster at a The miR15/16 miRNA family consists of 4 members who seem to be
proliferative signal. E2Fs are repressed by this cluster, thereby pre­ putative tumor suppressors. These are grouped into two independent
venting uncontrolled amplification of the positive feedback loop be­ clusters, miR15a / miR16-1 at 13q14.3 and miR15b / miR16-2 at 3q26
tween E2Fs and c-Myc. Additionally, the repression of E2F1 by miRNAs [5]. All four miRNAs share a seed region of 9 nucleotides, which targets
can help to minimize E2F1’s proapoptotic potential (Fig. 3). the 3’UTR of the antiapoptotic protein BCL2 for post-transcription
repression [5]. The oncogenic protein BCL2, typically found in
9.1.2. let-7 different tumors and hematopoietic malignancies, facilitates cell sur­
Initially described in C. elegans, let-7 was identified in a mutant vival by preventing apoptosis [5]. The miR15a/16-1 miRNA cluster is a
screen-seeking gene-regulating developmental timing [2]. Loss of this regular deletion site in 68% of patients with chronic B cell lymphocytic
miRNA function avoids a natural shift from late larval to adult cell fates. leukemia (CLL) [33]. In this scenario, the absence of miR15a/16-1 is
let-7 upregulation in adult stem cells is necessary for terminal differ­ correlated to a loss of regulation, resulting in elevated levels of BCL2
entiation and cell cycle exit. Without miRNA, the cells fail to differen­ proteins [5]. BCL2 blocks apoptosis by inhibiting the release of mito­
tiate and continue to divide appropriately, a phenotype similar to the chondrial cytochrome C necessary to activate the caspase pathway of
behavior of some cancer cells [129]. Many let-7 genes were mapped to enzymes responsible for guiding programmed cell death [142,143].
regions within the human genome that are often altered or deleted in Regular miR15a/16-1 expression by mediating BCL2 repression induces
different cancers [30]. This finding, along with many other types of intrinsic apoptosis pathways [5]. miR15b / miR16-2 have similar out­
research, implicated let-7 as a tumor suppressor. Oncogenes Ras and comes as described for miR15a/16-1 [5]. Furthermore, evidence indi­
high mobility group AT-hook 2 (HMGA2) are the two well-defined let-7 cated that miR15b / miR16-2 plays a role in regulating chemosensitivity
targets [5]. as BCL2 suppression via miR15b / miR16-2 reduces the sensitivity of a
Ras is a signal transmitting GTPase that transmits signals from cell cell to drug-induced apoptosis [5].
surface receptors to functional intracellular pathways that affect cell
proliferation, growth organization of cytoskeletons, cell movements and 9.1.4. miR-143 and miR-145
survival [5]. In a variety of human cancers, including pancreas, colon, Even though no causative role has been established in tumorigenesis,
thyroid and lung, constitutively active Ras mutants (H-Ras, K-Ras, and decreased expression of miR-143 and miR-145 is a frequent feature of
N-Ras) are found [137,138]. Ras, K-Ras, and N-Ras expression are colorectal tumors [101]. miR-145 downregulation is highly prevalent in

9
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Table 5
MicroRNAs function in human cancer as oncogenes or tumor suppressor genes [79,129].
MicroRNA Chromosome Tumor Profiling method Target Tumor Note
suppressor/
Oncogene

miR-15 and 13q14.2 Chronic Lymphocytic Bead BCL2 Tumor miR-15 and miR-16 target BCL2 ultimately led to
miR 16-1 3q25.33 Leukemia CloningArray Suppressor apoptosis in a model of the leukemia cell line.
Colon cancer Array qRT-PCR COX-2 miR-16 targets COX-2; elevated HuR levels
Cloning miSAGE antagonize the function of miR-16;
Follicular Lymphoma CHK-1 miRNA profiles are linked to increased
proliferation and late germinal-center B cell
phenotype
Fibroblast CEBPβ, CDC-25a, Upon re-entry of the cell cycle, the accelerated
CCNE1 degradation of miR-16 alleviates the suppression
of the target genes, enabling the cell cycle to be
properly resumed
Fibroblast VEGF, VEGFR2, miR-16 plays an important role in the
FGFR1 modulation of endothelial cell-intrinsic
angiogenic behavior
Cancer-associated fibroblast FGF2, FGFR1 miR-15 and miR-16 downregulation of cancer-
associated fibroblasts promotes tumor growth
and progression
not known CCNE1 miR-15 and miR-16 are established E2F
transcriptional targets
Multiple myeloma FGFR1, PI3KCA, miR-15/16 deletion is found in the early stages of
MDM4, VEGFa multiple myeloma
Breast cancer Bead qRT-PCR WIP1 miR-16 regulates WIP1 phosphatase in
Array mammary tumorigenesis and DNA-damage
response
Ovarian cancer Array BMI-1 BMI-1 targeted by miR-15a and miR-16 leads to
low proliferation and clonal growth
Lung cancer CCND1, CCND2, miR-15/16 over-expression contributes to
CCNE1 detention at G1-G0
Colon cancer Array qRT-PCR SIRT1 miR-34 targets SIRT1, which leads to apoptosis in
Cloning miSAGE wild type p53 only
miR-34 1p36.22 Gastric cancer BCL2, NOTCH, Tumor miR-34 targets NOTCH, BCL2, and HMGA2
11q23.1 HMGA2 suppressor
Fibroblast MYC miR-34a inhibits MYC during senescence and
regulates many cell-cycle regulators
Lung cancer AXL miR-34a and miR-199a / b targets the AXL
receptor; the promoter methylation silences both
miRNAs
Ovarian cancer Array MET MET miR-34 targets MET
Colon cancer Array SNAIL 1 A new link has been identified between p53, miR-
qRT-PCR 34, and SNAIL1 in the regulation of epithelial-to-
Cloning miSAGE mesenchymal transition programs for cancer
cells
Lung cancer Array KRAS The let-7 family negatively regulates let-60/
KRAS and lung tumors in Caenorhabditis elegans
let-7 family 9q22.32 Not known HMGA2 Tumor Chromosomal translocations interrupt HMGA2
Xp11.22 Suppressor repression by let-7 miRNA
22q13.31 Burkitt lymphoma MYC Dysregulation of let-7 is involved in the genesis
21q21.1 and maintenance of Burkitt lymphoma and other
19q13.41 cancers with MYC dysregulation.
11q24 Not known IMP- 1 Let-7 regulated oncofetal proteins could be novel
13q21.1 therapeutic targets and potential cancer
12q14.1 biomarkers for treatment.
Not known DICER There is a regulatory mechanism to regulate
DICER and multiple miRNAs in an equilibrated
state.
Fibroblast CDC-32 Let-7 suppresses CDC-34, stabilizes WEE1 kinase,
and raises the cell fraction of G2-M in primary
fibroblasts
Breast cancer Bead IL-6 Inflammation activates a positive feedback loop,
qRT-PCR which maintains the transformed epigenetic state
Prostate cancer Array E2F2, CCND2 E2F2 and CCND2 are targeted by let-7a, which
act as a tumor suppressor in prostate cancer
Liver cancer BCL-XL Let-7 targets BCL-XL and can potentiate
apoptosis caused by sorafenib
Breast cancer Bead ZEB1 and ZEB2 miR-200 family down-regulation can be a
qRT-PCR significant step in tumor progression
Array
miR-200 1p36.33 Bladder cancer ERRFI-1 Tumor miR-200 restores dependency on EGFR
family 12p13.31 Suppressor
Nasopharyngeal carcinoma ZEB1, CTNNB miR-200a blocks the growth, migration, and
invasion of cells
Pancreatic cancer BMI-1
(continued on next page)

10
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Table 5 (continued )
MicroRNA Chromosome Tumor Profiling method Target Tumor Note
suppressor/
Oncogene

By suppressing the miR-200 family, ZEB1 links


epithelial-to-mesenchymal transition and
stemness maintenance and thus promotes
migrations.
Breast cancer Bead PLCγ1 miR-200 negatively regulates the invasion,
qRT-PCR viability and cell-cycle progression of EGF-driven
Array cells
Not known FAP1 miR-200c sensitizes cells to CD95-mediated
apoptosis
Breast cancer Bead SUZ12 miR-200b – Suz12–cadherin pathway supports
qRT-PCR growth and invasiveness of cancer stem cells
Array
Lung cancer FLT1, VEGFR1 miR-200 suppresses metastasis by FLT1 targeting
not known JAG1, MALM2, Explains increased NOTCH signaling in certain
MALM3 types of cancers where mutations in the NOTCH
genes are rare.
Breast and endometrial FN1, LEPR, NTRK2, miR-200c blocks cell motility and anoikis
cancer ARHGAP19 resistance
Ovarian cancer Array p38α Signature of miR200a-dependent stress
correlates with better survival and treatment
response
miR-17/92 13q23.1 Colon Array TSP-1, CTGF Oncogene Upregulation of KRAS, c-Myc, and non-
qRT-PCR functional p53 in colonocytes
Cloning miSAGE
Prostate cancer, Burkitt E2F2, E2F3 There is an autoregulatory feedback loop
lymphoma, testis carcinoma between the factors E2F and miR-17/92
Myc-induced lymphoma BIM, PTEN Transgenic mice have a greater lymphocyte
expression of miR-17/92
Lung cancer Array HIF1α A complex and finely tuned circuit that includes
c-Myc, miR-17/92, and HIF1α
Cervix tumor cell line Array PTPRO PTPRO controlled by E2F1 and miR-17/92
Myeloid cell p63 By suppressing p63, miR-92 increases cell
proliferation
T cell acute lymphoblastic BIM, PTEN, Functional genomics reveals miR-19 repression
leukemia PRKAA1, PPP2R5e of regulators of PI3K survival signals.
Endothelial cell JAK1 miR-17/92 family provides a therapeutic
perspective to improve angiogenesis in the
therapy
Breast cancer Bead HBP1 miR-17/92 family inhibits HBP1 by activating
qRT-PCR Wnt/β-catenin and regulates invasion
Array
Ras-induced senescent p21WAF1 Senescence disrupted by miR-17/92 family
fibroblast
Glioblastoma Array TGF-βII, SMAD4 The family miR-17/92 suppresses TGF-β,
promoting angiogenesis and development of
tumor cells
Prostate cancer Array MnSOD, GPX2, miR-17/92 family suppresses tumorigenicity by
TRXR2 inhibiting antioxidant enzymes in the
mitochondria
miR-222/ Xp11.3 Glioblastoma, prostate, and Array p27Kip1 Oncogene High miR-222/221 keeps p27Kip1 low and
221 thyroid carcinoma boosts proliferation
Normal fibroblast p57Kip2 Upregulation initiates S phase with signaling
pathways for the growth factor that induce cell
proliferation
Non–small cell lung cancer PTEN, TIMP3 Target PTEN and TIMP3, induce TRAIL
and hepatocellular resistance and improve cell migration; MET
carcinoma oncogene activates miR-222/221 by
transcription factor c-Jun
Breast cancer Bead FOXO3A Aim FOXO3A at the transcriptional stage to
qRT-PCR suppress p27Kip1
Array
Endothelial cell KIT miR-222 targets c-Kit, which controls endothelial
cells ability to form new capillaries
Breast cancer Bead ESR1 Modulation of ERαis linked to antiestrogen
qRT-PCR therapy
Array
Glioblastoma Array PUMA Regulating apoptosis directly by targeting PUMA
Breast cancer Bead TRSP1 Promote a transition from epithelial to
qRT-PCR mesenchymal and contribute to the more
Array aggressive clinical behavior of basal-like breast
cancers
Non–small cell lung cancer PTPμ Target PTPμ and regulate tumorigenesis by
glioblastoma
Breast cancer DICER DICER inhibited in ERα-negative breast cancers
(continued on next page)

11
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Table 5 (continued )
MicroRNA Chromosome Tumor Profiling method Target Tumor Note
suppressor/
Oncogene

Bead
qRT-PCR
Array
Non–small cell lung cancer APAF1 Activated by EGFR and MET; targeting APAF1,
responsible for the resistance to gefitinib
miR-21 17q23.1 Cholangiocarcinoma cinoma PTEN Oncogene Gemcitabine-induced apoptosis modulated by
PTEN-dependent activation of PI3K
Breast cancer Bead TPM1 Suppression hinders the growth of tumors
qRT-PCR
Array
Not known SPRI1 Null- mice showed a significant reduction in the
formation of papilloma’s compared to wild mice
Glioblastoma Array RECK, TIMP3 Inhibition offers a new therapeutic approach for
the physiological modulation of multiple
proteins whose expression in cancer is
deregulated
p63, JMY, TOPORS, Several important components of p53, TGF-β,
TP53BP2, DAXX, and mitochondrial apoptosis tumor-suppressive
HNRPK, TGF-βRII pathways.
Prostate cancer Array MARKS Fosters tolerance to apoptosis, motility, and
invasion
miR-155 21q21.3 Breast cancer Bead SOCS1 Oncogene May act as a link between inflammation and
qRT-PCR cancer
Array
Acute myeloid leukemia Bead CEBPB, PU.1, Contributes to the physiological expansion of
qRT-PCR CUTL1, PICALM granulocytes/macrophages during inflammation
Array and certain pathological characteristics
associated with acute myeloid leukemia
not known BACH1, ZIC3 Epstein-Barr virus (EBV) induction leads to an
EBV-mediated signaling
Human cord blood CD34+ ETS1, MEIS1 Needed for proliferation and differentiation of
megakaryocytes
Lymphocyte C-MAF BIC / miR-155 plays a central role in the
homeostasis and immune system function
Diffuse large B cell HGAL Cell proliferation and aggressiveness are a
lymphoma symptom of diffuse large B cell lymphoma,
usually with high levels of miR-155 and no
expression of HGAL
Nasopharyngeal carcinoma JMJD1A LMP1 and LMP2A trigger miR-155 for JMJD1A
suppression
Breast cancer Bead WEE1 WEE1 targets and improves mutation rates by
qRT-PCR reducing the effectiveness of the DNA safeguard
Array mechanisms
Pancreatic cancer Array TP53INP1 not known
qRT-PCR of pre-
miRNA
not known SMAD1, SMAD5, Role in the control of cellular processes mediated
HIVEP2, by BMP
CEBPB,
RUNX2,
MYO10
Breast cancer Bead Molecular linkages between miR-155 and
qRT-PCR FOXO3A influences cell survival as well as
Array chemotherapy response
Colon cancer Array hMSH2, hMSH6, Inactivated mismatch repair by hMSH6, hMLH1
qRT-PCR hMLH1
Cloning miSAGE
Diffuse large B cell SMAD5 not known
lymphoma
miR-191 3p21.31 Prostate cancer RNA isolation TIMP3 Oncogene Prostate cancer cell growth and invasion
and qRT-PCR regulated by miR-191 via targeting tissue
inhibitor of metalloprotease 3 (TIMP3).

breast carcinomas and breast cancer cell lines [109]. These miRNAs are regulation of this miRNA and to determine the pathological signifi­
contained within a frequently deleted genomic interval of the myelo­ cance of the loss of miR143/145 expression [129].
dysplastic syndrome [30]. Ironically, the drop-in colon cancer abun­
dance of miR-143/145 is attributed to a block in Dicer processing since 9.2. Oncogenic miRNAs
the miRNAs precursor (pre-miRNAs) exhibits natural abundance [129].
These miRNAs occur at low levels in several cell lines, including those 9.2.1. BIC/miR-155
originating from the breast, cervical and lymphoid cancers, but pre- B-cell integration cluster (BIC) non-coding RNA was described as a
miRNA is often detectable, again indicating a block in the Dicer pro­ transcript arising from a specific retroviral integration site in chicken
cessing [101]. Future studies need to confirm the post-transcription lymphoma cells caused by avian leukosis virus [144]. But, this non-

12
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Fig. 3. Diagrammatic illustration of miRNA that


contributes to metastasis. Metastasis progresses by a
succession of steps: local invasion, intravasation,
extravasation, and colonization (as depicted by the
yellow boxes). Protein-coding genes and microRNAs
(miRNAs) that assist metastasis are (shown in blue),
those who prevent are (shown in red) and the inhi­
bition is depicted by (red arrow). miR-200 effectively
suppresses the action of the mesenchymal markers
zinc finger E-box-binding homeobox 1 (ZEB1) and
ZEB2 in sustained cells. As a consequence, ZEB1 and
ZEB2 expression are increased during the epithelial to
mesenchymal transition (EMT) as the miR-200 levels
are declined. E-cadherin, a cell adhesion molecule
deficient in aggressive and metastatic tumors, is
transcriptionally repressed by ZEB1 and ZEB2; hence
cells deficient in miR-200 are more adept to spread
and invade adjoining tissues. Owing to its miRNA-
processing ability DICER is positioned upstream of
miR-200 DICER1 itself is positively regulated by the
full-length isoform of p63 (TAp63) at the transcrip­
tional level and is inhibited by miR-103, miR-107, and let-7. Besides blocking the expression of DICER, let-7 also inhibits the oncogenic RAS – RAF – MEK pathway
and HMGA2, an upstream SNAIL activator. SNAIL blocks E-cadherin, contributing to local invasion and intravasation. Anti-metastatic miRNAs beyond the E-cadherin
pathway comprise miR-10b, which suppresses the expression of HOXD10; miR-335, which impairs SOX4 and tenascin C (TNC) signaling; and miR-31, which inhibits
all metastasizing steps via inhibiting integrin-α5, radixin (RDX), and RHOA expression RKIP, a protein inhibitory of RAF kinase.

coding RNA, and its final exon, was later shown to promote MYC TPM1 is an actin-binding protein active in regulating anchorage-
mediated lymphomagenesis in a chicken model [145]. Today, it is a fact independent development and organizing cellular microfilaments.
that BIC is a pri-miRNA, and miR-155 is in the final exon of this tran­ miR-21 repression of TMP1 is hypothesized to contribute to cytoskeleton
script [146]. BIC / miR-155 was, therefore, the first miRNA transcript modifications that facilitate neoplastic transformation, cell invasion,
undoubtedly demonstrated to have tumor-promoting behavior [129]. and metastasis [155]. Likewise, suppression of PDCD4 and mapsin fa­
Perfectly aligned with the oncogenic properties of this miRNA, BIC / cilitates cancer progression by promoting cell invasion and metastasis
miR-155 overexpression was repeatedly found in various types of tu­ [5]. These molecules are concerned with apoptosis and urokinase re­
mors, including B-cell lymphoma, and breast, lung, colon and thyroid ceptor regulation (uPAR), which is complicit in extracellular matrix
cancers [146,130,132]. Transgenic mice with BIC / miR-155 driven by degradation [5]. Thus, depicting a role in the progression and metastasis
the B-cell-specific Em enhancer were recently generated. Remarkably, of tumors.
these mice established a polyclonal B-cell malignancy rapidly, indi­
cating that this miRNA mutation is adequate to cause lymphomagenesis 9.2.3. miR-372 and miR-373
in combination with little or no other genetic changes [147]. The evi­ Oncogenic activation of Ras in primary cells results in an irreversible
dence available hence strongly implies BIC / miR-155 as an oncogene arrest of growth, called cellular senescence. Acquisition of additional
[129] though the pathways by which BIC / miR-155 induces tumori­ genetic anomalies, such as loss of p53 enables cancer cells to overcome
genesis are still unknown. this response and to continue proliferation [156]. Using a novel retro­
viral miRNA expression library, Agami and colleagues conducted a cell-
9.2.2. miR-21 based screen to recognize miRNAs that could replace p53 loss when
Oncogenic function of miR-21 was came into notice, while screening over-expressed and enable continued proliferation in the Ras activation
abnormally expressing miRNAs in glioblastoma [148]. miR-21 is an context [157]. Such miRNAs potentiate Ras-mediated cellular transition
antiapoptotic factor that downregulates genes associated with in addition to preventing the Ras-induced cellular senescence. These
advancing apoptosis, which is overexpressed in human glioblastoma miRNAs do not downregulate p53 in response to Ras activation as miR-
tumors [148]. Subsequently, miR-21 overexpression was observed in a 372- and miR-373-expressing cells show normal induction of p53 and its
variety of human cancer, including breast, colon, liver, brain, pancreas, downstream target p21. These miRNAs make cells resistant to the in­
and prostate cancers [5]. It has been revealed that miR-21 down­ fluence of a typical p53 allele. The authors hypothesized that tumors
regulates four tumor suppressor genes: mapsin, programmed cell death associate. Indeed, testicular germ cell tumors, which rarely show p53
4 (PDCD4), tropomyosin1 (TPM1), and phosphatase and tensin homolog function loss, often exhibit miR-372/373 overexpression with normal
(PTEN) [149]. miR-21 binds directly to the 3’UTR of the gene transcripts p53 function may show pathological elevation of miR-372 and miR-373.
preventing their translation. Repression of these tumor suppressor genes Such results together offer compelling evidence of an oncogenic function
by miR-21 promotes cell transformation, growth of tumors, invasion, for such miRNAs [129].
and metastasis [5,150]. Questioning the processes underlying miR-372/373 oncogenic
PTEN is a phosphatidylinositol phosphatase (PIP) that synchronizes behavior, the authors monitored the function of cycline-dependent ki­
with phosphoinositide 3kinase (PI3 K) to balance the levels of PIP3 that nase 2 (CDK2) in the miRNA expressing cell lines.Induction of p21 (an
regulate the Akt pathway [151]. To maintain the essential equilibrium efficient CDK inhibitor) after activation of Ras typically results in CDK2
of PIP3 levels, PTEN dephosphorylates PIP3, and PI3K phosphorylates inhibition and a subsequent cell cycle arrest [158]. Surprisingly, CDK2
PIP2 [152]. Translational PTEN repression by miR-21 results in an activity remained elevated in the presence of activated Ras despite
accumulation of PIP3, which over activates the Akt pathway, triggering regular p21 expression, when miR-372/373 was expressed. Down­
several downstream pathways that promote cell growth and survival regulation of the large homologous tumor suppressor 2 (LATS2), a
[5]. Besides, PTEN loss is associated with an increase in focal adhesion recognized inhibitor of CDK2, was revealed by the microarray analysis
kinase (FAK) activity that facilitates cell proliferation and metastasis by of miR-372/373-expressing cells [159]. LATS2’s 3’ UTR harbor possible
increasing multiple matrix metalloproteases expression [150,153,154]. binding sites miR-372/373, and luciferase reporter assays reinforce the

13
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

direct regulation of LATS2 by miR-373/373. Data recommend a model for the miRNAs’ pro-proliferative function since artificial p27(Kip1)
in which miR-372 and miR-373 decreased LATS2 expression, hence knockdown represents the phenotype of these miRNAs. miR221 and
releasing CDK2 from repression and encouraging cellular proliferation -222 are over-expressed in other cancers, which imply they play a role in
to continue in the existence of active Ras [129]. a broad spectrum of cancers. In addition to regulating p27(Kip1), miR­
NAs also regulate certain proteins in the cell cycle including Cdk6,
9.2.4. The miR-222/221 Cluster Cdc25A, cyclin D2, Cdk4, Rb-family protein and p180 DNA polymerase
The miR-222/221 cluster includes the miRNAs that are often upre­ α [89].
gulated in human cancers, notably hepatocellular carcinoma, where
70–80% of cases display a substantially elevated level [160]. Callegari 10.4. Invasion and metastasis
et al. [161] explained the oncogenic repercussions of overexpression
miRNA (Fig. 4). It has been documented that liver-specific over­ Malignant tumor characteristics that are diverse from benign tumors
expression of miR-221 induced elevated predisposition to the develop­ comprise invasion and metastasis. Malignant tumors are fatal mostly
ment of hepatic tumors. [161]. since they are dexterous of invading neighboring tissues capable of
Oncogenic and tumor suppressive miRNAs have been summarized in metastasizing to remote organs by the means of the bloodstream. miR-
Table-5. 125 ectopic expression has been reported to impair cell motility and
invasion in a breast cancer cell line [89]. The miR-21 as already dis­
10. MicroRNAs governed aspects of cancer biology cussed in above sections, promotes cell motility as well as invasion by
targeting PTEN (a tumor suppressor thought to impede cell invasion
10.1. Angiogenesis through suppressing the expression of various matrix metalloproteases)
[89]. miR-10b is another miRNA that is involved in metastasis and has
Blood vasculature recruitment is critical for the subsistence of been found to be upregulated in metastatic breast cancer cells [125].
neoplastic cells. miR-17-92 cluster was distinguished in this process. c- The ectopic expression of miR-10b facilitates the invasion, intra­
Myc stimulates neovascularization through the downregulation of the vasation, and metastasis of otherwise non-invasive or non-metastatic
anti-angiogenic factor Tsp-1. c-Myc suppresses Tsp-1 and the associated breast cancer cells. miR-10b directly targets HOXD10, the reduction of
protein, connective tissue growth factor (CTGF) by activating the miR- which induces the expression of the well-characterized premetastatic
17-92 cluster. The direct targets of miR-19 and-18 are Tsp-1 and CTGF gene RhoC [125].
respectively [89]. miR-17-92 cluster’s ectopic expression is sufficient for
promoting angiogenesis [36]. Other miRNAs —miR378 and -27a—may 10.5. Programmed cell death
play a role in angiogenesis [111,162]. Tsp-1 is a direct target of Kaposi’s
sarcoma-associated herpesvirus (KSHV) miRNAs, which signifies that Although an equilibrium between proliferation and apoptosis is
viral miRNAs may also play a role in angiogenesis [163]. crucial for tissue homeostasis and proper differentiation, abnormal
apoptosis can correspond to tumors. By directly targeting the anti­
10.2. p53 apoptotic genes miRNAs contribute to tumorigenesis. For example,
repression of Mcl-1 and Bcl-2 antiapoptotic genes miR-29b [89] and by
p53, a transcription factor acts as the genome’s guardian because of miR-34s [126], -15a and -16” [32], respectively. The loss of such miR­
its critical role in cell cycle regulation and apoptosis against genomic NAs due to p53 mutation or 13q14 chromosome deletion contributes to
damage [89]. It modulates the transcription of several target genes when enhanced antiapoptotic gene expression and tumor cell survival that
stimulated by genotoxic stress and activation of oncogenes and functions would usually be eliminated by apoptosis. miRNAs are likely to target
as a tumor suppressor. Its significance is highlighted in nearly 50 percent other genes in the apoptotic pathway as apoptosis is linked with the
of human cancers as it has been reported to be mutated [89]. miRNA transfection or expression of a variety of miRNAs [89].
profiling after p53 induction via genotoxic stress has documented miR-
34a, -34b, and-34c (collectively, miR-34s) to be the most upregulated 11. Tumor sensitization to chemotherapy
miRNA [89,128]. miR34b and -34c are clustered in chromosome 11,
while miR-34a is separately located in the genomic locus. p53 directly The capability of miRNAs or antagomirs to stimulate resistant cells to
activates both pri-miRNAs. The miR-34s appear to be critical down­ frequently employed therapeutic agents is analyzed under miRNAs’
stream effectors of p53 since the miR-34’s ectopic expression re­ sheer ability to target signaling pathways that are frequently disturbed
capitulates the phenotype of p53 activation [89]. It has been reported in cancer. Since the field is evolving, in vivo analysis is limited; never­
that miR-34s promotes cell cycle arrest, apoptosis and senescence [89]. theless, more miRNA-based sensitization studies in preclinical animal
The repression of several miR-34 direct targets such as Bcl-2, Cdk4 and models are likely to be revealed soon owing to reassuring in vitro ex­
the receptor of the hepatocyte growth factor explains these effects periments [166]. Resistant cancers do not respond to therapy due to
[164]. In addition to the miR-34s, numerous different miRNAs could altered expression of genes conferring resistance against drugs. Since
also be significant in the p53 pathway. miR30c, -103, -26a, -107 and resistant cancer possess efficient DNA repair capability, alterations in
-182 were distinctly, albeit less robustly, triggered by p53-dependent cell cycle checkpoints and apoptosis regulation therefore, inhibitors of
DNA damage [164]. the DDR pathway may be of great potential for chemo- and radio-
sensitization of various cancers. Similarly, tumor resistant cells usually
10.3. Cell cycle have cell cycle alterations and frequently show a decreased suscepti­
bility to drug-induced apoptosis as a consequence of an up-regulation of
Cell cycle regulators frequently serve as oncogenes or tumor sup­ anti-apoptotic proteins and a down-regulation of pro-apoptotic proteins
pressors, with the cell cycle inhibitor p27 (Kip1) being the best- [167].
described example. p27(Kip1) is a tumor suppressor, as shown in Several reports have shown the involvement of various miRNAs i.e.
some tumors by its low levels. Additionally, when exposed to carcino­ miR-181b, miR-451, miR-497 and miR-200bc/429 cluster in sensitizing
gens, the mutation p27(Kip1) predisposes cells to tumorigenesis. p27 the cells to platinum induced cell death by down-regulating the
(Kip1) binds to Cdk2–cyclin E and prevents a transition from G1 to S expression of anti- apoptotic BCL-2 gene whose overexpression results in
phase [89]. In glioblastomas and prostate cancer cells, p27(Kip1) is a drug resistance [167,168]. It has been documented that miR-133a
direct target of miR-221 and -222 [165]. In these cancer cells, p27(Kip1) sensitized colorectal cancer cells to chemotherapeutic drug treatment
is anti-correlated with miR-221 and -222. Targeting p27(Kip1) is vital with doxorubicin or oxaliplatin by targeting the RFFL gene which

14
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Fig. 4. (A) and (B). A schematic model which shows


the molecular mechanisms of cancer pathogenesis
involving micro RNA. Arrow in red depicts inhibition.
Abbreviations employed in the figure,BCL2:B cell
lymphoma 2, an antiapoptotic gene; BIC: an evolu­
tionarily conserved non-coding RNA; CDK: cyclin-
dependent kinase; c-Myc; oncogenes; E2F1: a cell
cycle transcription factor; LAST2: Large Tumor Sup­
pressor homolog 2, a serine-threonine kinase; MAPK:
mitogen-activated protein kinase; p53: a critical
tumor suppressor that is involved in most, if not all,
tumorigenesis; PTEN: phosphatase and tensin ho­
mology, a tumor suppressor gene; RAS: a common
proto-oncogene.

regulates the p53/p21 signaling pathway [167]. miR-224 targets based sensitization studies for enhancing cellular sensitivity to cancer
p21Waf1/CIP1, responsible for cell cycle control thus blocking the treatments.
transition from phase G1 to phase S. Furthermore, down-regulation of
miR-27a could sensitize gastric cancer cells to drugs by decreasing cyclin
11.1. Tamoxifen
D1 expression and up-regulating p21expression [167]. Cells over­
expressing miR-383, which targets the DDR gene GADD45G, exhibited a
It has been reported that miRNAs may sensitize cancer cells to
more severe cell death than control cells exposed to cisplatin treatment
Tamoxifen, the most extensively employed non-steroidal “selective es­
[167]. Therefore, combining normal chemotherapy and radiotherapy
trogen receptor modulator (SERM)”, owing to the fact miR-21, miR-
using the RNA interference (RNAi) technique to specifically knock-down
146a, miR-148a, miR-34a and miR-27a serve important roles in medi­
the expression of the resistance genes, may sensitize the resistant cells to
ating TAM resistance in breast cancer [170]. Since Tamoxifen compete
the anti-cancer treatment efficiently [167]. Interestingly, by targeting
with the binding of estrogen to ERα on mammary glands, ultimately
miRNAs that targets the genes involved in multidrug resistance, resistant
perturbing the ERα signalling pathway, ERα truncations or the loss of
tumor cells could be sensitize. Enhanced drug excretion by ATP-binding
their expression facilitate resistance. This is particularly complicated in
cassette (ABC) transporters usually occurs in multidrug resistance. Two
people whose tumors over-express the ERBB2 receptor tyrosine kinase,
of such transporters i.e. ABCC3 and ABCC6 are directly induced by SOX2
promoting growth and differentiation. An ERBB2-induced survival
(a transcription factor involved in stemness, tumor-initiation and drug-
mechanism comprises repression of tumor-suppressive miRNAs miR-15
resistant properties) [169]. Enhanced expression of SOX2 is linked with
and miR-16 to restore antiapoptotic BCL-2 expression [166]. Conse­
return of the drugs back into the extracellular environment. miR-9 has
quently, the restoration of miR-15 and miR-16 diminishes BCL-2 levels
been documented by Kim and his colleagues [169], as a SOX2’s negative
and therefore sensitizes cells to tamoxifen. Oncomir miR-21 has also
regulator. Artificial expression of miR-9 in a chemotherapy-resistant
been reported to be triggered by ERBB2, which implies that antago­
stem cell line of glioma repressed expression of SOX2, which leaded to
nizing miR-21 may additionally induce cells to tamoxifen. Analogous
decreased expression of ABC transporters and eventually drug retention
results were reported for miR-342, which was down-regulated in
[169]. Further, SOX2 is also able to induce pluripotent stem cells thus
tamoxifen-resistant cells and thus resistant cells may be sensitized to
miR-9 replacement therapy, therefore, can reduce stemness and drug
tamoxifen-induced apoptosis by its over-expression [171]. Eventually,
efflux [169].
the overexpression of the miR-221~222 cluster was correlated with
In the section below, we have represented the examples of some
tamoxifen resistance by the downregulation of the p27KIP1 cell cycle
established drugs with diverse modes of action in reference to miRNA-
inhibitor; the ectopic expression p27KIP1 restored tamoxifen mediated

15
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

cell death [172]. If affirmed in vivo these miRNAs may be used to boost various transcription factors, disorganized cellular drug uptake and
the sensitivity to tamoxifen [166]. accumulation, reestablishment of DNA repair mechanisms, activation of
TS, etc. Since miRNAs simultaneously affect a large number of genes and
11.2. Gefitinib signaling pathways, their use in therapy may lead to improve outcomes
rather directing only one specific gene or protein [181]. let-7, miR-20b,
The level of miRNAs that targets cell survival signaling pathways in -135b, -182, -200c, -204, -224, and -587 miRNAs has been reported to
therapy-resistant cells are frequently reduced therefore their re- impair 5-FU sensitivity by regulating proteins related to the PI3K/AKT
expression may provide a finer possibility of curing the tumor. Gefiti­ signalling pathway. Whereas, miR-125b, -149, -320, -181a-5p etc.
nib an Epidermal growth factor receptor-tyrosine kinase inhibitors change cancer cell resistance to 5-FU by controlling the Wnt/β-catenin
selectively binds to the EGFR-tyrosine kinase domain, preventing ATP signalling pathway [181] Recently it has been suggested that restoration
from binding and blocking subsequent receptor autophosphorylation, of miR-375-3p levels could be a future novel therapeutic strategy to
which results in the inhibition of signal transduction. Gefitinib, have enhance chemosensitivity to 5-FU [182].
been found to be clinically effective in the treatment of patients with
non-small cell lung cancer (NSCLC) and for the primary treatment for 11.5. Taxanes
EGFR-positive cancers of lung and breast cancers that acts via sup­
pressing the downstream signaling by RAS and PI3 K eventually pre­ Taxanes acts by blocking cell cycle progression through centrosomal
venting uncontrolled growth and proliferation [174]. However, impairment, induction of abnormal spindles and suppression of spindle
development of gefitinib resistance leads to limit its therapeutic effect by microtubule dynamics. Multiple cancers are treated using taxanes,
deregulation of various miRNAs like miR-506-3p, miR-216, miR-762 however resistance can develop eventually. Expression of various genes
and miR-450a-5p [173–176]. Therefore, it is conferred that targeting conferring drug resistance can be altered by treatment incorporating
of such miRNAs may be therapeutic potential against gefitinib resistance taxanes. miRNAs have since established to regulate many of such genes,
by sensitizing the cells to conventional chemotherapeutics. Moreover, for instance mitogen- and stress-activated protein kinase 1 (governed by
recently it has been documented that exosomal miR-564 and miR-658 miR-148a), CASP8 and FADD like apoptosis regulator (inhibited by miR-
derived from gefitinib-resistant lung cancer cells induce drug resis­ 512-3p), BCL-2 antagonist killer 1 (suppressed by miR-125b), SIRT1and
tance in sensitive cells. Therefore, cell-to-cell interaction via exosomal BCL-2 (inhibited by miR-34) and inositol monophosphatase 1 (inhibited
microRNAs might be a novel mechanism and therapeutic target of by let-7 family members) [166]. Forced expression of such tumor-
resistance against gefitinib [177]. suppressive miRNAs in all these cases sensitized cells to taxanes.
Adenomatous polyposis coligene (APC) is a multi-functional tumour
11.3. Oxaliplatin suppressor also involved in regulating cell adhesion and migration,
microtubule networks, spindle formation and chromosome segregation.
Oxaliplatin (OX), a platinum based anticancer medication and an miR-135a was found to be associated with reduced APC in paclitaxel-
alkylating agent, can form DNA adducts to produce inter- or intra-chain resistant NSCLC cell lines and in vivo models, probably through inter­
crosslinks thereby preventing the separation of DNA strands during fering with the mitotic spindle checkpoint [183]. Interestingly, muta­
transcription and translation eventually inducing cancer cell apoptosis. tions in β -tubulin accounts for disruption of microtubule assembly, that
However, its frequent application has been reported to be linked with confers taxanes resistance. Moreover, β III-tubulin is one of the direct
eventual resistance. The miR -107, -744, -23b, -46146, -19b -3p, and -21 targets of miR-200c and the other members of miR-200 family (miR-
are among the miRNAs that drive OX resistance in cancer cells and make 141, miR-200a, miR-200b, and miR-429). Restoration of miR-200c has
them unresponsive to OX -mediated inhibitory effects on proliferation been documented to enhance the sensitivity to paclitaxel in endometrial,
and migration [178]. Moreover, miR -27b -3p, -22 -3p, -200b -3p, - 122, breast and ovarian cancer cell lines [184]. Moreover, dysregulation of
-160a, -133b, -130a, -567, -128 -3p, -483 -3p, -29b, -134, -143, -181a Microtubule-associated protein tau (MAPT) involved in tubulin assem­
and -506 are among the miRNAs that enhance the number of cancer cells bly and microtubule stabilization by miR-34c-5p was reported to be
undergoing apoptosis during OX chemotherapy [178]. Therefore, tar­ critical in the chemosensitivity of gastric cancer to paclitaxel [185].
geting of these may be of crucial importance in dealing OX resistance. furthermore, the regulation of cytokine networks by miRNAs and its role
Interestingly, in a recent study a panel of plasma exosomal miRNAs in chemo-resistance was explored and miR-335-5p and let-7c-5p has
have been identified containing miR-100, miR-92a, miR-16, miR-30e, been found as potent regulatory factors responsible for taxanes-
miR-144-5p, and let-7i, that could remarkably distinguish chemo­ resistance in breast cancer cells that downstream target chemokines
resistant patients from chemosensitive colorectal cancer (CRC) patients CXCL9, CCR7, and SOCS1 genes [186]. Recently, integrative miRNA and
[179]. The detection of circulating exosomal miRNAs thus may serve as gene expression analysis provide evidence for new epigenetically regu­
an possible way to monitor CRC patient responses to chemotherapy and lated miRNAs-induced modulation of signalling pathways in paclitaxel
targeting of these may also be a promising strategy for CRC treatment. resistant ovarian cancer cells providing new therapeutic strategies to
Moreover, in a study it has been reported that inhibition of checkpoint combat chemoresistance [187].
kinase 1 (Chk1) by miR-320c increases oxaliplatin responsiveness in miRNAs involved in therapeutic uses have been summarized in
triple-negative breast cancer thereby increasing the oxaliplatin’s effec­ Table 6.
tiveness. Thus miR-320c may serve as a prognostic marker of oxaliplatin
by regulating DNA damage response through the expression of Chk1 12. miRNA as a biomarker in body fluids
[180].
Although, miRNA profiling of tumors could be a brilliant prognostic
11.4. 5-Fluorouracil test, sampling requires invasive techniques for instance biopsy, aspira­
tion biopsy or excision surgery of prevailing tumors [110]. An expeller
The fluoropyrimidine 5-fluorouracil (5-FU) is an antimetabolite drug biomarker needs to be attainable utilizing non-invasive techniques,
that is widely used for the treatment of cancer, particularly for colorectal reasonably sensitive to distinguish early tumor presence prior to clinical
cancer. 5-FU exerts its anticancer effects through inhibition of thymi­ symptoms and non-existent or low in healthy, tumor-free people [166].
dylate synthase (TS) and incorporation of its metabolites into RNA and In body fluids such as serum, urine, saliva, tears, breast milk, seminal
DNA. fluid, and fecal matter, stable, degradation resistant, extracellular
The effects of miRNAs on 5-FU resistance is complex phenomenon miRNAs could be detected. These miRNAs relate to different patho­
that involves atypical activation of cancer signaling, augmentation of physiological conditions [194]. Though studies imply that circulating

16
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

Table 6
In vivo therapeutic implementation of miRNAs and antagomirs.
miRNA Method of delivery Model used Phenotypes References

let-7 Viral particles delivered intranasally KrasG12D/+ autochthonous NSCLC Suppression of initiation of lung tumor when delivered [166]
mouse simultaneously with transgene activation
Intertumoral, lipid-based mimetic injection Subcutaneous H460 NSCLC xenografts Interfered with the further growth of tumors [166]
Viral particles delivered intranasally KrasG12D/+ autochthonous NSCLC Reduced tumor burden permitted to preform 10 weeks [166]
mouse prior to let-7 therapy
Lipid-based mimetic delivered systemically KrasG12D/+ autochthonous NSCLC Decreased tumor burden permitted to preform 10 [166]
mouse weeks prior to let-7 therapy
Prior to transplantation transfected into cells Subcutaneous human U251 or U87 Reduced formation of tumors [166]
glioblastoma cells
miR-143 and Prior to transplantation transduced into cells. Subcutaneous MiaPaCa2 and Panc1 Inefficiency in forming a tumor [166]
miR-145 PDAC xenografts
Lipid-based expression vectors delivered Subcutaneous MiaPaCa2 and Panc1 Growth inhibition [166]
systemically PDAC xenografts
Lipid-based expression vectors delivered Orthotopic MiaPaCa2 PDAC Growth inhibition [166]
systemically xenografts
miR-143 Anti-miR-143 delivered systemically p21-HBx HCC mouse Primary tumor along with local and distant metastatic [166]
growth inhibited
miR-34 Lipid-based mimetic delivered intratumorally Subcutaneous H460 NSCLC xenografts Growth inhibition [166]
Lipid-based expression vectors delivered Subcutaneous H460 and A549 NSCLC Growth inhibition [166]
systemically xenografts
Oligonucleotides transfected into cells before Subcutaneous prostate cancer Tumor incidence reduced [166]
transplantation xenografts (multiple cell lines)
Prior to transplantation, cells transduced with Subcutaneous prostate cancer Tumor incidence reduced [166]
virus encoding precursor (pre)-miR-34 xenografts (multiple cell lines)
Lipid-based mimetic injected intratumorally Subcutaneous PPC1 prostate cancer Growth inhibition [166]
xenografts
Lipid-based mimetic delivered systemically Orthotopic PC3 prostate cancer Tumor burden reduced [ [166]
xenografts
Lipid-based mimetic delivered systemically Orthotopic LAPC9 prostate cancer Reduced lung metastasis without affecting the growth [166]
xenografts of primary tumors; increased survival
Lipid-based expression vectors delivered Subcutaneous and orthotopic Growth inhibition [166]
systemically MiaPaCa2 PDAC xenografts
miR-122 Before transplantation, transduced into cells Orthotopic SKHEP1 HCC xenografts Tumorigenesis, angiogenesis and intrahepatic [166]
metastasis reduced
Nucleic acid antagomir-miR-122 delivered HCV-infected non-human primates HCV viremia suppressed and liver pathology improved [ [166]
systemically
Lysine–lipid nanoparticle antagomir-miR-12 C57BL/6J mice Plasma cholesterol levels reduced [166]
delivered systemically
Antagomir-miR-122 delivered systemically C57BL/6J mice Plasma cholesterol levels reduced [166]
miR-26a Adeno-associated virus delivered systemically HCC liver cancer model: MYC is driven Proliferation inhibition; apoptosis induction; disease [166]
by the liver activator promoter protection
miR-379 Delivered intravenously MSC-EVs Substantial reduction in tumor volume in breast cancer [188]
miR-133b Delivered intravenously Cationic lipoplexes A notable increase in premiR-133b expression at lungs [189]
in lung cancer
miR-660 Delivered intraperitoneally and intravenously CCL nanoparticles Tumor growth decreased significantly in lung cancer [190]
miR-634 Delivered intravenously Lipid-based nanoparticles Tumor growth decreased significantly in pancreatic [191]
cancer
miR-185 Delivered topically MSC-EVs Incidence of transformation to OSCC significantly [189]
reduced in OPMD
miR-100 and Delivered intranasally Gold-iron oxide nanoparticles NPs progressively accumulates in the prefrontal cortex [192]
anti-miR-21 and thus longer survival in patients with glioblastoma.
miR-16 Delivered intravenously Bacterial minicells Tumor growth inhibition in lung cancer [189]
miR-655-3p Delivered intraperitonially Nanoscale coordination polymers Liver tumor development suppressed in patients with [193]
and OXL colorectal liver metastases

HCC, hepatocellular carcinoma; HCV, hepatitis C virus; miRNA, microRNA; NSCLC, non-small-cell lung cancer; PDAC, pancreatic ductal adenocarcinoma; CLL, coated
cationic lipid; MSC-EVs, mesenchymal stem cell-derived extracellular vesicles; OSCC, oral squamous cell cancer; OMPD, oral potentially malignant disorders; NPs,
nanoparticles; OXL, oxaliplatin.

miRNAs could conceive from a non-specific discharge by lysed or nec­ complex mechanisms behind its biogenesis. Micro RNA discovery
rotized cells [195], others have demonstrated that miRNAs are actively brought another aspect to the investigation of the regulation of gene
transported outside of cells packed as exosomes [196] or as free miRNAs expression. Dysregulation of its biogenesis eventually alters the mRNA
attached to Argonaut proteins [197]. Intracellular and secreted miRNA profile in a cell, which in turn, via a feedback loop, influences miRNA
populations vary, implying that an extracellular miRNA secretion in­ expression and function. Thus, the gene expression regulatory networks
volves a selective process. Their stability, low complexity, and accessi­ that cover both miRNA expression and its effect on mRNA targets, need
bility of inexpensive detection techniques and profiling could make to be examined extensively. While discrepancies remain in our knowl­
extracellular miRNAs exemplar for several diseases [166]. edge of how this post-transcriptional gene expression pathway functions
and the manner it is controlled in various types of cells, an increasing
13. Conspectus number of pieces of evidence ties deregulated miRNA expression to
many cancers’ etiology. Upcoming research thus strives to highlight the
Over the past 15 years, researchers have significantly highlighted an most critical miRNAs in light of distinct classes of cancer and is likely to
essential group of regulators among all multicellular species and the unveil additional mechanisms that govern the expression of specific

17
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

miRNAs or miRNA sub-sets. miRNAs play a significant role in virtually [8] J.M. Thomson, J. Parker, C.M. Perou, S.M. Hammond, A custom microarray
platform for analysis of microRNA gene expression, Nat. Methods 1 (2004)
all cancer biology facets, including proliferation, apoptosis, invasion/
47–53, https://doi.org/10.1038/nmeth704.
metastasis, and angiogenesis. From the illustrations put forward in this [9] B. Zhang, X. Pan, G.P. Cobb, T.A. Anderson, microRNAs as oncogenes and tumor
review, it becomes apparent that several of these alterations in the suppressors, Dev. Biol. 302 (1) (2007) 1–12, https://doi.org/10.1016/j.
expression are not merely a secondary outcome of the transformation ydbio.2006.08.028.
[10] J. Lu, G. Getz, E.A. Miska, E. Alvarez-Saavedra, J. Lamb, D. Peck, J.R. Downing,
process. Instead, the loss- or gain-of-function of particular miRNAs MicroRNA expression profiles classify human cancers, Nature 435 (7043) (2005)
seems to be a crucial incident in the emergence of various cancers. Such 834–838, https://doi.org/10.1038/nature03702.
studies reflect the requirement to incorporate the analysis of miRNA [11] T.D. Schmittgen, J. Jiang, Q. Liu, L. Yang, A high-throughput method to monitor
the expression of microRNA precursors, Nucleic Acids Res. 32 (4) (2004) e43,
expression and function in ongoing molecular analysis of cancer path­ https://doi.org/10.1093/nar/gnh040.
ogenesis to attain a full comprehension of this group of malignancies. [12] S.D. Fiedler, M.Z. Carletti, L.K. Christenson, Quantitative RT-PCR methods for
Since miRNAs have several targets, their role in tumorigenesis may be mature microRNA expression analysis, Methods Mol. Biol. 630 (2010) 49–64.
[13] Q. He, Y. Fang, F. Lu, J. Pan, L. Wang, W. Gong, F. Fei, J. Cui, J. Zhong, R. Hu,
due to a few different targets being controlled, perhaps only one or M. Liang, L. Fang, H. Wang, M. Yu, Z. Zhang, Analysis of differential expression
more. A potential challenge will be discovering all the targets of the profile of miRNA in peripheral blood of patients with lung cancer, J. Clin. Lab.
miRNAs concerned with cancer and evaluating their share to malignant Anal. 33 (2019), e23003, https://doi.org/10.1002/jcla.23003.
[14] L. Gan, B. Denecke, Profiling pre-microRNA and mature microRNA expressions
transformation. An added goal would be to recognize all the miRNAs using a single microarray and avoiding separate sample preparation, Microarrays
disrupted by pathways that are frequently dysregulated in district forms (Basel, Switzerland) 2 (1) (2013) 24–33.
of human cancers. Hopefully, miRNA-based procedures will expedi­ [15] D. Waggott, K. Chu, S. Yin, B.G. Wouters, F.F. Liu, P.C. Boutros, Nano string norm:
an extensible R package for the pre-processing of NanoString mRNA and miRNA
tiously enter the clinic as an essential diagnostic and promising thera­
data, Bioinformatics 28 (11) (2012) 1546–1548, https://doi.org/10.1093/
peutic tools. However, the analysis of microRNA-based interventions is bioinformatics/bts188.
still in its infancy, and the adverse effects of these therapies must be [16] H.F. Tsang, V.W. Xue, S.P. Koh, Y.M. Chiu, L.P.W. Ng, S.C.C. Wong, NanoString, a
investigated. novel digital color-coded barcode technology: current and future applications in
molecular diagnostics, Expert. Rev. Mol. Diagn. 17 (1) (2017) 95–103, https://
doi.org/10.1080/14737159.2017.1268533.
Funding [17] J.M. Eastel, K.W. Lam, N.L. Lee, W.Y. Lok, A.H.F. Tsang, X.M. Pei, A.K.C. Chang,
W.C.S. Cho, S.C.C. Wong, Application of NanoString technologies in companion
diagnostic development, Expert. Rev. Mol. Diagn. 19 (7) (2019) 591–598,
This research received no external funding. https://doi.org/10.1080/14737159.2019.1623672.
[18] V.A. Valera, R. Parra-Medina, B.A. Walter, P. Pinto, M.J. Merino, microRNA
Author contributions expression profiling in young prostate cancer patients, J. Cancer 11 (14) (2020)
4106, https://doi.org/10.7150/jca.37842.
[19] P.P. Reis, S.A. Drigo, R.F. Carvalho, R.M. Lopez Lapa, T.F. Felix, D. Patel,
Supervision: Manisha Nigam and Abhay Prakash Mishra; Project D. Chang, M. Pintilie, G. Lui, M.S. Tsao, Circulating miR-16-5p, miR-92a-3p, and
administration: Manisha Nigam and Henrique Douglas Melo Coutinho; miR-451a in plasma from lung cancer patients: potential application in early
detection and a regulatory role in tumorigenesis pathways, Cancers 12 (8) (2020)
Methodology: Abhay Shikhar Panwar and Mridul Budakoti; Final draft 2071, https://doi.org/10.3390/cancers12082071.
of the manuscript: Manisha Nigam, Mridul Budakoti and Abhay Prakash [20] A.M. Coenen-Stass, I. Magen, T. Brooks, I.Z. Ben-Dov, L. Greensmith,
Mishra; Resources: Diksha Molpa, Rahul Kunwar Singh and Dietrich E. Hornstein, P. Fratta, Evaluation of methodologies for microRNA biomarker
detection by next generation sequencing, RNA Biol. 15 (8) (2018) 1133–1145, e,
Büsselberg.
https://doi.org/10.1080/15476286.2018.1514236.
[21] H. Bisgin, B. Gong, Y. Wang, W. Tong, Evaluation of bioinformatics approaches
Declaration of Competing Interest for next-generation sequencing analysis of microRNAs with a toxicogenomics
study design, Front. Genet. 9 (2018) 22, https://doi.org/10.3389/
fgene.2018.00022.
The authors declare no conflict of interest. [22] S. Motameny, S. Wolters, P. Nürnberg, B. Schumacher, Next generation
sequencing of miRNAs–strategies, resources and methods, Genes 1 (1) (2010)
Acknowledgments 70–84, https://doi.org/10.3390/genes1010070.
[23] V.G. LeBlanc, M.A. Marra, Next-generation sequencing approaches in cancer:
where have they brought us and where will they take us? Cancers 7 (3) (2015)
NM acknowledges the Portuguese Foundation for Science and 1925–1958, https://doi.org/10.3390/cancers7030869.
Technology under the Horizon 2020 Program (PTDC/PSI-GER/28076/ [24] G. Matullo, A. Naccarati, B. Pardini, Micro RNA expression profiling in bladder
cancer: the challenge of next-generation sequencing in tissues and biofluids, Int.
2017). J. Cancer 138 (10) (2016) 2334–2345, https://doi.org/10.1002/ijc.29895.
[25] B.J. Reinhart, F.J. Slack, M. Basson, et al., The 21-nucleotide let-7 RNA regulates
References developmental timing in Caenorhabditis elegans, Nature 403 (2000) 901–990.
[26] A.E. Pasquinelli, B.J. Reinhart, F. Slack, et al., Conservation of the sequence and
temporal expression of let-7 heterochronic regulatory RNA, Nature 408 (2000)
[1] R.C. Lee, R.L. Feinbaum, V. Ambros, The C. elegans heterochronic gene lin-4
86–89.
encodes small RNAs with antisense complementarity to lin-14, Cell 75 (5) (1993)
[27] G. Michlewski, S. Guil, C.A. Semple, J.F. Cáceres, Posttranscriptional regulation
843–854, https://doi.org/10.1016/0092-8674(93)90529-Y.
of miRNAs harboring conserved terminal loops, Mol. Cell 32 (3) (2008) 383–393,
[2] B.J. Reinhart, F.J. Slack, M. Basson, A.E. Pasquinelli, J.C. Bettinger, A.E. Rougvie,
https://doi.org/10.1016/j.molcel.2008.10.013.
G. Ruvkun, The 21-nucleotide let-7 RNA regulates developmental timing in
[28] S.R. Viswanathan, G.Q. Daley, Lin28: A microRNA regulator with a macro role,
Caenorhabditis elegans, Nature 403 (6772) (2000) 901–906, https://doi.org/
Cell 140 (4) (2010) 445–449, https://doi.org/10.1016/j.cell.2010.02.007.
10.1038/35002607.
[29] L. Han, P.D.W. Witmer, E. Casey, D. Valle, S. Sukumar, DNA methylation
[3] Y. Peng, C.M. Croce, The role of MicroRNAs in human cancer, Sig. Transduc.
regulates MicroRNA expression, Cancer Biol. & Ther. 6 (8) (2007) 1290–1294,
Targeted Ther. 1 (1) (2016) 1–9, https://doi.org/10.1038/sigtrans.2015.4.
https://doi.org/10.4161/cbt.6.8.4486.
[4] F. Wahid, A. Shehzad, T. Khan, Y.Y. Kim, MicroRNAs: synthesis, mechanism,
[30] G.A. Calin, C. Sevignani, C.D. Dumitru, T. Hyslop, E. Noch, S. Yendamuri, C.
function, and recent clinical trials, Biochimica et Biophysica Acta (BBA)-
M. Croce, Human microRNA genes are frequently located at fragile sites and
Molecular Cell Research 1803 (11) (2010) 1231–1243, https://doi.org/10.1016/
genomic regions involved in cancers, Proc. Natl. Acad. Sci. 101 (9) (2004)
j.bbamcr.2010.06.013.
2999–3004, https://doi.org/10.1073/pnas.0307323101.
[5] L.A. MacFarlane, R. Murphy, P., MicroRNA: biogenesis, function and role in
[31] L. Zhang, J. Huang, N. Yang, J. Greshock, M.S. Megraw, A. Giannakakis, G. Yao,
cancer, Current genomics 11 (7) (2010) 537–561, https://doi.org/10.2174/
microRNAs exhibit high frequency genomic alterations in human cancer, Proc.
138920210793175895.
Natl. Acad. Sci. 103 (24) (2006) 9136–9141, https://doi.org/10.1073/
[6] Y. Hayashita, H. Osada, Y. Tatematsu, H. Yamada, K. Yanagisawa, S. Tomida,
pnas.0508889103.
Y. Yatabe, K. Kawahara, Y. Sekido, T. Takahashi, A polycistronic microRNA
[32] A. Cimmino, G.A. Calin, M. Fabbri, M.V. Iorio, M. Ferracin, M. Shimizu,
cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell
L. Rassenti, miR-15 and miR-16 induce apoptosis by targeting BCL2, Proc. Natl.
proliferation, Cancer Res. 65 (21) (2005) 9628–9632, https://doi.org/10.1158/
Acad. Sci. 102 (39) (2005) 13944–13949, https://doi.org/10.1073/
0008-5472.CAN-05-2352.
pnas.0506654102.
[7] N.B. Aziz, R.G. Mahmudunnabi, M. Umer, S. Sharma, M.A. Rashid,
[33] G.A. Calin, C.D. Dumitru, M. Shimizu, R. Bichi, S. Zupo, E. Noch, L. Rassenti,
Y. Alhamhoom, Y.B. Shim, C. Salomon, M.J. Shiddiky, MicroRNAs in ovarian
Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at
cancer and recent advances in the development of microRNA-based biosensors,
Analyst 145 (6) (2020) 2038–2057, https://doi.org/10.1039/C9AN02263E.

18
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

13q14 in chronic lymphocytic leukemia, Proc. Natl. Acad. Sci. 99 (24) (2002) [58] J.W. Catto, S. Miah, H.C. Owen, H. Bryant, K. Myers, E. Dudziec, S. Larré,
15524–15529, https://doi.org/10.1073/pnas.242606799. M. Milo, I. Rehman, D.J. Rosario, E. Di Martino, Distinct microRNA alterations
[34] A. Kotani, D. Ha, D. Schotte, S.A. Armstrong, H.F. Lodish, A novel mutation in the characterize high-and low-grade bladder cancer, Cancer Res. 69 (21) (2009)
miR-128b gene reduces miRNA processing and leads to glucocorticoid resistance 8472–8481, https://doi.org/10.1158/0008-5472.CAN-09-0744.
of MLL-AF4 acute lymphocytic leukemia cells, Cell Cycle 9 (6) (2010) [59] M. Yan, H.Y. Huang, T. Wang, Y. Wan, S.D. Cui, Z.Z. Liu, Q.X. Fan, Dysregulated
1037–1042, https://doi.org/10.4161/cc.9.6.11011. expression of dicer and drosha in breast cancer, Pathol. Oncol. Res. 18 (2) (2012)
[35] K.A. O’Donnell, E.A. Wentzel, K.I. Zeller, C.V. Dang, J.T. Mendell, c-Myc- 343–348, https://doi.org/10.1007/s12253-011-9450-3.
regulated microRNAs modulate E2F1 expression, Nature 435 (7043) (2005) [60] M. Sand, M. Skrygan, D. Georgas, C. Arenz, T. Gambichler, D. Sand, P. Altmeyer,
839–843, https://doi.org/10.1038/nature03677. F.G. Bechara, Expression levels of the microRNA maturing microprocessor
[36] M. Dews, A. Homayouni, D. Yu, D. Murphy, C. Sevignani, E. Wentzel, A. Thomas- complex component DGCR8 and the RNA-induced silencing complex (RISC)
Tikhonenko, Augmentation of tumor angiogenesis by a Myc-activated microRNA components argonaute-1, argonaute-2, PACT, TARBP1, and TARBP2 in epithelial
cluster, Nat. Genet. 38 (9) (2006) 1060, https://doi.org/10.1038/ng1855. skin cancer, Mol. Carcinog. 51 (11) (2012) 916–922, https://doi.org/10.1002/
[37] C.P. Bracken, P.A. Gregory, N. Kolesnikoff, A.G. Bert, J. Wang, M.F. Shannon, G. mc.20861.
J. Goodall, A double-negative feedback loop between ZEB1-SIP1 and the [61] S. Ambs, R.L. Prueitt, M. Yi, R.S. Hudson, T.M. Howe, F. Petrocca, T.A. Wallace,
microRNA-200 family regulates epithelial-mesenchymal transition, Cancer Res. C.G. Liu, S. Volinia, G.A. Calin, H.G. Yfantis, Genomic profiling of microRNA and
68 (19) (2008) 7846–7854, https://doi.org/10.1158/0008-5472.CAN-08-1942. messenger RNA reveals deregulated microRNA expression in prostate cancer,
[38] Y. Lee, K. Jeon, J.T. Lee, S. Kim, V.N. Kim, MicroRNA maturation: stepwise Cancer Res. 68 (15) (2008) 6162–6170, https://doi.org/10.1158/0008-5472.
processing and subcellular localization, EMBO J. 21 (17) (2002) 4663–4670, CAN-08-0144.
https://doi.org/10.1093/emboj/cdf476. [62] B. Kim, J.H. Lee, J.W. Park, T.K. Kwon, S.K. Baek, I. Hwang, S. Kim, An essential
[39] Y. Zeng, B.R. Cullen, Sequence requirements for micro RNA processing and microRNA maturing microprocessor complex component DGCR8 is up-regulated
function in human cells, RNA 9 (1) (2003) 112–123, https://doi.org/10.1261/ in colorectal carcinomas, Clin. Exp. Med. 14 (3) (2014) 331–336, https://doi.org/
rna.2780503. 10.1007/s10238-013-0243-8.
[40] Y. Lee, C. Ahn, J. Han, H. Choi, J. Kim, J. Yim, V.N. Kim, The nuclear RNase III [63] Y. Guo, P. Tian, C. Yang, Z. Liang, M. Li, M. Sims, L. Lu, Z. Zhang, H. Li, L.
Drosha initiates microRNA processing, Nature 425 (6956) (2003) 415–419, M. Pfeffer, J. Yue, Silencing the double-stranded RNA binding protein DGCR8
https://doi.org/10.1038/nature01957. inhibits ovarian cancer cell proliferation, migration, and invasion, Pharm. Res. 32
[41] J. Han, Y. Lee, K.H. Yeom, Y.K. Kim, H. Jin, V.N. Kim, The Drosha-DGCR8 (3) (2015) 769–778, https://doi.org/10.1007/s11095-013-1219-9.
complex in primary microRNA processing, Genes Dev. 18 (24) (2004) [64] A. Torres, K. Torres, T. Paszkowski, B. Jodłowska-Jędrych, T. Radomański,
3016–3027, https://doi.org/10.1101/gad.1262504. A. Książek, R. Maciejewski, Major regulators of microRNAs biogenesis Dicer and
[42] A.M. Denli, B.B. Tops, R.H. Plasterk, R.F. Ketting, G.J. Hannon, Processing of Drosha are down-regulated in endometrial cancer, Tumor Biol. 32 (4) (2011)
primary microRNAs by the microprocessor complex, Nature 432 (7014) (2004) 769–776, https://doi.org/10.1007/s13277-011-0179-0.
231–235, https://doi.org/10.1038/nature03049. [65] D.X. Zhu, L. Fan, R.N. Lu, C. Fang, W.Y. Shen, Z.J. Zou, Y.H. Wang, H.Y. Zhu, K.
[43] R.I. Gregory, K.P. Yan, G. Amuthan, T. Chendrimada, B. Doratotaj, N. Cooch, R. Miao, P. Liu, W. Xu, Downregulated Dicer expression predicts poor prognosis
R. Shiekhattar, The microprocessor complex mediates the genesis of microRNAs, in chronic lymphocytic leukemia, Cancer Sci. 103 (5) (2012) 875–881, https://
Nature 432 (7014) (2004) 235–240, https://doi.org/10.1038/nature03120. doi.org/10.1111/j.1349-7006.2012.02234.x.
[44] M. Landthaler, A. Yalcin, T. Tuschl, The human DiGeorge syndrome critical [66] X. Wang, X. Xu, Z. Ma, Y. Huo, Z. Xiao, Y. Li, Y. Wang, Dynamic mechanisms for
region gene 8 and Its D. melanogaster homolog are required for miRNA pre-miRNA binding and export by Exportin-5, Rna 17 (8) (2011) 1511–1528. http
biogenesis, Curr. Biol. 14 (23) (2004) 2162–2167, https://doi.org/10.1016/j. ://www.rnajournal.org/cgi/doi/10.1261/rna.2732611.
cub.2004.11.001. [67] K. Wu, J. He, W. Pu, Y. Peng, The role of exportin-5 in microRNA biogenesis and
[45] J. Han, Y. Lee, K.H. Yeom, J.W. Nam, I. Heo, J.K. Rhee, V.N. Kim, Molecular basis cancer, Genomics, Proteomics & Bioinform. 16 (2) (2018) 120–126, https://doi.
for the recognition of primary microRNAs by the Drosha-DGCR8 complex, Cell org/10.1016/j.gpb.2017.09.004.
125 (5) (2006) 887–901, https://doi.org/10.1016/j.cell.2006.03.043. [68] S.A. Melo, C. Moutinho, S. Ropero, G.A. Calin, S. Rossi, R. Spizzo, H. Yamamoto,
[46] Y. Zeng, B.R. Cullen, Efficient processing of primary microRNA hairpins by A genetic defect in exportin-5 traps precursor microRNAs in the nucleus of cancer
Drosha requires flanking nonstructured RNA sequences, J. Biol. Chem. 280 (30) cells, Cancer Cell 18 (4) (2010) 303–315, https://doi.org/10.1016/j.
(2005) 27595–27603, https://doi.org/10.1074/jbc.M504714200. ccr.2010.09.007.
[47] T. Du, P.D. Zamore, microPrimer: the biogenesis and function of microRNA, [69] D. Leaderer, A.E. Hoffman, T. Zheng, A. Fu, J. Weidhaas, T. Paranjape, Y. Zhu,
Development 132 (21) (2005) 4645–4652, https://doi.org/10.1242/dev.02070. Genetic and epigenetic association studies suggest a role of microRNA biogenesis
[48] B. Muralidhar, D. Winder, M. Murray, R. Palmer, N. Barbosa-Morais, H. Saini, gene exportin-5 (XPO5) in breast tumorigenesis, Int. J. Mol. Epidemiol.Genet. 2
N. Coleman, Functional evidence that Drosha overexpression in cervical (1) (2011) 9. PMID: 21552306.
squamous cell carcinoma affects cell phenotype and microRNA profiles, J. Pathol. [70] H.L. Sun, R. Cui, J. Zhou, K.Y. Teng, Y.H. Hsiao, K. Nakanishi, C.M. Croce, ERK
224 (4) (2011) 496–507, https://doi.org/10.1002/path.2898. activation globally downregulates miRNAs through phosphorylating exportin-5,
[49] B. Muralidhar, L.D. Goldstein, G. Ng, D.M. Winder, R.D. Palmer, E.L. Gooding, N. Cancer Cell 30 (5) (2016) 723–736.
L. Barbosa-Morais, G. Mukherjee, N.P. Thorne, I. Roberts, M.R. Pett, Global [71] Z. Ali Syeda, S.S.S. Langden, C. Munkhzul, M. Lee, S.J. Song, Regulatory
microRNA profiles in cervical squamous cell carcinoma depend on Drosha mechanism of microrna expression in cancer, Int. J. Mol. Sci. 21 (5) (2020) 1723,
expression levels, J. Pathol. A J. Pathol. Soc. Great Britain and Ireland 212 (4) https://doi.org/10.3390/ijms21051723.
(2007) 368–377, https://doi.org/10.1002/path.2179. [72] H. Zhang, F.A. Kolb, V. Brondani, E. Billy, W. Filipowicz, Human Dicer
[50] N. Sugito, H. Ishiguro, Y. Kuwabara, M. Kimura, A. Mitsui, H. Kurehara, T. Ando, preferentially cleaves dsRNAs at their termini without a requirement for ATP,
R. Mori, N. Takashima, R. Ogawa, Y. Fujii, RNASEN regulates cell proliferation EMBO J. 21 (21) (2002) 5875–5885, https://doi.org/10.1093/emboj/cdf582.
and affects survival in esophageal cancer patients, Clin. Cancer Res. 12 (24) [73] P. Provost, D. Dishart, J. Doucet, D. Frendewey, B. Samuelsson, O. Rådmark,
(2006) 7322–7328, https://doi.org/10.1158/1078-0432.CCR-06-0515. Ribonuclease activity and RNA binding of recombinant human Dicer, EMBO J. 21
[51] D.J. Papachristou, E. Sklirou, D. Corradi, C. Grassani, V. Kontogeorgakos, U. (21) (2002) 5864–5874, https://doi.org/10.1093/emboj/cdf578.
N. Rao, Immunohistochemical analysis of the endoribonucleases Drosha, Dicer [74] K. Saito, A. Ishizuka, H. Siomi, M.C. Siomi, Processing of pre-microRNAs by the
and Ago2 in smooth muscle tumours of soft tissues, Histopathology 60 (6B) Dicer-1–Loquacious complex in Drosophila cells, PLoS Biol. 3 (7) (2005) l30,
(2012) E28–E36, https://doi.org/10.1111/j.1365-2559.2012.04192.x. https://doi.org/10.1371/journal.pbio.0030235.
[52] R.J. Lin, Y.C. Lin, J. Chen, H.H. Kuo, Y.Y. Chen, M.B. Diccianni, L.Y. Alice, [75] T.P. Chendrimada, R.I. Gregory, E. Kumaraswamy, J. Norman, N. Cooch,
microRNA signature and expression of Dicer and Drosha can predict prognosis K. Nishikura, R. Shiekhattar, TRBP recruits the Dicer complex to Ago2 for
and delineate risk groups in neuroblastoma, Cancer Res. 70 (20) (2010) microRNA processing and gene silencing, Nature 436 (7051) (2005) 740–744,
7841–7850, https://doi.org/10.1158/0008-5472.CAN-10-0970. https://doi.org/10.1038/nature03868.
[53] G.S. Shu, L.P. Yang, Z.L. Yang, S. Jiang, Expression of CDC6 and GDF-9 and their [76] Y. Tomari, C. Matranga, B. Haley, N. Martinez, P.D. Zamore, A protein sensor for
clinicopathological significances in benign and malignant lesions of the siRNA asymmetry, Science 306 (5700) (2004) 1377–1380, https://doi.org/
gallbladder, Cancer Biomarkers 11 (2–3) (2012) 107–114, https://doi.org/ 10.1126/science.1102755.
10.3233/CBM-2012-0267. [77] S. Diederichs, D.A. Haber, Dual role for argonautes in microRNA processing and
[54] S. Lin, R.I. Gregory, MicroRNA biogenesis pathways in cancer, Nat. Rev. Cancer posttranscriptional regulation of microRNA expression, Cell 131 (6) (2007)
15 (6) (2015) 321–333, https://doi.org/10.1038/nrc3932. 1097–1108, https://doi.org/10.1016/j.cell.2007.10.032.
[55] S.E. Grund, M. Polycarpou-Schwarz, C. Luo, S.B. Eichmüller, S. Diederichs, Rare [78] G.B. Robb, T.M. Rana, RNA helicase A interacts with RISC in human cells and
Drosha splice variants are deficient in microRNA processing but do not affect functions in RISC loading, Mol. Cell 26 (4) (2007) 523–537, https://doi.org/
general microRNA expression in cancer cells, Neoplasia (New York, NY) 14 (3) 10.1016/j.molcel.2007.04.016.
(2012) 238, https://doi.org/10.1593/neo.111586. [79] C.G. Lee, J. Hurwitz, A new RNA helicase isolated from HeLa cells that
[56] O. Tchernitsa, A. Kasajima, R. Schäfer, R.J. Kuban, U. Ungethüm, B. Györffy, catalytically translocates in the 3’to 5’direction, J. Biol. Chem. 267 (7) (1992)
U. Neumann, E. Simon, W. Weichert, M.P. Ebert, C. Röcken, Systematic 4398–4407. 1537828.
evaluation of the miRNA-ome and its downstream effects on mRNA expression [80] J. Friedemann, F. Grosse, S. Zhang, Nuclear DNA helicase II (RNA helicase A)
identifies gastric cancer progression, J. Pathol. 222 (3) (2010) 310–319, https:// interacts with Werner syndrome helicase and stimulates its exonuclease activity,
doi.org/10.1002/path.2759. J. Biol. Chem. 280 (35) (2005) 31303–31313, https://doi.org/10.1074/jbc.
[57] O. Vaksman, T.E. Hetland, C.G. Trope, R. Reich, B. Davidson, Argonaute, Dicer, M503882200.
and Drosha are up-regulated along tumor progression in serous ovarian [81] M.S. Kumar, J. Lu, K.L. Mercer, T.R. Golub, T. Jacks, Impaired microRNA
carcinoma, Hum. Pathol. 43 (11) (2012) 2062–2069, https://doi.org/10.1016/j. processing enhances cellular transformation and tumorigenesis, Nat. Genet. 39
humpath.2012.02.016. (5) (2007) 673–677, https://doi.org/10.1038/ng2003.

19
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

[82] M.S. Kumar, R.E. Pester, C.Y. Chen, K. Lane, C. Chin, J. Lu, T. Jacks, Dicer1 controls developmental time and is regulated by microRNAs, Dev. Cell 4 (5)
functions as a haploinsufficient tumor suppressor, Genes Dev. 23 (23) (2009) (2003) 625–637, https://doi.org/10.1016/S1534-5807(03)00127-8.
2700–2704, https://doi.org/10.1101/gad.1848209. [107] M.D. Jansson, A.H. Lund, MicroRNA and cancer, Mol. Oncol. 6 (6) (2012)
[83] B. Kim, J.H. Lee, J.W. Park, T.K. Kwon, S.K. Baek, I. Hwang, S. Kim, An essential 590–610, https://doi.org/10.1016/j.molonc.2012.09.006.
microRNA maturing microprocessor complex component DGCR8 is up-regulated [108] M.L. Si, S. Zhu, H. Wu, Z. Lu, F. Wu, Y.Y. Mo, miR-21-mediated tumor growth,
in colorectal carcinomas, Clin. Exp. Med. 14 (3) (2014) 331–336, https://doi.org/ Oncogene 26 (19) (2007) 2799–2803, https://doi.org/10.1038/sj.onc.1210083.
10.1007/s10238-013-0243-8. [109] M.V. Iorio, M. Ferracin, C.G. Liu, A. Veronese, R. Spizzo, S. Sabbioni, S. Ménard,
[84] D.A. Hill, J. Ivanovich, J.R. Priest, C.A. Gurnett, L.P. Dehner, D. Desruisseau, MicroRNA gene expression deregulation in human breast cancer, Cancer Res. 65
G. Williams, DICER1 mutations in familial pleuropulmonary blastoma, Science (16) (2005) 7065–7070, https://doi.org/10.1158/0008-5472.CAN-05-1783.
325 (5943) (2009) 965, https://doi.org/10.1126/science.1174334. [110] Y.W. Kong, D. Ferland-McCollough, T.J. Jackson, M. Bushell, microRNAs in
[85] A. Heravi-Moussavi, M.S. Anglesio, S.W.G. Cheng, J. Senz, W. Yang, L. Prentice, cancer management, Llancet Oncol. 13 (6) (2012) e249–e258, https://doi.org/
N. Hamel, Recurrent somatic DICER1 mutations in nonepithelial ovarian cancers, 10.1016/S1470-2045(12)70073-6.
New England J. Med. 366 (3) (2012) 234–242, https://doi.org/10.1056/ [111] D.Y. Lee, Z. Deng, C.H. Wang, B.B. Yang, MicroRNA-378 promotes cell survival,
NEJMoa1102903. tumor growth, and angiogenesis by targeting SuFu and Fus-1 expression, Proc.
[86] M.S. Anglesio, Y. Wang, W. Yang, J. Senz, A. Wan, A. Heravi-Moussavi, G. Natl. Acad. Sci. 104 (51) (2007) 20350–20355, https://doi.org/10.1073/
B. Morin, Cancer-associated somatic DICER1 hotspot mutations cause defective pnas.0706901104.
miRNA processing and reverse-strand expression bias to predominantly mature [112] K.B. Reddy, MicroRNA (miRNA) in cancer, Cancer Cell Int. 15 (1) (2015) 38,
3p strands through loss of 5p strand cleavage, J. Pathol. 229 (3) (2013) 400–409, https://doi.org/10.1186/s12935-015-0185-1.
https://doi.org/10.1002/path.4135. [113] A. Lagana, F. Russo, C. Sismeiro, R. Giugno, A. Pulvirenti, A. Ferro, Variability in
[87] I. Slade, C. Bacchelli, H. Davies, A. Murray, F. Abbaszadeh, S. Hanks, the incidence of miRNAs and genes in fragile sites and the role of repeats and CpG
H. Jenkinson, DICER1 syndrome: clarifying the diagnosis, clinical features and islands in the distribution of genetic material, PLoS One 5 (6) (2010), https://doi.
management implications of a pleiotropic tumour predisposition syndrome, org/10.1371/journal.pone.0011166.
J. Med. Genet. 48 (4) (2011) 273–278, https://doi.org/10.1136/ [114] C. Mayr, M.T. Hemann, D.P. Bartel, Disrupting the pairing between let-7 and
jmg.2010.083790. Hmga2 enhances oncogenic transformation, Science 315 (5818) (2007)
[88] D.X. Zhu, L. Fan, R.N. Lu, C. Fang, W.Y. Shen, Z.J. Zou, Y.H. Wang, H.Y. Zhu, K. 1576–1579, https://doi.org/10.1126/science.1137999.
R. Miao, P. Liu, W. Xu, Downregulated Dicer expression predicts poor prognosis [115] P.J. Mishra, P.J. Mishra, D. Banerjee, J.R. Bertino, MiRSNPs or MiR-
in chronic lymphocytic leukemia, Cancer Sci. 103 (5) (2012) 875–881, https:// polymorphisms, new players in microRNA mediated regulation of the cell:
doi.org/10.1111/j.1349-7006.2012.02234.x. Introducing microRNA pharmacogenomics, Cell Cycle 7 (7) (2008) 853–858,
[89] Y.S. Lee, A. Dutta, MicroRNAs in cancer, Ann. Rev. Pathol.: Mech. Dis. 4 (2009) https://doi.org/10.4161/cc.7.7.5666.
199–227, https://doi.org/10.1146/annurev.pathol.4.110807.092222. [116] P. Lopez-Serra, M. Esteller, DNA methylation-associated silencing of tumor-
[90] J. Liu, M.A. Valencia-Sanchez, G.J. Hannon, R. Parker, MicroRNA-dependent suppressor microRNAs in cancer, Oncogene 31 (13) (2012) 1609–1622, https://
localization of targeted mRNAs to mammalian P-bodies, Nat. Cell Biol. 7 (7) doi.org/10.1038/onc.2011.354.
(2005) 719–723, https://doi.org/10.1038/ncb1274. [117] Y. Saito, G. Liang, G. Egger, J.M. Friedman, J.C. Chuang, G.A. Coetzee, P.A. Jones,
[91] C.Y. Chu, T.M. Rana, Translation repression in human cells by microRNA-induced Specific activation of microRNA-127 with downregulation of the proto-oncogene
gene silencing requires RCK/p54, PLoS Biol. 4 (7) (2006), https://doi.org/ BCL6 by chromatin-modifying drugs in human cancer cells, Cancer Cell 9 (6)
10.1371/journal.pbio. 0040210. (2006) 435–443, https://doi.org/10.1016/j.ccr.2006.04.020.
[92] A. Grimson, K.K.H. Farh, W.K. Johnston, P. Garrett-Engele, L.P. Lim, D.P. Bartel, [118] U. Lehmann, B. Hasemeier, M. Christgen, M. Müller, D. Römermann, F. Länger,
MicroRNA targeting specificity in mammals: determinants beyond seed pairing, H. Kreipe, Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast
Mol. Cell 27 (1) (2007) 91–105, https://doi.org/10.1016/j.molcel.2007.06.017. cancer, J. Pathol. 214 (1) (2008) 17–24, https://doi.org/10.1002/path.2251.
[93] I. Behm-Ansmant, J. Rehwinkel, T. Doerks, A. Stark, P. Bork, E. Izaurralde, mRNA [119] M. Toyota, H. Suzuki, Y. Sasaki, R. Maruyama, K. Imai, Y. Shinomura, T. Tokino,
degradation by miRNAs and GW182 requires both CCR4: NOT deadenylase and Epigenetic silencing of microRNA-34b/c and B-cell translocation gene 4 is
DCP1: DCP2 decapping complexes, Genes Dev. 20 (14) (2006) 1885–1898, associated with CpG island methylation in colorectal cancer, Cancer Res. 68 (11)
https://doi.org/10.1101/gad.1424106. (2008) 4123–4132, https://doi.org/10.1158/0008-5472.CAN-08-0325.
[94] M.P. Gantier, A.J. Sadler, B.R. Williams, Fine-tuning of the innate immune [120] A. Lujambio, S. Ropero, E. Ballestar, M.F. Fraga, C. Cerrato, F. Setién, I. Spiteri,
response by microRNAs, Immunol. Cell Biol. 85 (6) (2007) 458–462, https://doi. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells,
org/10.1038/sj.icb.7100091. Cancer Res. 67 (4) (2007) 1424–1429, https://doi.org/10.1158/0008-5472.CAN-
[95] R. Garzon, G.A. Calin, C.M. Croce, MicroRNAs in cancer, Annu. Rev. Med. 60 06-4218.
(2009) 167–179, https://doi.org/10.1146/annurev.med.59.053006.104707. [121] B. Brueckner, C. Stresemann, R. Kuner, C. Mund, T. Musch, M. Meister, F. Lyko,
[96] C.S. Sullivan, D. Ganem, MicroRNAs and viral infection, Mol. Cell 20 (1) (2005) The human let-7a-3 locus contains an epigenetically regulated microRNA gene
3–7, https://doi.org/10.1016/j.molcel.2005.09.012. with oncogenic function, Cancer Res. 67 (4) (2007) 1419–1423, https://doi.org/
[97] M.N. Poy, L. Eliasson, J. Krutzfeldt, S. Kuwajima, X. Ma, P.E. Macdonald, 10.1158/0008-5472.CAN-06-4074.
S. Pfeffer, T. Tuschl, N. Rajewsky, P. Rorsman, M. Stoffel, A pancreatic islet- [122] G.K. Scott, M.D. Mattie, C.E. Berger, S.C. Benz, C.C. Benz, Rapid alteration of
specific microRNA regulates insulin secretion, Nature 432 (7014) (2004) microRNA levels by histone deacetylase inhibition, Cancer Res. 66 (3) (2006)
226–230, https://doi.org/10.1038/nature03076. 1277–1281, https://doi.org/10.1158/0008-5472.CAN-05-3632.
[98] S. Chang, R.J. Johnston, C. Frøkjær-Jensen, S. Lockery, O. Hobert, MicroRNAs act [123] M. Fabbri, R. Garzon, A. Cimmino, Z. Liu, N. Zanesi, E. Callegari, S. Volinia,
sequentially and asymmetrically to control chemosensory laterality in the MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting
nematode, Nature 430 (7001) (2004) 785–789, https://doi.org/10.1038/ DNA methyltransferases 3A and 3B, Proc. Natl. Acad. Sci. 104 (40) (2007)
nature02752. 15805–15810, https://doi.org/10.1073/pnas.0707628104.
[99] F. Fazi, A. Rosa, A. Fatica, V. Gelmetti, M.L. De Marchis, C. Nervi, I. Bozzoni, [124] S. Varambally, Q. Cao, R.S. Mani, S. Shankar, X. Wang, B. Ateeq, J.C. Brenner,
A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and Genomic loss of microRNA-101 leads to overexpression of histone
C/EBPα regulates human granulopoiesis, Cell 123 (5) (2005) 819–831, https:// methyltransferase EZH2 in cancer, Science 322 (5908) (2008) 1695–1699,
doi.org/10.1016/j.cell.2005.09.023. https://doi.org/10.1126/science.1165395.
[100] S. Yekta, I.H. Shih, D.P. Bartel, MicroRNA-directed cleavage of HOXB8 mRNA, [125] L. Ma, J. Teruya-Feldstein, R.A. Weinberg, Tumour invasion and metastasis
Science 304 (5670) (2004) 594–596, https://doi.org/10.1126/science.1097434. initiated by microRNA-10b in breast cancer, Nature 449 (7163) (2007) 682–688,
[101] M.Z. Michael, S.M. O’Connor, N.G. van Holst Pellekaan, G.P. Young, R.J. James, https://doi.org/10.1038/nature06174.
Reduced Accumulation of Specific MicroRNAs in Colorectal Neoplasia11Note: [126] G.T. Bommer, I. Gerin, Y. Feng, A.J. Kaczorowski, R. Kuick, R.E. Love, Y. Zhai, T.
Susan M. O’Connor and Nicholas G. van Holst Pellekaan contributed equally to J. Giordano, Z.S. Qin, B.B. Moore, O.A. MacDougald, p53-mediated activation of
this work, Mol. Cancer Res. 1 (12) (2003) 882–891. PMID: 14573789. miRNA34 candidate tumor-suppressor genes, Curr. Biol. 17 (15) (2007)
[102] L. He, J.M. Thomson, M.T. Hemann, E. Hernando-Monge, D. Mu, S. Goodson, 1298–1307, https://doi.org/10.1016/j.cub.2007.06.068.
S. Powers, C. Cordon-Cardo, S.W. Lowe, G.J. Hannon, S.M. Hammond, [127] V. Tarasov, P. Jung, B. Verdoodt, D. Lodygin, A. Epanchintsev, A. Menssen,
A microRNA polycistron as a potential human oncogene, Nature 435 (7043) G. Meister, H. Hermeking, Differential regulation of microRNAs by p53 revealed
(2005) 828–833, https://doi.org/10.1038/nature03552. by massively parallel sequencing: miR-34a is a p53 target that induces apoptosis
[103] Q. Jing, S. Huang, S. Guth, T. Zarubin, A. Motoyama, J. Chen, F. Di Padova, S. and G1-arrest, Cell Cycle 6 (13) (2007) 1586–1593, https://doi.org/10.4161/
C. Lin, H. Gram, J. Han, Involvement of microRNA in AU-rich element-mediated cc.6.13.4436.
mRNA instability, Cell 120 (5) (2005) 623–634, https://doi.org/10.1016/j. [128] L. He, X. He, L.P. Lim, E. De Stanchina, Z. Xuan, Y. Liang, W. Xue, L. Zender,
cell.2004.12.038. J. Magnus, D. Ridzon, A.L. Jackson, A microRNA component of the p53 tumour
[104] P. Xu, S.Y. Vernooy, M. Guo, B.A. Hay, The Drosophila microRNA Mir-14 suppressor network, Nature 447 (7148) (2007) 1130–1134, https://doi.org/
suppresses cell death and is required for normal fat metabolism, Curr. Biol. 13 (9) 10.1038/nature05939.
(2003) 790–795, https://doi.org/10.1016/S0960-9822(03)00250-1. [129] O.A. Kent, J.T. Mendell, A small piece in the cancer puzzle: microRNAs as tumor
[105] F.J. Slack, M. Basson, Z. Liu, V. Ambros, H.R. Horvitz, G. Ruvkun, The lin-41 suppressors and oncogenes, Oncogene 25 (46) (2006) 6188–6196, https://doi.
RBCC gene acts in the C. elegans heterochronic pathway between the let-7 org/10.1038/sj.onc.1209913.
regulatory RNA and the LIN-29 transcription factor, Mol. Cell 5 (4) (2000) [130] H. He, K. Jazdzewski, W. Li, S. Liyanarachchi, R. Nagy, S. Volinia, G.A. Calin, C.
659–669, https://doi.org/10.1016/S1097-2765(00)80245-2. G. Liu, K. Franssila, S. Suster, R.T. Kloos, The role of microRNA genes in papillary
[106] J.E. Abrahante, A.L. Daul, M. Li, M.L. Volk, J.M. Tennessen, E.A. Miller, A. thyroid carcinoma, Proc. Natl. Acad. Sci. 102 (52) (2005) 19075–19080, https://
E. Rougvie, The Caenorhabditis elegans hunchback-like gene lin-57/hbl-1 doi.org/10.1073/pnas.0509603102.

20
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

[131] A. Ota, H. Tagawa, S. Karnan, S. Tsuzuki, A. Karpas, S. Kira, Y. Yoshida, M. Seto, [157] P.M. Voorhoeve, C. Le Sage, M. Schrier, A.J. Gillis, H. Stoop, R. Nagel,
Identification and characterization of a novel gene, C13orf25, as a target for E. Zlotorynski, A genetic screen implicates miRNA-372 and miRNA-373 as
13q31-q32 amplification in malignant lymphoma, Cancer Res. 64 (9) (2004) oncogenes in testicular germ cell tumors, Cell 124 (6) (2006) 1169–1181, https://
3087–3095, https://doi.org/10.1158/0008-5472.CAN-03-3773. doi.org/10.1016/j.cell.2006.02.037.
[132] S. Volinia, G.A. Calin, C.G. Liu, S. Ambs, A. Cimmino, F. Petrocca, R. Visone, [158] W.S. El-Deiry, T. Tokino, V.E. Velculescu, D.B. Levy, R. Parsons, J.M. Trent,
M. Iorio, C. Roldo, M. Ferracin, R.L. Prueitt, A microRNA expression signature of D. Lin, W.E. Mercer, K.W. Kinzler, B. Vogelstein, WAF1, a potential mediator of
human solid tumors defines cancer gene targets, Proc. Natl. Acad. Sci. 103 (7) p53 tumor suppression, Cell 75 (4) (1993) 817–825, https://doi.org/10.1016/
(2006) 2257–2261, https://doi.org/10.1073/pnas.0510565103. 0092-8674(93)90500-p.
[133] H. Tagawa, K. Karube, S. Tsuzuki, K. Ohshima, M. Seto, Synergistic action of the [159] Z. Li, S. Van Calcar, C. Qu, W.K. Cavenee, M.Q. Zhang, B. Ren, A global
microRNA-17 polycistron and Myc in aggressive cancer development, Cancer Sci. transcriptional regulatory role for c-Myc in Burkitt’s lymphoma cells, Proc. Natl.
98 (9) (2007) 1482–1490, https://doi.org/10.1111/j.1349-7006.2007.00531.x. Acad. Sci. 100 (14) (2003) 8164–8169, https://doi.org/10.1073/
[134] H. Tagawa, M. Seto, A microRNA cluster as a target of genomic amplification in pnas.1332764100.
malignant lymphoma, Leukemia 19 (11) (2005) 2013–2016, https://doi.org/ [160] Q. Song, Q. An, B. Niu, X. Lu, N. Zhang, X. Cao, Role of miR-221/222 in Tumor
10.1038/sj.leu.2403942. Development and the Underlying Mechanism, J. Oncol. 2 (2019) 7252013,
[135] J.H. Schulte, S. Horn, T. Otto, B. Samans, L.C. Heukamp, U.C. Eilers, M. Krause, https://doi.org/10.1155/2019/7252013.
K. Astrahantseff, L. Klein-Hitpass, R. Buettner, A. Schramm, MYCN regulates [161] E. Callegari, B.K. Elamin, F. Giannone, M. Milazzo, G. Altavilla, F. Fornari,
oncogenic MicroRNAs in neuroblastoma, Int. J. Cancer 122 (3) (2008) 699–704, L. Giacomelli, L. D’Abundo, M. Ferracin, C. Bassi, B. Zagatti, Liver tumorigenicity
https://doi.org/10.1002/ijc.23153. promoted by microRNA-221 in a mouse transgenic model, Hepatology 56 (3)
[136] Y. Sylvestre, V. De Guire, E. Querido, U.K. Mukhopadhyay, V. Bourdeau, F. Major, (2012) 1025–1033, https://doi.org/10.1002/hep.25747.
G. Ferbeyre, P. Chartrand, An E2F/miR-20a autoregulatory feedback loop, J. Biol. [162] S.U. Mertens-Talcott, S. Chintharlapalli, X. Li, S. Safe, The oncogenic microRNA-
Chem. 282 (4) (2007) 2135–2143, https://doi.org/10.1074/jbc.M608939200. 27a targets genes that regulate specificity protein transcription factors and the
[137] K. Woods, J.M. Thomson, S.M. Hammond, Direct regulation of an oncogenic G2-M checkpoint in MDA-MB-231 breast cancer cells, Cancer Res. 67 (22) (2007)
micro-RNA cluster by E2F transcription factors, J. Biol. Chem. 282 (4) (2007) 11001–11011, https://doi.org/10.1158/0008-5472.CAN-07-2416.
2130–2134, https://doi.org/10.1074/jbc.C600252200. [163] M.A. Samols, R.L. Skalsky, A.M. Maldonado, A. Riva, M.C. Lopez, H.V. Baker,
[138] J.L. Bos, Ras oncogenes in human cancer: a review, Cancer Res. 49 (17) (1989) R. Renne, Identification of cellular genes targeted by KSHV-encoded microRNAs,
4682–4689. 2547513. PLoS Pathog. 3 (5) (2007), https://doi.org/10.1371/journal.ppat.0030065.
[139] S.M. Johnson, H. Grosshans, J. Shingara, M. Byrom, R. Jarvis, A. Cheng, F. [164] T.C. Chang, E.A. Wentzel, O.A. Kent, K. Ramachandran, M. Mullendore, K.H. Lee,
J. Slack, RAS is regulated by the let-7 microRNA family, Cell 120 (5) (2005) D.E. Arking, Transactivation of miR-34a by p53 broadly influences gene
635–647, https://doi.org/10.1016/j.cell.2005.01.014. expression and promotes apoptosis, Mol. Cell 26 (5) (2007) 745–752, https://doi.
[140] R. Reeves, Molecular biology of HMGA proteins: hubs of nuclear function, Gene org/10.1016/j.molcel.2007.05.010.
277 (1-2) (2001) 63–81, https://doi.org/10.1016/S0378-1119(01)00689-8. [165] S. Galardi, N. Mercatelli, E. Giorda, S. Massalini, G.V. Frajese, S.A. Ciafrè, M.
[141] T. Wang, X. Zhang, L. Obijuru, J. Laser, V. Aris, P. Lee, J.J. Wei, A micro-RNA G. Farace, miR-221 and miR-222 expression affects the proliferation potential of
signature associated with race, tumor size, and target gene activity in human human prostate carcinoma cell lines by targeting p27Kip1, J. Biol. Chem. 282
uterine leiomyomas, Genes Chromosom. Cancer 46 (4) (2007) 336–347, https:// (32) (2007) 23716–23724, https://doi.org/10.1074/jbc.M701805200.
doi.org/10.1002/gcc.20415. [166] A.L. Kasinski, F.J. Slack, MicroRNAs en route to the clinic: progress in validating
[142] X. Jiang, Wang X. Cytochrome-c-mediated apoptosis, Annu. Rev. Biochem. 73 and targeting microRNAs for cancer therapy, Nat. Rev. Cancer 11 (12) (2011)
(2004) 87–106, https://doi.org/10.1146/annurev.biochem.73.011303.073706. 849–864, https://doi.org/10.1038/nrc3166.
[143] S. Cory, D.C. Huang, J.M. Adams, The Bcl-2 family: roles in cell survival and [167] M. Maddalena, C. Lucia, MicroRNAs used in combination with anti-cancer
oncogenesis, Oncogene 22 (53) (2003) 8590–8607, https://doi.org/10.1038/sj. treatments can enhance therapy efficacy, Mini-Rev. Med. Chem. 15 (2015)
onc.1207102. 1052–1062, https://doi.org/10.2174/1389557515666150709115355.
[144] W. Tam, in: W. Tam, D. Ben-Yehuda, W.S. Hayward (Eds.), Mol. Cell. Biol. 17 [168] W. Zhu, H. Xu, D. Zhu, H. Zhi, T. Wang, J. Wang, B. Jiang, Y. Shu, P. Liu, miR-
(1997) 1490–1502. 200bc/429 cluster modulates multidrug resistance of human cancer cell lines by
[145] W. Tam, S.H. Hughes, W.S. Hayward, P. Besmer, Avian bic, a gene isolated from a targeting BCL2 and XIAP, Cancer Chemother. Pharmacol. 69 (2012) 723–731,
common retroviral site in avian leukosis virus-induced lymphomas that encodes a https://doi.org/10.1007/s00280-011-1752-3.
noncoding RNA, cooperates with c-myc in lymphomagenesis and [169] H.M. Jeon, Y.W. Sohn, S.Y. Oh, S.H. Kim, S. Beck, S. Kim, H. Kim, ID4 imparts
erythroleukemogenesis, J. Virol. 76 (9) (2002) 4275–4286, https://doi.org/ chemoresistance and cancer stemness to glioma cells by depressing miR-
10.1128/JVI.76.9.4275-4286.2002. 9*–mediated suppression of SOX2, Cancer Res. 71 (9) (2011) 3410–3421, https://
[146] P.S. Eis, W. Tam, L. Sun, A. Chadburn, Z. Li, M.F. Gomez, J.E. Dahlberg, doi.org/10.1158/0008-5472.CAN-10-3340.
Accumulation of miR-155 and BIC RNA in human B cell lymphomas, Proc. Natl. [170] P. Ye, C. Fang, H. Zeng, Y. Shi, Z. Pan, N. An, K. He, L. Zhang, X. Long, Differential
Acad. Sci. 102 (10) (2005) 3627–3632, https://doi.org/10.1073/ microRNA expression profiles in tamoxifen-resistant human breast cancer cell
pnas.0500613102. lines induced by two methods, Oncol. Lett. 15 (3) (2018) 3532–3539, https://doi.
[147] S. Costinean, N. Zanesi, Y. Pekarsky, E. Tili, S. Volinia, N. Heerema, C.M. Croce, org/10.3892/ol.2018.7768.
Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in Eμ- [171] D.M. Cittelly, P.M. Das, N.S. Spoelstra, S.M. Edgerton, J.K. Richer, A.D. Thor, F.
miR155 transgenic mice, Proc. Natl. Acad. Sci. 103 (18) (2006) 7024–7029, E. Jones, Downregulation of miR-342 is associated with tamoxifen resistant breast
https://doi.org/10.1073/pnas.0602266103. tumors, Mol. Cancer 9 (1) (2010) 317, https://doi.org/10.1186/1476-4598-9-
[148] J.A. Chan, A.M. Krichevsky, K.S. Kosik, MicroRNA-21 is an antiapoptotic factor in 317.
human glioblastoma cells, Cancer Res. 65 (14) (2005) 6029–6033, https://doi. [172] T.E. Miller, K. Ghoshal, B. Ramaswamy, S. Roy, J. Datta, C.L. Shapiro,
org/10.1158/0008-5472.CAN-05-0137. S. Majumder, MicroRNA-221/222 confers tamoxifen resistance in breast cancer
[149] L. Qi, J. Bart, L.P. Tan, et al., Expression of miR-21 and its targets (PTEN, PDCD4, by targeting p27Kip1, J. Biol. Chem. 283 (44) (2008) 29897–29903, https://doi.
TM1) in flat epithelial atypia of the breast in relation to ductal carcinoma in situ org/10.1074/jbc.M804612200.
and invasive carcinoma, BMC Cancer 9 (2009) 163, https://doi.org/10.1186/ [173] J. Zhu, L. Tao, L. Jin, MicroRNA-506-3p reverses gefitinib resistance in non-small
1471-2407-9-163. cell lung cancer by targeting Yes-associated protein 1, Mol. Med. Rep. 19 (2)
[150] F. Meng, R. Henson, Wehbe–Janek, H., Ghoshal, K., Jacob, S. T., & Patel, T., (2019) 1331–1339, https://doi.org/10.3892/mmr.2018.9710.
MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human [174] M. Zhong, X. Ma, C. Sun, L. Chen, MicroRNAs reduce tumor growth and
hepatocellular cancer, Gastroenterology 133 (2) (2007) 647–658, https://doi. contribute to enhance cytotoxicity induced by gefitinib in non-small cell lung
org/10.1053/j.gastro.2007.05.022. cancer, Chem. Biol. Interact. 184 (3) (2010) 431–438, https://doi.org/10.1016/j.
[151] R.H. Kim, T.W. Mak, Tumours and tremors: how PTEN regulation underlies both, cbi.2010.01.025.
Br. J. Cancer 94 (5) (2006) 620–624, https://doi.org/10.1038/sj.bjc.6602994. [175] P. Ge, L. Cao, X. Chen, et al., miR-762 activation confers acquired resistance to
[152] M. Iijima, P. Devreotes, Tumor suppressor PTEN mediates sensing of gefitinib in non-small cell lung cancer, BMC Cancer 19 (2019) 1203, https://doi.
chemoattractant gradients, Cell 109 (5) (2002) 599–610, https://doi.org/ org/10.1186/s12885-019-6416-4.
10.1016/s0092-8674(02)00745-6. [176] Y. Liu, L. Yang, F. Liao, et al., MiR-450a-5p strengthens the drug sensitivity of
[153] S. Zhu, H. Wu, F. Wu, D. Nie, S. Sheng, Y.Y. Mo, MicroRNA-21 targets tumor gefitinib in glioma chemotherapy via regulating autophagy by targeting EGFR,
suppressor genes in invasion and metastasis, Cell Res. 18 (3) (2008) 350–359, Oncogene 39 (2020) 6190–6202, https://doi.org/10.1038/s41388-020-01422-9.
https://doi.org/10.1038/cr.2008.24. [177] Y. Azuma, T. Yokobori, A. Mogi, et al., Cancer exosomal microRNAs from
[154] M.J. Park, M.S. Kim, I.C. Park, H.S. Kang, H. Yoo, S.H. Park, S.H. Lee, PTEN gefitinib-resistant lung cancer cells cause therapeutic resistance in gefitinib-
suppresses hyaluronic acid-induced matrix metalloproteinase-9 expression in sensitive cells, Surg. Today 50 (2020) 1099–1106, https://doi.org/10.1007/
U87MG glioblastoma cells through focal adhesion kinase dephosphorylation, s00595-020-01976-x.
Cancer Res. 62 (21) (2002) 6318–6322. PMID: 12414663. [178] M. Ashrafizadeh, A. Zarrabi, K. Hushmandi, F. Hashemi, F. Hashemi,
[155] S. Zhu, M.L. Si, H. Wu, Y.Y. Mo, MicroRNA-21 targets the tumor suppressor gene S. Samarghandian, M. Najafi, MicroRNAs in cancer therapy: Their involvement in
tropomyosin 1 (TPM1), J. Biol. Chem. 282 (19) (2007) 14328–14336, https://doi. oxaliplatin sensitivity/resistance of cancer cells with a focus on colorectal cancer,
org/10.1074/jbc.M611393200. Life Sci. 117973 (2020), https://doi.org/10.1016/j.lfs.2020.117973.
[156] M. Serrano, A.W. Lin, M.E. McCurrach, D. Beach, S.W. Lowe, Oncogenic ras [179] J. Han, W. Sun, R. Liu, Z. Zhou, H. Zhang, X. Chen, Y. Ba, Plasma exosomal
provokes premature cell senescence associated with accumulation of p53 and miRNA expression profile as oxaliplatin-based chemoresistant biomarkers in
p16INK4a, Cell 88 (5) (1997) 593–602, https://doi.org/10.1016/S0092-8674 colorectal adenocarcinoma, Front. Oncol. 10 (2020) 1495. https://www.frontier
(00)81902-9. sin.org/article/10.3389/fonc.2020.01495.

21
M. Budakoti et al. Cellular Signalling 83 (2021) 109995

[180] S. Lim, Y. Kim, S.B. Lee, et al., Inhibition of Chk1 by miR-320c increases [189] C.P. O’Neill, R.M. Dwyer, Nanoparticle-based delivery of tumor suppressor
oxaliplatin responsiveness in triple-negative breast cancer, Oncogenesis 9 (2020) microRNA for cancer therapy, Cells 9 (2) (2020) 521, https://doi.org/10.3390/
91, https://doi.org/10.1038/s41389-020-00275-x. cells9020521.
[181] R.M. Marjaneh, M. Khazaei, G.A. Ferns, A. Avan, S.H. Aghaee-Bakhtiari, The role [190] M. Moro, D. Di Paolo, M. Milione, G. Centonze, V. Bornaghi, C. Borzi,
of microRNAs in 5-FU resistance of colorectal cancer: Possible mechanisms, P. Gandellini, P. Perri, U. Pastorino, M. Ponzoni, G. Sozzi, Coated cationic lipid-
J. Cell. Physiol. 234 (3) (2018) 2306–2316, https://doi.org/10.1002/jcp.27221. nanoparticles entrapping miR-660 inhibit tumor growth in patient-derived
[182] F. Xu, M.L. Ye, Y.P. Zhang, W.J. Li, M.T. Li, H.Z. Wang, X. Qiu, Y. Xu, J.W. Yin, xenografts lung cancer models, J. Control. Release 308 (2019) 44–56, https://doi.
Q. Hu, W.H. Wei, Y. Chang, L. Liu, Q. Zhao, MicroRNA-375-3p enhances org/10.1016/j.jconrel.2019.07.006.
chemosensitivity to 5-fluorouracil by targeting thymidylate synthase in colorectal [191] K. Gokita, J. Inoue, H. Ishihara, K. Kojima, J. Inazawa, Therapeutic potential of
cancer, Cancer Sci. 111 (5) (2020) 1528–1541, https://doi.org/10.1111/ lnp-mediated delivery of miR-634 for cancer therapy, Mol. Therapy-Nucleic Acids
cas.14356. 19 (2020) 330–338, https://doi.org/10.1016/j.omtn.2019.10.045.
[183] A. Holleman, I. Chung, R.R. Olsen, et al., miR-135a contributes to paclitaxel [192] U.K. Sukumar, R.J. Bose, M. Malhotra, H.A. Babikir, R. Afjei, E. Robinson,
resistance in tumor cells both in vitro and in vivo, Oncogene 30 (2011) 4386–4398, Y. Zeng, E. Chang, F. Habte, R. Sinclair, S.S. Gambhir, Intranasal delivery of
https://doi.org/10.1038/onc.2011.148. targeted polyfunctional gold–iron oxide nanoparticles loaded with therapeutic
[184] D.R. Cochrane, N.S. Spoelstra, E.N. Howe, et al., MicroRNA-200c mitigates microRNAs for combined theranostic multimodality imaging and presensitization
invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic of glioblastoma to temozolomide, Biomaterials 218 (2019) 119342, https://doi.
agents, Mol. Cancer Ther. 8 (2009) 1055–1066, https://doi.org/10.1158/1535- org/10.1016/j.biomaterials.2019.119342.
7163.MCT-08-1046. [193] G. Oshima, N. Guo, C. He, M.E. Stack, C. Poon, A. Uppal, S.C. Wightman,
[185] H. Wu, M. Huang, M. Lu, et al., Regulation of microtubule-associated protein tau A. Parekh, K.B. Skowron, M.C. Posner, W. Lin, In vivo delivery and therapeutic
(MAPT) by miR-34c-5p determines the chemosensitivity of gastric cancer to effects of a microRNA on colorectal liver metastases, Mol. Ther. 25 (7) (2017)
paclitaxel, Cancer Chemother. Pharmacol. 71 (2013) 1159–1171, https://doi. 1588–1595, https://doi.org/10.1016/j.ymthe.2017.04.005.
org/10.1007/s00280-013-2108-y. [194] J.A. Weber, D.H. Baxter, S. Zhang, D.Y. Huang, K. How Huang, M. Jen Lee,
[186] D. Chen, C. Bao, F. Zhao, H. Yu, G. Zhong, L. Xu, S. Yan, Exploring specific K. Wang, The microRNA spectrum in 12 body fluids, Clin. Chem. 56 (11) (2010)
miRNA-mRNA axes with relationship to taxanes-resistance in breast cancer, 1733–1741, https://doi.org/10.1373/clinchem.2010.147405.
Front. Oncol. 10 (2020) 1397. https://www.frontiersin.org/article/10.33 [195] K. Wang, S. Zhang, B. Marzolf, P. Troisch, A. Brightman, Z. Hu, D.J. Galas,
89/fonc.2020.01397. Circulating microRNAs, potential biomarkers for drug-induced liver injury, Proc.
[187] M.K. Hassan, A.A. Waly, W. Elsayed, et al., Integrative microRNA and gene Natl. Acad. Sci. 106 (11) (2009) 4402–4407, https://doi.org/10.1073/
expression analysis identifies new epigenetically regulated microRNAs mediating pnas.0813371106.
taxane resistance in ovarian cancer, Sci. Rep. 11 (2021) 562, https://doi.org/ [196] H. Valadi, K. Ekström, A. Bossios, M. Sjöstrand, J.J. Lee, J.O. Lötvall, Exosome-
10.1038/s41598-020-78596-5. mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic
[188] K.P. O’brien, S. Khan, K.E. Gilligan, H. Zafar, P. Lalor, C. Glynn, C. O’Flatharta, exchange between cells, Nat. Cell Biol. 9 (6) (2007) 654–659, https://doi.org/
H. Ingoldsby, P. Dockery, A. De Bhulbh, J.R. Schweber, Employing mesenchymal 10.1038/ncb1596.
stem cells to support tumor-targeted delivery of extracellular vesicle (EV)- [197] J.D. Arroyo, J.R. Chevillet, E.M. Kroh, I.K. Ruf, C.C. Pritchard, D.F. Gibson, J.
encapsulated microRNA-379, Oncogene 37 (16) (2018) 2137–2149, https://doi. F. Tait, Argonaute2 complexes carry a population of circulating microRNAs
org/10.1038/s41388-017-0116-9. independent of vesicles in human plasma, Proc. Natl. Acad. Sci. 108 (12) (2011)
5003–5008, https://doi.org/10.1073/pnas.1019055108.

22

You might also like