ArticlePDF Available

Optical waveguides formed by silver ion exchange in Schott SG11 glass for waveguide evanescent field fluorescence microscopy: Evanescent images of HEK293 cells

Authors:

Abstract

Planar glass waveguides with a specific number of modes were fabricated by Ag(+)-Na(+) exchange in Schott SG11 glass. The effective refractive indices were determined using m-line spectroscopy in both s- and p-polarization. By using the reversed Wentzel-Kramers-Brillouin approximation, the index profiles were described by a nonlinear diffusion equation. The diffusion coefficients for Ag(+) were established, as well as the penetration depth of the evanescent field in an aqueous environment for the different modes. The integrals of \E\(2) fields for the evanescent-guided fields were investigated. These are important when evanescent fields are used for illumination in interface microscopy, an alternative method to total internal reflection fluorescence (TIRF) microscopy. The photoluminescent behavior of the waveguides was investigated as a function of ion exchange time and excitation wavelengths. Comparable images were obtained of fluorescently labeled HEK293 cells using TIRF microscopy and waveguide evanescent field fluorescence microscopy. Imaging was performed using HEK293 cells, delivering similar images and information.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
Available online 1 October 2022
0141-8130/© 2022 Published by Elsevier B.V.
Review
Non-coding RNAs targeting notch signaling pathway in cancer: From
proliferation to cancer therapy resistance
Mehrdad Hashemi
a
,
b
, Sahar Hasani
b
, Shima Hajimazdarany
b
,
c
, Seyed Reza Mirmazloomi
b
,
Sara Makvandy
d
, Abbas Zabihi
e
, Yeganeh Goldoost
b
, Nazanin Gholinia
b
,
Amirabbas Kakavand
b
, Alireza Tavakolpournegari
f
, Shokooh Salimimoghadam
g
,
*
,
Noushin Nabavi
h
, Ali Zarrabi
i
, Afshin Taheriazam
b
,
j
,
**
, Maliheh Entezari
a
,
b
,
***
,
Kiavash Hushmandi
k
,
*
a
Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
b
Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
c
Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
d
Department of Biology, Zarghan Branch, Islamic Azad University, Zarghan, Iran
e
Department of Biology, Faculty of Basic Sciences, Islamic Azad University, Hamedan Branch, Hamedan, Iran
f
Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Aut`
onoma de Barcelona, Cerdanyola del Vall`
es, 08193 Barcelona,
Spain
g
Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
h
Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
i
Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey
j
Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
k
Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
ARTICLE INFO
Keywords:
Cancer
Notch
miRNA
lncRNA
Cancer therapy
ABSTRACT
Cancer is a challenging to treat disease with a high mortality rate worldwide, nevertheless advances in science
has led to a decrease in the number of death cases caused by cancer. Aberrant expression of genes occurs during
tumorigenesis therefore targeting the signaling pathways that regulate these genes' expression is of importance in
cancer therapy. Notch is one of the signaling pathways having interactions with other vital cell signaling mol-
ecules responsible for cellular functions such as proliferation, apoptosis, invasion, metastasis, epithelial-to-
mesenchymal transition (EMT), angiogenesis, and immune evasion. Furthermore, the Notch pathway is
involved in response to chemo- and radiotherapy. Thus, targeting the Notch signaling pathway in cancer therapy
can be benecial for overcoming the therapeutic gaps. Non-coding RNAs (ncRNAs) are a class of RNAs that
include short ncRNAs (such as micro RNAs) and long ncRNAs (lncRNAs). MicroRNAs (miRNAs) are ~22 nu-
cleotides in length while lncRNAs have more than 200 nucleotides. Both miRNAs and lncRNAs control vital
cellular mechanisms in cells and affect various signaling pathways and Notch is among them. The current review
aims to discuss the critical role of ncRNAs in the regulation of the Notch signaling pathway by focusing on
different cancer hallmarks including proliferation, apoptosis, autophagy, EMT, invasion, metastasis, and resis-
tance to therapies.
* Corresponding authors.
** Correspondence to: A. Taheriazam, Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
*** Correspondence to: M. Entezari, Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University,
Tehran, Iran.
E-mail addresses: Shokoohmoghadam@yahoo.com (S. Salimimoghadam), a.taheriazam@iautmu.ac.ir (A. Taheriazam), mentezari@iautmu.ac.ir (M. Entezari),
Kiavash.hushmandi@gmail.com (K. Hushmandi).
Contents lists available at ScienceDirect
International Journal of Biological Macromolecules
journal homepage: www.elsevier.com/locate/ijbiomac
https://doi.org/10.1016/j.ijbiomac.2022.09.203
Received 3 September 2022; Accepted 22 September 2022
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1152
1. Introduction
As the most life-threatening disease, cancer is one of the main causes
of death worldwide. In the United States, almost 2 million new cancer
cases were recognized and the number of patients who die from cancer is
estimated to be more than 600,000 in 2022 [1]. Common causes of
death from cancer are local recurrence and distant metastasis [2]. Many
cancers have an invasive nature and among them, lung, breast, prostate,
and gastrointestinal cancers have devoted the most mortality rate to
themselves. Thanks to the past and current progress against cancer,
early-stage detection, targeted therapies, and aggressive techniques like
surgeries have brought cancer treatment under control, but it remains
the most difcult-to-treat disease yet [1].
During tumorigenesis, normal cells are converted to tumor cells and
genes have a signicant role in this process. Tumor cells are known by
certain characteristics called cancer hallmarks that include sustained
proliferation, growth suppressors' dysfunction, metabolic reprogram-
ming, resistance to apoptosis, angiogenesis promotion, invasion, and
metastasis [3]. Increasing evidence suggests that tumorigenesis is highly
regulated by cellular signaling pathways. One of the fascinating
signaling pathways is Notch which is broadly involved in cancer biology
and attracts notable attention for designing Notch-targeting therapies
for cancer as a promising tool [4]. Notch is a highly carcinogenic
pathway that regulates various cancer hallmarks and plays a critical role
in cancer cell proliferation, apoptosis inhibition, and cell fate. Thus, it
could be a valuable diagnostic and therapeutic target in cancer [5].
Recent insights have linked non-coding RNAs (ncRNAs) to cancer
biology, therapy, and resistance to conventional therapies [6,7]. More
than a fth of the human genome is transcribed to RNA during different
conditions. However, only 2 % of these transcriptomes can be translated
into proteins. The vast part of the remaining untranslated transcriptome
includes a notable number of ncRNAs. ncRNAs are the dominant form of
RNA. More than 95 % of them are classied by their length; short non-
coding RNAs (sncRNAs) and long non-coding RNAs (lncRNAs) which
have less and more than 200 nucleotides, respectively. ncRNAs tightly
regulate cellular function by targeting gene expression, protein trans-
lation, and reorganizing the genome. Moreover, ncRNAs have been
explored extensively in cancer progression for their roles in carcino-
genesis or tumor suppression [811].
This review details the regulation of Notch signaling by ncRNAs in
different cancers by focusing on cancer hallmarks including prolifera-
tion, cell survival, invasion, and metastasis. Moreover, ncRNAs' role in
cancer therapy and resistance through targeting Notch pathways is
discussed.
2. Non-coding RNAs
The most well-studied part of the genome belongs to protein-coding
genes. However, this proportion accounts for 2 % of the genome. The
other large remaining sequences of the genome are related to non-
protein coding genes which are responsible for physiological and
developmental functions. These functions are particularly apparent for a
class of ncRNAs called microRNAs (miRNAs). During the past two de-
cades, it has become gradually evident that genetic and epigenetic al-
terations in the miRNAs genome can be considered as disease hallmarks
[12,13]. ncRNAs are divided into two main classes based on their length:
small non-coding RNAs (sncRNAs) with no more than 200 nucleotides,
and long non-coding RNAs with greater than 200 nucleotides. Despite
this classication, lncRNAs have similarities to miRNAs. For example,
both of them are transcribed by RNA polymerase II. An explosion of
studies for sncRNAs suggests that they represent a diverse group and
come in a variety of molecules with different sizes and functions,
including tumor suppressors and oncogenic molecules. miRNA, piwi-
interacting RNA (piRNA), small interfering RNA (siRNA), and transfer
RNA-derived small RNAs (tsRNAs) are among very known sncRNAs
[14,15].
As master regulators, miRNAs control the expression of a vast
number of genes at different levels. Among sncRNAs, miRNAs are widely
studied in cancer biology and therapy. MiRNAs exert their function in
cancer in a tumor suppressor or tumor-promoting manner. Interestingly,
some miRNAs have a dual role of either tumor promoter or suppressor
depending on cellular context [15]. For example, miRNA-29 has
different expression levels in indolent and aggressive types of B-cell
chronic lymphocytic leukemia [16].
RNA polymerase II (Pol II) is responsible for the transcription of
miRNAs that produce pri-miRNAs with at least one hairpin region as the
substrate for Microprocessors. Microprocessors are complexes
comprising of Drosha, RNase III enzyme, and RNA-binding protein Di
George Syndrome Critical Region Gene 8 (DGCR8). After binding of
Microprocessors, Drosha cleaves pri-miRNA to produce the precursor
miRNA (pre-miRNA) with ~60 nucleotides. Once formed, pre-miRNAs
translocate into the cytoplasm by the action of Exportin 5 and Ran-
GTP complex. After being processed by the RNase III enzyme Dicer, a
miRNA duplex is generated by the function of Dicer. Then, the formed
miRNA duplex is loaded into an Argonaute protein, leading to the pro-
duction of a complex named RNA-induced silencing complex (RISC)
which contains the mature miRNA of ~22 nucleotides [17,18]. MiRNAs
regulate their targets at the post-transcriptional level by binding to the 3
untranslated region (3UTR) of their messenger RNA (mRNA). This
interaction leads to degradation of the mRNA or translational inhibition
of functional proteins [19].
A very newly discovered class of sncRNAs is tsRNAs which are also
named tRNA-derived stress-induced RNAs (tiRNAs) and tRNA fragments
(tRFs). Although tsRNAs have not been extensively studied yet, their
role in cancer is shown. They have a wide range of targets including
Argonaute and Piwi proteins which include miRNAs and piRNAs,
respectively [20]. These sncRNAs are derived from tRNA. tRNAs are
transcribed by RNA polymerase III (RNA Pol III) to produce pre-tRNA.
Then, 5and 3ends are removed by RNase P and Z, and tRNA nucleo-
tide transferase adds a CCA sequence to the 3end. The mature tRNA has
four loops including (1) D-loop, (2) anticodon loop, (3) variable loop,
and (4) pseudouracil loop (T
ψ
C loop). TsRNAs are derived from
different parts of tRNA using Dicers and/or Angiogenin. For example,
tRF-5, tRF-3, and tiRNA are derived from D-loop, T
ψ
C loop, and anti-
codon loop respectively [21]. tsRNA functions are include RNA
silencing, RNA stability, regulation of RNA reverse transcription, and
regulating protein translation. Moreover, a body of studies has illumi-
nated the role of tsRNAs in different cancers [21,22].
CircRNAs, as a class of long ncRNAs, are single-strand RNA mole-
cules without 5caps and 3tails which are generated from pre-mRNAs
transcripts during the process of back-splicing [23] or through lariat-
driven circularization. Back-splicing is mediated by cis-acting and
trans-acting factors. CircRNAs are grouped based on their site of origin:
exonic circRNAs, circular intronic RNAs, and exon-intron circRNAs
[2427]. They have different functions in the cells including miRNA
regulation, regulating translation, RNA maturation, protein localization,
role as decoys, and scaffolding. Besides, circRNA's roles in the regulation
of human disorders such as cancer have been studied. These studies
suggest that circRNAs are involved in drug resistance and oncogenesis
and can be used as promising diagnostic and prognostic biomarkers
[2833].
H19 and XIST were the rst discovered and studied lncRNAs during
studies of embryonic development and inactivation of the X chromo-
some [15,34,35]. With a length of more than 200 nucleotides, lncRNAs
are transcribed by RNA Pol II. LncRNAs genomic region is enriched with
histone H3 trimethylation at Lys36 (H3K36) through the body of related
genes and histone H3 Lys4 trimethylated (H3K4) at the start site of
transcription. Being spliced through canonical pathways, matured
lncRNAs have their 5ends capped by 7-methyl guanosine with a pol-
yadenylated 3tail [3639]. Based on the site of lncRNAs function, they
can be divided into two groups: lncRNAs that function in cis (exert local
effects close to their transcription site) or trans (exert their effects on
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1153
distant cellular or genomic regions). X-inactive specic transcript (XIST)
and HOX antisense intergenic RNA (HOTAIR) are two well-known
lncRNAs that exert their functions in cis and trans regions, respectively
[40]. LncRNAs have a myriad of functional roles in cells and affect
different genes and factors to regulate their function. For example,
lncRNAs modulate the pre-mRNA splicing process to regulate mRNA.
Moreover, they regulate the translation, and repression process medi-
ated by miRNAs, act as scaffolds, increasing genetic diversity, and play
roles in cellular signaling pathways (Fig. 1) [41].
3. Notch signaling pathway
Notch receptor gene which encodes a large transmembrane receptor
of Type I was rst studied in 1985 [42]. The notch pathway is one of the
highly conserved signaling pathways involved in creating diversity be-
tween various species and different cell types. The notch signaling
pathway has a critical role in regulating cellular processes including cell
growth, cell death, and differentiation to promote or suppress them in a
context-dependent manner. Regarding its extensive role in a high range
of cell processes and different tissues, dysregulation of Notch is linked to
various cellular dysfunctions and human disorders, cancer being among
them [43].
After being produced in the endoplasmic reticulum, Notch co-
operates with an O-fucosyl transferase (O-Fut) and is translocated into
the cytoplasm to be processed in the Golgi apparatus using an enzyme
named Furin-like convertase. Next, Notch is glycosylated by O-Fut and
glycosyltransferase factors like Fringe and is transported to the cell
membrane. By binding to its ligands, the Notch pathway is activated.
Notch ligands are proteins located on the cell membrane including an N-
terminal domain called DSL (Delta, Serrate, and LAG-2) with extracel-
lular domains that are contained repeats of epidermal growth factor
(EGF). Ligands are classied according to the absence or existence of
cysteine-rich (CR) areas into two groups: Delta or Delta-like (DLL1/3/4),
and Jagged (Jagged1/2). Notch receptor has three parts of extracellular,
transmembrane, and intracellular domains. The extracellular domain is
characterized by EGF repeats, CR LIN repeats, and the linker region that
links the extracellular part to the other parts. The intracellular domain
contains, an RBP-J association molecule (RAM), ankyrin repeats (Ank),
and a C-terminal PEST domain. Moreover, different Notch receptors may
have various protein-protein interaction motifs [4446].
Notch signaling can be activated when a ligand binds to the Notch
receptor. An appropriate ligand should effectively induce receptor
Fig. 1. Biogenesis of non-coding RNAs.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1154
proteolysis which results in releasing an active fragment of Notch from
the membrane tether, called Notch intracellular domain (NICD). Re-
ceptor proteolysis is mediated by ADAM10 or TACE (TNF-a-converting
enzyme) metalloprotease which paves the way for the function of the
γ-secretase complex to mediate a second cleavage. The produced NICD is
translocated into the nucleus to associate with a ubiquitous DNA binding
protein named CSL (CBF1/suppressor of Hairless/LaG1), the co-
activator Mastermind (Mam), Ski-interacting protein (SKIP), and other
transcription factors to mediate a transcriptional complex and regulate
downstream genes. A co-repressor (Co-R) complex that inhibits target
gene transcription is released after NICD binding to its targets. For
activation and elongation of transcription of target genes, Histone ace-
tyltransferases (HATs; including p300 and PCAF/GCN5) and chromatin-
remodeling complexes (including Dom, BRM, and TRA1/TRRAP) are
recruited. Then, cyclin-dependent kinase-8 (CDK8) and SEL10 ligase
modify NICD to make it suitable for the function of the proteasome to be
degraded. By degradation of NICD, Co-R complexes are binding to CSL
again until re-initiating the cycle by NICD (Fig. 2) [4345,47].
In addition to the canonical pathways, there is a non-canonical
pathway for activation of Notch signaling, in which Notch interacts
with other transcription factors rather than CSL, such as NF-κB, β-cat-
enin, and HIF-1
α
[46]. In non-canonical Notch signaling, Notch is acti-
vated by a non-canonical ligand or is activated without ligands.
Moreover, some forms of non-canonical activation of Notch do not
require the cleavage of Notch receptor. In another form, other nuclear or
cytoplasmic factors are recruited instead of CSL protein [48]. One of the
examples of the non-canonical pathway of Notch is the association be-
tween IKKa and Notch1 to mediate an NF-kB dependent transcription
which is active in cervical cancer [49].
There are four types of Notch receptors including Notch1 to 4.
Notch3 and 4 are shorter in their extracellular domain than Notch1 and
2. Moreover, they don't have the intracellular transcriptional activating
domain. Among these four Notch receptors, Notch4 is the only receptor
that lacks the cytokine response region [50]. Initially, the function of the
Notch pathway in cancer was dened through a study of chromosomal
translocation in T-cell lymphoblastic leukemia. Studies have shown that
a Drosophila Notch-like gene exists in the human genome and its rear-
ranged form may be involved in tumor pathogenesis [51]. After
Fig. 2. Notch signaling pathway activation, function, and degradation.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1155
identifying the Notch pathway as an oncogenic factor, a large number of
studies target this signaling pathway as a therapeutic approach such as
γ-secretase suppressors and monoclonal antibodies that target Notch
ligands or receptors, leading to Notch inhibition [48]. Designing a so-
phisticated approach for Notch-based cancer therapy requires more
complex and precise investigations of this signaling pathway [45]. The
outcomes of the Notch pathway are controlled by various types of reg-
ulations. For example, the expression pattern of receptors, ligands, and
modifying enzymes is one of the common strategies in their regulation.
Interestingly, a cis-inhibition mechanism exerted by ligands and re-
ceptors of the same cells could regulate this pathway [52].
Notch receptors are involved in a different types of cancers by
regulating vital cellular processes such as apoptosis, drug resistance,
invasion, epithelial-mesenchymal transition (EMT), metastasis, and
promoting tumor vasculature [45] (Table 1). The data in the
Table provided indicates that the Notch function is context-dependent
and is an oncogene or tumor suppressor based on the cancer type or
even a different cancer cell line of the same cancer type.
4. Regulation of Notch signaling pathway by miRNAs in cancer
4.1. Cancer progression
Accumulating research conrms the critical function of the Notch
signaling pathway in cancer cell proliferation [98103]. It is now known
that miRNAs and Notch signaling either work together or in opposite
ways to shape cellular activities. Understanding the molecular cues and
mechanisms between miRNAs and Notch signaling and their impact on
cancer biology can pave the way for the development of promising
therapeutic strategies. Here, we provide the current knowledge
regarding the interaction between miRNAs and Notch signaling pathway
in the context of different cancers. As mentioned previously, miRNAs
can display either oncogenic or tumor suppressor functions. MiRNA-34
is a tumor suppressor miRNA with a decreased expression in ovarian
cancer. MiRNA-34 is capable of inducing tumor cell apoptosis and
autophagy to decrease tumor cell proliferation and in this case, miRNA-
34 increases the expression level of Bax, as an apoptosis-associated
factor, and LC3 II/p62 as an autophagy-related protein. In one study,
it was shown that miRNA-34 induces ovarian cancer cell death by tar-
geting Notch1 and suppressing it [104]. In contrast, miRNA-223 is a
tumor-promoting agent which positively affects Notch signaling
pathway in colorectal cancer. For this, miRNA-223 inhibits FBXW7 to
increase tumor cell proliferation. FBXW7, in turn, is a tumor suppressor
factor with critical roles in cancer including the regulation of prolifer-
ation, differentiation, and cell division. Reversely affecting FBXW7,
miRNA-223 binds to the 3UTR region of FBXW7 and decreases its
expression levels to increase Notch signaling function. Ultimately, the
Notch pathway increases tumor cell proliferation [105].
Various other target genes have been identied to affect Notch
signaling such as Hairy enhance of split (HES), NF-kB, Hairy/enhancer
of split related with YPRW motif (Hey), vascular growth factor receptor
(VEGF), CYCLIND1, Akt, p21, mammalian target of rapamycin (mTOR),
p27, and c-Myc [19]. HES family including HES1, -5, and -7 subtypes are
primary targets of Notch that act as a transcriptional repressor and
negatively regulate downstream target genes. Potential target genes of
HES1 are CD4, p21, and HES1 itself [106]. A study on miRNA-140-5p
has shown that the expression level of this miRNA is decreased in
gastric cancer. MiRNA-140-5p is a tumor suppressor factor that exerts its
function on the Notch pathway by suppressing Thymus cell antigen 1
(THY1; known as CD90), a tumor-promoting factor with high expression
in gastric cancer. By activation of Notch signaling, its downstream target
genes HES1 and HES5 are increased which subsequently regulate their
targets. HES1 and HES5 prevent apoptosis by upregulation of Bcl-2 and
downregulation of Bax and caspase-3. Therefore, miRNA-140-p5 nega-
tively affects the Notch pathway to inhibit gastric cancer cell progres-
sion through induction of apoptosis [107]. Similarly, a study on miRNA-
Table 1
The function of different types of Notch in cancer.
Notch
receptor
Cancer type Function Ref.
Notch1 Breast cancer Invasion, poor overall survival [53]
Gastric carcinoma Tumor aggressiveness, poor
survival rate, invasion,
migration
[54]
Lung cancer Tumor suppressor (inducing cell
cycle arrest, apoptosis, and
suppressing proliferation, cell
growth, EMT, metastasis)
[5558]
Oncogene (apoptosis inhibition,
pro-survival effects under
hypoxia, proliferation, poor
survival, colony formation, EMT,
metastasis)
[5963]
T cell acute
lymphoblastic
leukemia/lymphoma
Growth, survival [64]
Prostate cancer Growth, proliferation, invasion,
tumor sphere formation,
metastasis
[65]
Colorectal cancer Invasion,
tumornodemetastasis, lymph
node metastases, poor overall
survival
[66]
Head and neck
squamous cell
carcinomas
Both tumor inhibitor and
oncogene
[67]
Ovarian cancer Sphere formation, chemo-
resistance, increasing expression
of cancer stem cells-associated
genes
[68]
Tongue cancer Tumor stage, differentiation,
proliferation, invasion,
metastasis, apoptosis inhibition
[69]
Notch2 Breast cancer Cellular dormancy,
hematopoietic stem cell
mimicry, mobilization in bone
[70]
Gastric cancer Proliferation, growth, colony
formation, invasion, migration
[71]
Colorectal cancer Tumor suppressor [72]
Bladder cancer Tumor growth, EMT, metastasis,
cell-cycle progression, stemness
[73]
Liver cancer stem cells Stemness, clinical severity and
prognosis, self-renewal
[74]
Pancreatic
intraepithelial
neoplasia/pancreatic
ductal
adenocarcinoma
Tumor progression, prolongs
survival, malignant
transformation
[75]
Medullary thyroid
cancer
Tumor suppressor, induction of
apoptosis
[76]
Bladder cancer Proliferation, invasion,
metastasis
[77]
Tongue squamous cell
carcinoma
Proliferation, invasion,
migration
[78]
Esophageal carcinoma Proliferation, migration,
tumorigenicity
[79]
Notch3 Colorectal cancer Tumor formation, proliferation,
proliferation, clonogenic
capacity, aggressive phenotype
[80]
Breast cancer Self-renewal, invasion,
metastatic growth, poor
outcome, proliferation
[81,82]
Ovarian high-grade
serous carcinoma
Recurrent post-chemotherapy,
reduced overall survival, worse
clinical outcome, shortened
progression-free survival,
carboplatin resistance
[83]
Lung cancer Both tumor suppressor and
oncogene
[84]
Gastric cancer Good prognostic biomarker, low
inltration of CD8+T cells, high
inltration of
immunosuppressive cells
[85]
(continued on next page)
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1156
340 has conrmed the anti-cancer function of this miRNA inducing
apoptosis and suppressing proliferation in osteosarcoma cancer cells. To
function, miRNA-340 inhibits b-catenin (cadherin-associated protein) 1
(CTNNB1). Crucial for cell-to-cell junctions and regulation of gene
transcriptions, CTNNB1 is necessary for the function of Wnt signaling
pathway. As Wnt signaling is connected to a wide range of cellular
pathways, dysregulation of CTNNB1 is relevant to a variety of disorders,
especially cancer metastasis. In this study, the authors have shown that
miRNA-340 suppresses CTNNB1 to decrease its targets including HES1
protein of Notch signaling. Overall, miRNA-340 induces apoptosis by
upregulating pro-apoptotic factors (Bim and Bax) and antiapoptotic
proteins (Bcl-2) downregulation while inhibiting proliferation through
suppressing Notch via inactivation of CTNNB1 [108].
In addition to proliferation, Notch signaling is also involved in cancer
invasion and metastasis. Activation of EMT in cancer cells is crucial for
their survival, invasion, and metastasis to other organs. Certain molec-
ular mechanisms are required to induce EMT. During EMT, E-cadherin is
decreased while N-cadherin and vimentin are increased in expression.
Furthermore, other factors include the extracellular matrix (e.g., TGF-b
and Wnt) as well as paracrine signals that mediate the activation of EMT-
inducing transcription factors including zinc-nger E-box (ZEB) proteins
1 and 2, Snail, Slug, and Twist. These factors are responsible for the
induction of EMT, and subsequent invasion and metastasis. During EMT,
cells lose their cell-to-cell adhesion, leading to entry into the blood-
stream through blood vessels [109112]. Based on conrmed evidence,
Notch is one of the critical regulators of EMT. For instance, NICD targets
snail1 via directly interacting with its promoter region and increasing
EMT. Moreover, Notch affects various signaling pathways and factors (e.
g., NF-kB and integrins) to increase EMT and to incur metastasis and
invasion [113].
MiRNA-873-5p is a tumor suppressor factor with a down-regulated
expression in cervical cancer. MiRNA-873-5p suppresses cancer cell
proliferation and induces apoptosis. Moreover, one study demonstrates
that ZEB1 is one of the target genes of miRNA-873-5p. Indeed, miRNA-
873-5p binds to the 3-UTR region of ZEB1 to suppress its activities
including invasion and metastasis. Furthermore, one of the ZEB1 targets
is Notch signaling proteins such as Jagged1, Hey1, and Maml2, and
ZEB1 which increase invasion and metastasis by activating the Notch
pathway. Overall, miRNA-873-5p exerts an inhibitory effect on metas-
tasis and invasion of ovarian cancer cells by suppressing ZEB1, leading
to a subsequent inhibition of the Notch pathway [114]. Another
example of targeting EMT by Notch results in increasing metastasis as
studied by Li et al. They show that the tumor suppressor activity of
miRNA-139-5p on glioma via targeting Notch1 signaling. Notch is sup-
pressed via miRNA-139-5p which ultimately leads to the depletion of
Snail to enhance EMT, invasion, and metastasis. Besides, the activity of
miRNA-139-5p decreases the expression of N-cadherin, vimentin, and
bronectin, while increasing E-cadherin [115].
Similarly, another study on the role of miRNA-424-3p was done by
Chen et al. using pituitary adenoma cells. They have demonstrated that
overexpression of miRNA-424-3p inhibits EMT, proliferation, and
metastasis via negatively targeting Jagged1 and subsequently
decreasing matrix metalloproteinase2 (MMP2) and vimentin [116].
MMPs are a group of zinc-dependent proteolytic metalloenzyme pro-
teins that degrade the extracellular matrix during physiological (such as
wound healing) and pathological situations (such as cancer). Based on
their substrate specicity, MMPs are classied into different groups
including matrilysins, collagenases, stromelysins, and membrane-type
MMPs [117119]. The ability of Notch in targeting MMPs is demon-
strated in previous studies [120122]. In vitro, miRNA-130b-3p sup-
presses DLL1, MMP9/13, and VEGF in order to hamper invasion and
metastasis in breast cancer [123].
An interesting study carried out by Kawasaki et al. on laryngeal
cancer patients with or without nodal metastasis, reveals that miRNA-
449a has a low expression level in nodal metastasis conditions
compared to the group without nodal metastasis. They also found that
aberrant expression of miRNA-449a is associated with decreased inva-
sion, metastasis, and proliferation via targeting Notch1/2. Moreover,
they concluded that miRNA-449a could be a promising diagnostic
biomarker and a novel therapeutic target for laryngeal cancer patients
with nodal metastasis [124]. Furthermore, miRNA-34a reversely targets
Notch1 and its ligand, Jagged1, to suppress colon cancer cell growth,
invasion, and metastasis. By targeting Notch1, miRNA-34a down-regu-
lates vimentin and bronectin in cancer cells [125]. Another experiment
shows that the overexpression of miRNA-1179 leads to the inhibition of
Notch1/4 and HES1, the Notch downstream target, thereby suppressing
proliferation, invasion, and metastasis. Besides, the study suggests that a
low level of miRNA-1179 is associated with lymph node metastasis, poor
survival rate, and advanced clinical stage [126]. On the contrary,
miRNA-1275 exerts oncogenic roles in lung adenocarcinoma. To exert
its carcinogenic functions, miRNA-1275 activates Wnt/b-catenin and
Notch signaling pathways and promotes stemness, recurrence, and
metastasis. Moreover, HIF-1a was identied as the inducer of miRNA-
1275 [127131]. Intriguingly, miRNAs that target the Notch pathway
that increases metastasis of cancer cells could be introduced to these
cells via nanocarriers. Besides, triple-negative breast cancer cells express
high levels of Notch1 on their surface and using Notch1-antibody for
delivering anti-cancer miRNAs has revealed promising results. In an
effort, it was demonstrated that poly(lactic-co-glycolic acid) nano-
particles containing miRNA-34a which were modied with Notch 1
antibodies could signicantly inhibit breast tumor cells migration and
proliferation and induce cell senescence [132].
Another hallmark of cancer crucial for tumor cell survival and spread
is angiogenesis. Accumulating evidence conrms the importance of the
Table 1 (continued )
Notch
receptor
Cancer type Function Ref.
(regulatory T cells and M2
polarized macrophages),
increasing the expression level of
immune checkpoint inhibitors,
immune tolerance
Prostate cancer Proliferation, colony formation [86]
Pancreatic cancer Gemcitabine resistance, caspase-
induced apoptosis inhibition
[87]
Notch4 Endometrial cancer Tumor suppressor [88]
Breast cancer Proliferation, invasiveness,
increasing tumor volume, lymph
node metastasis, advanced
tumor metastasis stage, cancer
recurrence, aggressive
clinicopathological and
biological phenotypes, poor
prognosis
[89,90]
Prostate cancer Tumor cell viability,
proliferation, growth, apoptosis
inhibition, cell metastasis,
invasion
[91]
Gastric cancer Tumor cells growth,
proliferation
[92]
Colorectal cancer Lympho-vascular invasion, poor
overall survival, prognostic
biomarker
[93]
Head and neck
squamous cell
carcinoma
Proliferation, cisplatin
resistance, inhibition of
apoptosis, and cell-cycle
dysregulation, EMT, invasion,
metastasis
[94]
Pancreatic cancer Viability, migration, invasion,
docetaxel resistance
[95]
Salivary adenoid cystic
carcinoma
Invasion, metastasis, recurrence [96]
Melanoma Tumor suppressor,
mesenchymal-epithelial
transition, inhibition of invasion
and metastasis
[97]
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1157
regulatory role of Notch in physiological and pathological angiogenesis
[133135]. Moreover, a dysfunctional form of Jagged1 is seen in several
vascular anomalies, including embryonic lethal and embryonic hemor-
rhage. Further, the lack of Hey1/2 resulted in small vessels with no more
development. Various studies have indicated that VEGF induces the
Notch pathway by increasing angiogenesis via activation of other
pathways such as PI3K/Akt. Interestingly, activation of the Notch
pathway could also suppress angiogenesis. For instance, Notch4 has
been reported to attenuate angiogenesis via suppressing VEGF. Thus,
both pro-angiogenic and anti-angiogenic activities have been reported
for the Notch pathway [136139].
MiRNAs are involved in the angiogenesis process by targeting Notch
signaling. As an example, miRNA-34a is involved in the differentiation
of CD133+glioma stem cells into vascular endothelial cells via sup-
pressing the Notch pathway [140]. Another role of miRNA has been
suggested in retinal angiogenesis by targeting the Notch1 pathway
[141]. However, despite the critical role of Notch signaling in tumor
angiogenesis, several studies point to the regulatory role of this pathway
in association with miRNAs to affect tumor angiogenesis. Jeon et al.
shows an interaction between glioma initiating cells and endothelial
cells leading to cancer progression. They have shown that activation of
nitric oxide synthase via platelet-derived growth factor (PDGF) in both
cells leads to overexpression of nitric oxide-dependent inhibitor of
differentiation 4 (ID4). Upregulation of ID4 mediates suppression of
miRNA-129 to increase the activation of the Jagged1-Notch pathway
and HES1. Activation of the Notch pathway ultimately promotes glioma-
initiating cells' self-renewal and tumor angiogenesis. These results are
important as anti-cancer rationale therapies that target the Notch
pathway (Fig. 3) [142].
4.2. Therapy response
A major challenge for cancer therapy, chemo-resistance is also
responsible for the failure of anticancer drugs in suppressing tumor
progression. Different mechanisms are involved in chemo-resistance
including overexpression of genes responsible for increasing drug
resistance (such as multidrug resistance gene (MDR1)), mitochondrial
alteration, drug efux transporter pumps (such as P-gp), autophagy, as
well as cellular signaling pathways and factors (such as growth factors,
NF-κB, JAK/STAT3, PI3K/Akt/mTOR, and MAPK/ERK). Besides, DNA
repair mechanisms are also involved in drug resistance. There are
different mechanisms by which DNA undergoes repair following damage
including nucleotide and base excision repair, homologous recombina-
tion, and mismatch repair. Furthermore, the role of tumor microenvi-
ronment is also important in developing chemo-resistance. For instance,
hypoxia, pH, and paracrine signals can affect response of tumor cells to
Fig. 3. The role of miRNAs in regulating cancer progression via targeting the Notch signaling pathway.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1158
therapy [143147]. Various studies have revealed the critical function
of Notch signaling in regulating chemo-resistance [148150]. Islam
et al. investigates chemo-resistance in head and neck squamous cell
carcinoma and shows that the mutated form of Keap1 is key in devel-
oping chemo-radio resistance in cancer which results from the activation
of Notch signaling pathway. Keap1 suppresses Nrf2 function and nuclear
translocation. Inhibition of Keap1 resulted in the activation of Nrf2, its
translocation into nucleus, and subsequently overexpression of its
downstream target genes. This study shows that activation of Nrf2
following mutation in Keap1, leads to overexpression of its downstream
target, Notch, which causes cellular metabolic reprogramming that ac-
celerates clonal expansion. This further leads to gaining self-renewal
abilities that induce signaling pathways responsible for therapy resis-
tance [151]. Congruent with this study which addressed the role of
Notch receptor in chemo-resistance, another research demonstrates that
Notch ligands are also involved. Kumar et al. has shown that DLL1
ligand overexpression is associated with NF-κB activation followed by
increased chemo-resistance [152]. A review by Kumar et al. discusses
the crosstalk between Notch and two pathways of Hedgehog and Wnt.
HES1, Jagged1, and other components of Notch signaling pathway can
regulate Hedgehog in mediating chemo-resistance via targeting Gli
factors. However, Wnt signaling could be suppressed by Notch [153]. It
is accepted that miRNAs also play a role in the regulation of key cellular
processes such as Notch signaling to confer chemo-resistance. For
instance, overexpression of miRNA-628-5p is associated with Jagged1
suppression in prostate cancer. For this, miRNA-628-5p directly binds to
the 3UTR region of Jagged1 and suppresses Notch, Snail and Slug to
decrease EMT. Moreover, miRNA-628-5p activity ultimately results in
the inhibition of chemo-resistance in cancer cells under enzalutamide
and docetaxel's pressure [154]. In vitro, Notch1 suppresses miRNA-451
via the AP-1 transcription factor. MDR-1 is the downstream target of
miRNA-451 by which miRNA-451 increases the sensitivity of lung
adenocarcinoma cells under docetaxel therapy [155]. Further, the
impact of miRNA-34 in cancer chemo-resistance occurs through tar-
geting Notch signaling pathway as shown in several studies [156159].
MiRNA-34a is a tumor suppressor and is downregulated in breast cancer
cells after treatment with mutated form of p53 or p53 RNAi. MiRNA-34a
suppresses Notch1 to sensitize breast cancer cells to doxorubicin.
Additionally, miRNA-34a overexpression is associated with decreased
cancer stemness and a small tumor size [129]. Another study on miRNA-
34a demonstrates that its ectopic overexpression leads to sensitivity of
breast cancer cells to Adriamycin via targeting the 3UTR region of
Notch1 [160]. Furthermore, miRNA-34a overexpression suppresses
migration and invasion of breast cancer cells via Snail inhibition, but not
their growth. The administration of doxorubicin along with miRNA-34a
notably induces apoptosis and leads to the reduction of proliferation in
cancer cells. Besides, inhibition of Snail following miRNA-34a over-
expression results in an inhibition of Notch/NF-κB pathway as well as
the RAS/RAF/MEK/ERK axis which subsequently leads to drug resis-
tance suppression [161].
Likewise, miRNA-195-5p is downregulated in colorectal cancer cells.
Upregulation of miRNA-195-5p suppresses Notch2 and RBPJ to increase
sensitivity of cancer cells to 5-uorouracil. Besides, stem-like capacity of
cells, stemness, and tumor sphere formation is decreased while
apoptosis is induced following the miRNA-195-5p overexpression [162].
Another study shows that increased expression of miRNA-199b-5p
negatively regulate Jagged1 expression in ovarian cancer in order to
increase cisplatin-induced cytotoxicity [163]. Additionally, miRNA-136
targets Notch3 reversely and mediates the reduction of cell viability,
tumor cells proliferation, angiogenesis, spheroid formation of cancer
stem cells, and induction of apoptosis in paclitaxel resistant ovarian
cancer cells. In fact, miRNA-136 decreases the resistance of cancer cells
to paclitaxel therapy by reducing the expression levels of survivin, S6,
BCL2, BCL-Xl, Cyclin D1, NF-κB, while increasing that of Bid, Bax, and
Bim proteins [164]. In hepatocellular carcinoma, miRNA-760 over-
expression suppresses doxorubicin resistance via targeting Notch1/
HES1 pathway. Besides, upregulation of miRNA-760 augments PTEN
overexpression and decreases Akt phosphorylation [165].
In addition to chemo-resistance, nding a solution for increasing the
sensitivity of cancer cells to radiotherapy is important. Radiation ther-
apy leads to induction of DNA damage either directly or indirectly via
increasing free radicals. Various studies have shown the key role of
Notch signaling in regulation of radio-resistance, as Notch has critical
crosstalk with oncogenic cellular signaling pathways. Besides, other
studies show that some cancers have increased levels of Jagged1 on the
surface and Notch1 in the culture media following irradiation
[166172]. Similarly, miRNAs also play key roles in regulating the
response of cancer cells to radiation via targeting the Notch pathway.
For instance miRNA-124-3p suppresses Notch related proteins to pro-
mote radiation-induced apoptosis in nasopharyngeal carcinoma.
Furthermore lncRNA PTPRG-AS1 acts as an inhibitor of miRNA-124-3p
in order to increase radio-sensitivity [173]. MiRNA-153 has been shown
to sensitize pancreatic cancer cells to radiation therapy via targeting
Jagged1 [174]. Interestingly, in a study by Yang et al., miRNA-3163
negatively targets ADAM17, a factor responsible for activation of
Notch pathway, to mediate therapy resistance. MiRNA-3163 suppresses
Notch signaling pathway via targeting ADAM17, and promotes the
sensitivity of hepatocellular carcinoma cells to targeted agents (Fig. 4)
[175].
5. Regulation of Notch signaling pathway by lncRNAs in cancer
5.1. Cancer progression
In contrast to miRNAs, numerous studies have associated lncRNAs
with Notch signaling pathway and tumor progression. For instance, the
results of a study have revealed that lncRNA MEG3 is overexpressed in
breast cancer and enhances tumor progression. DNA methyltransferase-
1 suppresses MEG3 expression via inhibiting the hypermethylation of
the MEG3 promoter, thereby decreasing tumor proliferation, and
inducing apoptosis. Besides, Notch1 is the downstream target of MEG3
to induce EMT [176]. LncRNA ILF3-AS1 is another oncogenic factor
highly expressed in hepatocellular carcinoma. ILF3-AS1 increases can-
cer cell viability, EMT, invasion, and migration. For this, ILF3-AS1
competes with Meis Homeobox 2 (MEIS2) to suppress miRNA-628-5p
and increase Notch signaling pathway activities [177]. Another
example is lncRNA PTTG3P which suppresses miRNA-142-5p to pro-
mote Jagged1 overexpression, followed by Notch1 activation. Down-
stream targets of Notch including vimentin, c-Myc, Snail, Twist, and
Cyclin D1 are increased, which result in increased EMT, aggressiveness,
proliferation, and migration of tongue cancer cells [178]. LncRNA
LINC01806 attenuates miRNA-4428 expression to promote Notch2
signaling pathway and subsequently accelerate non-small cells of lung
cancer progression [179]. In contrast to these studies, He et al. has
shown that lncRNA DLEU2 enhances cervical tumor cells proliferation
and progression of cell cycle via inhibiting Notch signaling. to do this,
lncRNA DLEU2 suppresses p53 expression via interacting with ZFP36
Ring Finger Protein (ZFP36) [180]. However, another study shows that
lncRNA DLEU2 induces Notch signaling and increase tumor progression.
The authors of this study have demonstrated that lncRNA DLEU2
expression increases by STAT1, which in turn suppresses miRNA-23b-3p
to increase Notch2, thereby increasing gastric cancer cells proliferation,
invasion, and migration [181].
Furthermore, lncRNA MIR99AHG is overexpressed in pancreatic
cancers mediated by transcription factor forkhead box A1 (FOXA1).
Upregulation of this lncRNA results in sequestration of miRNA-3129-5p
and subsequently ELAV-like RNA binding protein 1 (ELAVL1), followed
by activation of the Notch2 signaling pathway. Activation of Notch2
ultimately results in tumor cell proliferation, migration, and invasion
[182]. In melanoma, lncRNA BASP1-AS1 expression increase is associ-
ated with tumor progression, proliferation, invasion, and migration.
LncRNA BASP1-AS1 up-regulates Notch3 expression via interacting with
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1159
YBX1. Activation of Notch3 results in an increase in downstream targets
including c-Myc, CDK4, and PCNA [183]. A group of lncRNAs called
Small nucleolar RNA host genes (SNHGs) host genes of small nucleolar
RNAs. Accumulating evidence has revealed that these lncRNAs are
involved in various disorders such as cancer [184188]. In a study on
pancreatic cancer cells, it was found that co-culturing of mesenchymal
stem cells and pancreatic cancer cells would increase lncRNA SNHG7 in
tumor cells. Upregulation of lncRNA SNHG7 is further associated with
tumor cell stemness and resistance against Folrinox by targeting the
Notch1/Jagged1/HES1 pathway [189]. SNHG7 could also affect tumor
progression via the miRNA/Notch pathway. For instance, lncRNA
SNHG7 has been proven to sponge miRNA-34a to increase Notch1 and
promote breast cancer progression [190]. As mentioned before, the
progression of tumor cells is increased in hypoxic conditions. In a study,
the authors elucidated that hypoxia could induce the expression of
lncRNAHILAR to promote renal cancer cell invasion and metastasis.
Renal cancer cells release exosomes in response to a hypoxic microen-
vironment that contains lncRNAHILAR. This lncRNA acts as competing
endogenous RNA (ceRNA) to suppress miRNA-613/206/1-1-3p, and
then increases the Jagged1/Notch pathway, followed by activation of C-
X-C motif chemokine receptor 4 (CXCR4). This pathway activity sub-
sequently increases invasive and metastatic capacity of renal cancer cells
[191]. LncRNAs also mediate the response of cancer cells to the immune
system via targeting the Notch pathway. For instance, LINC01355
overexpression is associated with oral squamous cell carcinoma cell
proliferation, invasion, and metastasis. LINC01355 suppresses CD8+T
cell immune response and increases their apoptosis to exert its function
and promote tumor cell progression. For these purposes, INC01355
augments Notch signaling pathway via increasing Notch1, Jagged1, and
HES1 [192]. In the case of angiogenesis, in a study, it was found that
exosomes derived from Jagged1 overexpressed triple-negative breast
cancer cells contained lncRNA MALAT1 which induces angiogenesis in
human vascular endothelial cells via targeting miRNA-140-5p and
subsequently activating the VEGFA pathway [193]. Thus, lncRNAs play
a critical role in cancer progression via targeting the Notch signaling
pathway. Table 2 represents other lncRNAs with proven regulatory
functions on the Notch pathway in cancer.
5.2. Therapy response
As mentioned before, various mechanisms are involved in resistance
to therapy by cancer cells and the Notch signaling pathway could
regulate these mechanisms. For instance, lncRNA SNHG15 suppresses
miRNA-451 to increase MDR1 promotion to mediate resistance to ge-
tinib in lung adenocarcinoma cells [222]. Similarly, lncRNA SNHG7 is
involved in cancer chemoresistance. A study shows the upregulation of
SNGH7 in breast cancer. SNHG7 negatively regulates the expression of
miRNA-34a to increase the percentage of CD44+/CD24- cells, stemness
(via targeting Oct4, SOX2, and Nanog), and sphere formation. Besides,
this interaction between lncRNA SNHG7 and miRNA-34a results in a
higher sensitivity response to chemo-resistance in breast cancer cells
[223]. It has been reported that Notch1 expression is increased in
cisplatin-resistant gastric cancer cells. Notch1 promotes the expression
of lncRNA AK022798 to increase multidrug resistance-associated pro-
tein 1 (MRP1) and P-gp while decreasing caspase 3/8 to suppress
apoptosis. Overall, Notch1 increases cisplatin resistance via targeting
lncRNA AK022798 [224]. In another study, it was demonstrated that
lncRNA DANCR acts as ceRNA for miRNA-34a-5p to increase Jagged1.
Fig. 4. The role of miRNAs in regulating therapy response via targeting the Notch signaling pathway.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1160
Table 2
The function of different lncRNAs in cancer via targeting Notch.
lncRNA Cancer type Signaling
network
Remark Ref
SNHG11 Lung
adenocarcinoma
miRNA-
193a-5p/
Notch3
SNHG11
overexpression
suppresses miRNA-
193a-5p to increase
Notch3 in promoting
tumor growth,
proliferation and
migration.
[194]
SNHG3 Ovarian cancer miRNA-
139-5p/
Notch1
SNHG3 upregulation
resulted in miRNA-
139-5p suppression,
followed by
activation of Notch1
pathway in increasing
proliferation and
migration.
[195]
SNHG3 Breast cancer miRNA-
154-3p/
Notch2
SNHG3 sponges
miRNA-154-3p to
increase Notch2
activation and
subsequently
promoting tumor
cells proliferation,
viability, invasion
and migration.
[196]
SNHG22 Gastric Cancer EZH2
miRNA-
200c-3p/
Notch1
ELK4 increases the
expression of
SNHG22.
SNHG22 targets
EZH2 to inhibit onco-
suppressor genes
expression.
For increasing the
expression level of
Notch1, SNHG22
sponges miRNA-
200c-3p, thereby
increasing
proliferation and
invasion.
Overexpression of
SNHG22 is associated
with poor prognosis.
[197]
SNHG12 Endometrial
cancer
ZIC2/
SNHG12/
Notch
ZIC2 up regulates
SNHG12 and
subsequently Notch
signaling pathway in
increasing tumor cells
proliferation,
migration, and
invasion.
[198]
SNHG12 Nasopharyngeal
carcinoma
Notch1 Increased level of
SNHG12 is associated
with clinical stage,
grade and poor
prognosis.
SNHG12 increases
proliferation,
migration and
invasion of cancer
cells via activating
Notch1 pathway.
[199]
LINC01410 Glioma miRNA-
506-3p/
Notch2
LINC01410
expression is
increased in glioma
cells via Myc.
Overexpressed
LINC01410
suppresses miRNA-
506-3p to increase
Notch2 in promoting
cancer progression.
[177]
SRA Ovarian cancer Notch [200]
Table 2 (continued )
lncRNA Cancer type Signaling
network
Remark Ref
Upregulation of SRA
is associated with
increased tumor
growth, invasion, and
migration via
targeting Notch
pathway.
HOXA-AS2 Cervical cancer RBP-JK/
Notch
HOXA-AS2 increases
Notch signaling
pathway via targeting
RBP-JK to promote
tumor progression.
[201]
HCG11 Pancreatic
carcinoma
miRNA-
579-3p/
MDM2/
Notch/
Hes1
HCG11 suppresses
miRNA-579-3p to
increase MDM2 and
subsequently Notch/
Hes1.
Activation of Notch/
Hes1 resulted in high
aggressiveness and
tumor progression.
[202]
FTX Colorectal cancer miRNA-
214-5p/
Jagged1
FTX suppresses
miRNA-214-5p to
increase Jagged1 and
promote
proliferation,
migration, and
invasion.
[203]
MEG8 Hemangioma miRNA-
203/
Jagged1/
Notch1
MEG8 overexpression
promotes Jagged1/
Notch1 pathway via
inhibition of miRNA-
203 in order to
increase proliferation
and invasion.
[181]
MEG3 Endometrial
carcinoma
Notch1/
HES1
MEG3 overexpression
inhibited tumor cells
proliferation and
growth via
suppressing Notch1/
HES1 signaling
pathway.
[204]
HAGLROS Esophageal
cancer
miRNA-
206/
Notch3
HAGLROS promotes
proliferation,
invasion and
metastasis via
targeting miRNA-206
and subsequently
Notch3.
[205]
UCA1 Tongue cancer miRNA-
124/
TGFβ1/
Jagged1/
Notch1
UCA1 is
overexpressed in
tongue cancer and
targets miRNA-124/
TGFβ1/Jagged1/
Notch1 pathway to
increase EMT and
invasion.
UCA1 upregulation is
associated with
poorer prognosis.
[206]
GHET1 Prostate cancer KLF2/HIF-
1
α
/Notch1
Overexpression of
CHET1 is associated
with increased cell
proliferation via
suppression of KLF2
and subsequently
increased HIF-1
α
/
Notch1axis.
Knockdown of CHET1
is associated with cell
cycle arrest at G0/G1
phase and enhanced
apoptosis.
[207]
FEZF1-AS1 Glioblastoma Upregulation of
FEZF1-AS1 is
[208]
(continued on next page)
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1161
Table 2 (continued )
lncRNA Cancer type Signaling
network
Remark Ref
miRNA-
34a/Notch-
1
associated with poor
survival.
FEZF1-AS1 inhibits
miRNA-34a to
increase Notch1 in
promoting invasion
and migration.
FEZF1-AS1 Non-small cell
lung cancer
miRNA-
34a/
Notch1
FEZF1-AS1
overexpression
suppresses miRNA-
34a to increase
Notch1 function.
Upregulation of
FEZF1-AS1 is
associated with tumor
cells invasion and
metastasis, while
decreased apoptosis.
[209]
PlncRNA-1 Glioma Jagged1/
Notch1/
HES1
PlncRNA-1 upturning
leads to activation of
Jagged1/Notch1/
HES1 axis in
increasing cancer
cells proliferation and
colony formation,
while suppressing
apoptosis.
[210]
XIST Non-small cell
lung cancer
miRNA-
137/
Notch1/
TGF-β1
XIST sponges miRNA-
137 to promote TGF-
β1 activity in
increasing EMT.
MiRNA-137
suppresses Notch1
pathway.
[211]
LET Non-small cell
lung cancer
NICD1 LET expression is
down regulated in
cancer cells.
Downregulation of
LET is associated with
advanced tumor
stages and poor
overall survival.
Overexpression of
LET leads to
suppression of
proliferation,
invasion, and
metastasis, while
enhance cell cycle
arrest and apoptosis
via suppressing
NICD1.
[212]
HNF1A-
AS1
Oral squamous
cell carcinoma
STAT3/
HNF1A-
AS1/
Notch1/
HES1
As an upstream
mediator, STAT3
increase the
expression of HNF1A-
AS1.
Upregulation of
HNF1A-AS1 resulted
in activation of
Notch1/HES1
pathway in increasing
cancer cells
proliferation,
migration and EMT.
[213]
FAM83H-
AS1
Colorectal
carcinoma
Jagged1/
Notch1
Upregulation of
FAM83H-AS1 is
associated with
clinical features and
poor prognosis.
FAM83H-AS1
increased cell
proliferation and
migration, while
suppressing apoptosis
[214]
Table 2 (continued )
lncRNA Cancer type Signaling
network
Remark Ref
via activation of
Jagged1/Notch1.
HOTAIR Cervical cancer Notch/Wnt HOTAIR
overexpression is
associated with
lympho-vascular
invasion, tumor size,
and lymph node
metastasis.
HOTAIR
overexpression leads
to enhanced tumor
growth, proliferation,
EMT, and invasion via
targeting Notch-Wnt
signaling pathway.
[215]
HOTAIR Pancreatic cancer miRNA-
613/
Notch3
HOTAIR
overexpression
suppresses miRNA-
613 to increase
Notch3 pathway.
Upregulation of
HOTAIR is associated
with tumor
differentiation,
advanced TNM stage,
nodal metastasis,
poor survival,
proliferation,
invasion and
migration.
Downregulation of
HOTAIR resulted in
tumor cells apoptosis
and cell cycle arrest at
G0/G1 phase.
[216]
ZFAS1 Glioma Notch Overexpression of
ZFAS1 is associated
with advanced tumor
stage and poor
survival rate.
ZFAS1 promotes
cancer cells
proliferation, EMT,
invasion and
migration via
activation of Notch
signaling pathway.
[217]
DSCAM-
AS1
Colorectal cancer miRNA-
137/
Notch1
DSCAM-AS1
suppresses miRNA-
137 to increase
Notch1, thereby
promoting
proliferation,
migration, and EMT.
[218]
MIR22HG Gastric cancer Notch2/
HEY1
MIR22HG is down
regulated in gastric
cancer and its
overexpression leads
to suppression of
Notch2 and HEY1.
Low expression level
of MIR22HG is
associated with poor
overall survival.
Upregulation of
MIR22HG resulted in
suppression of tumor
cells proliferation,
invasion and
migration.
[219]
Linc00514 Breast cancer Jagged1/
Notch/IL4
and IL-6
Linc00514 is
overexpressed in
breast cancer and
promotes Notch
[220]
(continued on next page)
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1162
This axis ultimately leads to docetaxel resistance in prostate cancer cells
[225]. Furthermore, another study has revealed that lncRNA MALAT1
interacts with Notch1 to promote ovarian cancer cells' resistance to
cisplatin. Following the knockdown of lncRNA MALAT1, Bax upregu-
lation and Bcl-2 downregulation occur in response to cisplatin [226].
LINC00152 is another lncRNA that mediates drug resistance via
targeting the Notch signaling pathway. LINC00152 sponges miRNA-
139-5p to increase Notch1 expression level. Activation of Noth1 pro-
motes resistance of colorectal cancer cells to 5-uorouracil [227].
Another long intergenic noncoding RNA named LINC00021 targets
Notch signaling pathway and suppresses p21 via affecting EZH2 to in-
crease chemo-resistance to temozolomide in glioblastoma cells [228]. In
the case of radio sensitivity, few studies address the role of lncRNAs in
regulating resistance/sensitivity of cancer cells to radiation therapy. An
example is lncRNA AGAP2-AS1 which is involved in increasing the
resistance of lung cancer cells to radiation therapy. For this purpose, M2
polarized macrophage-derived exosomes containing AGAP2-AS1 sponge
miRNA-296 to increase Notch2 to enhance radio-resistance [229].
6. Regulation of Notch signaling pathway by other ncRNAs in
cancer progression and therapy
The tRFs are a group of ncRNAs and accumulating research has
revealed their critical roles in cancer. They regulate gene expression,
gene silencing, protein translation, and RNA processing, thereby con-
trolling vital cellular processes such as cell proliferation and differenti-
ation. Moreover, their function in different cancers including breast
cancer, prostate cancer, leukemia, and lung and colorectal cancers is
elucidated [230]. Like miRNAs and lncRNAs, tRFs exert two different
functions of tumor suppression or promotion in cancers. By regulating
the expression of tumorigenic factors, tRFs could enhance cancer cell
proliferation and progression. Different tRF mechanisms exert their
functions in cancer cells such as suppressing the transcription process,
enhancing ribosomal protein's translation, and increasing the function of
mitosis regulators such as aurora kinase A [231]. Additionally, they
could also target invasion and metastasis, and are considered promising
biomarkers for cancer diagnosis and prognosis [232235]. Huang et al.
has a well-performed study to illustrate the role of tRFs in cancer by
targeting the Notch signaling pathway. They evaluate the effect of tRF/
miRNA-1280 on colorectal cancer stem-like cell progression. A 17-bp
fragment, tRF/miRNA-1280, is derived from tRNA
Leu
and has a low
expression in colorectal cancer. As a tumor suppressor factor, tRF/
miRNA-1280 diminishes tumor cell proliferation, colony formation,
and metastasis via negatively regulating JAG2. Moreover, suppression of
Notch by tRF/miRNA-1280 results in cancer stem-like cell phenotypes
inhibition, as well as Gata1/3 and miRNA-200b transcriptional sup-
pression. Suppression of miRNA-200b leads to decreased levels of SUZ12
and ZEB1 [236].
Another class of ncRNAs is circRNAs with critical functions in cancer
[237240]. It is demonstrated that circNFIX has a high expression level
in glioma. CircNFIX inhibits miRNA-34a to increase Notch1 to enhance
tumor cell proliferation and migration. Moreover, suppression of
circNFIX or upregulation of miRNA-34a is associated with cell apoptosis
[241]. In another study, it was shown that hsa_circ_0009910 over-
expression in ovarian cancer cells is associated with miRNA-145
downregulation which increase NF-kB and Notch/HES pathway acti-
vation. Activation of these pathways promotes tumor proliferation and
migration. Moreover, upregulation of hsa_circ_0009910 leads to an un-
favorable prognosis in cancer patients [242].
Another circRNA is circKIF4A (hsa_circ_0007255) which suppresses
miRNA-375/1231 in order to enhance bladder cancer cells progression
via increasing Notch2 [243]. Furthermore, hsa_circ_0058124 is associ-
ated with poor prognosis, increased papillary thyroid cancer cell pro-
liferation, invasion and metastasis via suppressing miRNA-218-5p.
Inhibition of miRNA-218-5p leads to upregulation of NUMB, a strong
inhibitor of Notch, which subsequently suppresses the Notch3/
GATAD2A axis [244].
The results of another experiment have revealed that circAPLP2 at-
tenuates miRNA-103-3p expression to increase colorectal cancer cells
proliferation and metastasis via activation of the Notch signaling
pathway [245]. Additionally, circ-NSD2 suppresses miRNA-199b-5p to
promote colorectal cancer cell metastasis and migration via increasing
Jagged1 and discoidin domain receptor tyrosine kinase 1 (DDR1)
expression [246].
Circular RNA FAT atypical cadherin 1 (circFAT1) expression has
been shown to be increased in breast cancer cells. In one study, it was
founded that overexpression of circFAT1 is associated with oxaliplatin
resistance via sponging miRNA-525-5p to increase spindle and
kinetochore-associated complex subunit 1 (SKA1). This axis ultimately
results in the activation of Notch and Wnt signaling pathways and de-
creases the sensitivity of breast cancer cells to oxaliplatin therapy [247].
7. Conclusion and remarks
Cancer is characterized by uninhibited growth and proliferation of
cells resulting from disrupted mechanisms that highly control cell di-
vision. Numerous studies have focused on various cell signaling path-
ways to bring cancer under control it remains hard to treat.
Conventional cancer therapies include surgeries, chemotherapies, and
radiation therapies. However, resistance to therapeutics by cancer cells
and recurrence is one of the obstacles in cancer therapy and remains to
be solved. Notch is an important signaling pathway that affects vital cell
processes such as proliferation, differentiation, invasion, and metastasis.
Notch is a cell membrane receptor and is activated after binding to its
ligands. Following activation, a part of Notch receptor named NICD is
separated and transferred to the nucleus to regulate its downstream
target genes and its degradation is mediated by proteasomes. Regulation
of Notch signaling is of importance for cancer therapy. The most known
class of ncRNAs is miRNAs are 22 nucleotides long. MiRNAs regulate
gene expression, protein translation, and function. Additionally, they
target various cellular signaling to affect tumorigenesis and response to
therapy by cancer cells. Another class of ncRNAs is lncRNAs with at least
200 nucleotides and critical functions in cells. It is highly accepted that
these ncRNAs are important regulators of cancer, and affect signaling
pathways to induce tumorigenesis or act as a tumor suppressor. One of
the pathways under the control of these ncRNAs is the Notch signaling
pathway. ncRNAs could induce or inhibit this pathway to regulate
cancer hallmarks. For instance, ncRNAs affect Notch and its downstream
targets to regulate apoptosis via mitochondrial pathways. Likewise, they
Table 2 (continued )
lncRNA Cancer type Signaling
network
Remark Ref
signaling pathway to
increase IL-4 and IL-6.
Overexpression of
Linc00514
accelerates tumor
proliferation,
invasion, increasing
the number of
macrophages
expressing M2
markers CD206 and
CD163, tumor size,
and pulmonary
metastatic nodules
NALT Acute
lymphoblastic
leukemia
Notch1 NALT overexpression
increased tumor cells
proliferation and
growth via activation
of Notch1 signaling
pathway through cis-
regulation.
[221]
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1163
regulate metastasis and invasion by affecting EMT-TFs, cadherins, and
MMPs. In addition, ncRNAs and Notch are associated with regulation of
tumor angiogenesis, stemness, and cancer recurrence. Intriguingly,
lncRNAs act as ceRNA for miRNA as their upstream mediator to
modulate Notch signaling in cancer. It bears noting that the interaction
between Notch and other ncRNAs such as circRNAs and tRFs are being
investigated. Interestingly, Notch can be an upstream regulator of
ncRNAs to regulate cellular processes. Overall, the relationship between
ncRNAs and the Notch pathway is promising and future studies should
address this issue for overcoming barriers in cancer treatment.
Funding
No funding was received.
CRediT authorship contribution statement
All authors have contributed at least in part in preparing this
manuscript. We conrm that we acknowledge authorship roles.
Declaration of competing interest
The authors declare no conict of interest.
References
[1] R.L. Siegel, K.D. Miller, H.E. Fuchs, A. Jemal, Cancer statistics, 2022, CA: a cancer
journal for clinicians (2022).
[2] F. Wu, J. Yang, J. Liu, Y. Wang, J. Mu, Q. Zeng, et al., Signaling pathways in
cancer-associated broblasts and targeted therapy for cancer, Signal Transduct.
Target. Ther. 6 (1) (2021) 135.
[3] ´
A. Nagy, G. Munk´
acsy, B. Gy˝
orffy, Pancancer survival analysis of cancer hallmark
genes, Sci. Rep. 11 (1) (2021) 110.
[4] G. Monticone, L. Miele, Notch pathway: a journey from notching phenotypes to
cancer immunotherapy, in: Notch Signaling in Embryology and Cancer, 2021,
pp. 201222.
[5] C.K. Jain, S. Bhargava, I. Jain, S. Varshney, Targeting notch pathway in cancer
diagnostics and therapeutics: an emerging approach, Recent patents on anti-
cancerDrug Discov. (2021).
[6] S. Mirzaei, M.D.A. Paskeh, E. Okina, M.H. Gholami, K. Hushmandi, M. Hashemi,
et al., Molecular landscape of LncRNAs in prostate cancer: a focus on pathways
and therapeutic targets for intervention, J. Exp. Clin. Cancer Res. 41 (1) (2022)
214.
[7] S. Mirzaei, M.H. Gholami, K. Hushmandi, F. Hashemi, A. Zabolian, I. Canadas, et
al., The long and short non-coding RNAs modulating EZH2 signaling in cancer,
J. Hematol. Oncol. 15 (1) (2022) 18.
[8] M. Ravo, A. Cordella, A. Rinaldi, G. Bruno, E. Alexandrova, P. Saggese, et al.,
Small non-coding RNA deregulation in endometrial carcinogenesis, Oncotarget 6
(7) (2015) 4677.
[9] J.R. Warner, The economics of ribosome biosynthesis in yeast, Trends Biochem.
Sci. 24 (11) (1999) 437440.
[10] S. Mirzaei, A. Zarrabi, F. Hashemi, A. Zabolian, H. Saleki, A. Ranjbar, et al.,
Regulation of nuclear factor-KappaB (NF-κB) signaling pathway by non-coding
RNAs in cancer: inhibiting or promoting carcinogenesis? Cancer Lett. 509 (2021)
6380.
[11] S. Ashrazaveh, M. Ashrazadeh, A. Zarrabi, K. Husmandi, A. Zabolian,
M. Shahinozzaman, et al., Long non-coding RNAs in the doxorubicin resistance of
cancer cells, Cancer Lett. 508 (2021) 104114.
[12] M. Esteller, Non-coding RNAs in human disease, Nat. Rev. Genet. 12 (12) (2011)
861874.
[13] T.R. Mercer, M.E. Dinger, J.S. Mattick, Long non-coding RNAs: insights into
functions, Nat. Rev. Genet. 10 (3) (2009) 155159.
[14] S. Salimimoghadam, S. Taefehshokr, R. Loveless, Y. Teng, G. Bertoli,
N. Taefehshokr, et al., The role of tumor suppressor short non-coding RNAs on
breast cancer, Crit. Rev. Oncol. Hematol. 158 (2021), 103210.
[15] F.J. Slack, A.M. Chinnaiyan, The role of non-coding RNAs in oncology, Cell 179
(5) (2019) 10331055.
[16] Y. Pekarsky, C.M. Croce, Is miR-29 an oncogene or tumor suppressor in CLL?
Oncotarget 1 (3) (2010) 224.
[17] E. Anastasiadou, A. Faggioni, P. Trivedi, F.J. Slack, The nefarious nexus of
noncoding RNAs in cancer, Int. J. Mol. Sci. 19 (7) (2018) 2072.
[18] D.P. Bartel, Metazoan micrornas, Cell 173 (1) (2018) 2051.
[19] Z. Wang, Y. Li, D. Kong, A. Ahmad, S. Banerjee, F.H. Sarkar, Cross-talk between
miRNA and notch signaling pathways in tumor development and progression,
Cancer Lett. 292 (2) (2010) 141148.
[20] V. Balatti, Y. Pekarsky, C.M. Croce, Role of the tRNA-derived small RNAs in
cancer: new potential biomarkers and target for therapy, Adv. Cancer Res. 135
(2017) 173187.
[21] Y. Hu, A. Cai, J. Xu, W. Feng, A. Wu, R. Liu, et al., An emerging role of the 5
termini of mature tRNAs in human diseases: current situation and prospects,
Biochim. Biophys. Acta, Mol. Basis Dis. 1868 (2) (2022), 166314.
[22] Y. Shen, X. Yu, Y. Ruan, Z. Li, Y. Xie, Z. Yan, et al., Global prole of tRNA-derived
small RNAs in gastric cancer patient plasma and identication of tRF-33-
P4R8YP9LON4VDP as a new tumor suppressor, Int. J. Med. Sci. 18 (7) (2021)
1570.
[23] J.N. Vo, M. Cieslik, Y. Zhang, S. Shukla, L. Xiao, Y. Zhang, et al., The landscape of
circular RNA in cancer, Cell 176 (4) (2019) 869881, e13.
[24] C. Wang, S. Tan, J. Li, W.-R. Liu, Y. Peng, W. Li, CircRNAs in lung cancer-
biogenesis, function and clinical implication, Cancer Lett. 492 (2020) 106115.
[25] J. Greene, A.-M. Baird, L. Brady, M. Lim, S.G. Gray, R. McDermott, et al., Circular
RNAs: biogenesis, function and role in human diseases, Front. Mol. Biosci. 4
(2017) 38.
[26] L.S. Kristensen, M.S. Andersen, L.V. Stagsted, K.K. Ebbesen, T.B. Hansen,
J. Kjems, The biogenesis, biology and characterization of circular RNAs, Nat. Rev.
Genet. 20 (11) (2019) 675691.
[27] L.-L. Chen, The biogenesis and emerging roles of circular RNAs, Nat. Rev. Mol.
Cell Biol. 17 (4) (2016) 205211.
[28] T.B. Hansen, T.I. Jensen, B.H. Clausen, J.B. Bramsen, B. Finsen, C.K. Damgaard, et
al., Natural RNA circles function as efcient microRNA sponges, Nature 495
(7441) (2013) 384388.
[29] K. Abdelmohsen, A.C. Panda, R. Munk, I. Grammatikakis, D.B. Dudekula, S. De, et
al., Identication of HuR target circular RNAs uncovers suppression of PABPN1
translation by CircPABPN1, RNA Biol. 14 (3) (2017) 361369.
[30] W.W. Du, W. Yang, Y. Chen, Z.-K. Wu, F.S. Foster, Z. Yang, et al., Foxo3 circular
RNA promotes cardiac senescence by modulating multiple factors associated with
stress and senescence responses, Eur. Heart J. 38 (18) (2017) 14021412.
[31] W.W. Du, W. Yang, E. Liu, Z. Yang, P. Dhaliwal, B.B. Yang, Foxo3 circular RNA
retards cell cycle progression via forming ternary complexes with p21 and CDK2,
Nucleic Acids Res. 44 (6) (2016) 28462858.
[32] L.M. Holdt, A. Stahringer, K. Sass, G. Pichler, N.A. Kulak, W. Wilfert, et al.,
Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and
atherosclerosis in humans, Nat. Commun. 7 (1) (2016) 114.
[33] Y. Geng, J. Jiang, C. Wu, Function and clinical signicance of circRNAs in solid
tumors, J. Hematol. Oncol. 11 (1) (2018) 120.
[34] M.S. Bartolomei, S. Zemel, S.M. Tilghman, Parental imprinting of the mouse H19
gene, Nature 351 (6322) (1991) 153155.
[35] C.J. Brown, A. Ballabio, J.L. Rupert, R.G. Lafreniere, M. Grompe, R. Tonlorenzi, et
al., A gene from the region of the human X inactivation Centre is expressed
exclusively from the inactive X chromosome, Nature 349 (6304) (1991) 3844.
[36] M.K. Iyer, Y.S. Niknafs, R. Malik, U. Singhal, A. Sahu, Y. Hosono, et al., The
landscape of long noncoding RNAs in the human transcriptome, Nat. Genet. 47
(3) (2015) 199208.
[37] L. Statello, C.-J. Guo, L.-L. Chen, M. Huarte, Gene regulation by long non-coding
RNAs and its biological functions, Nat. Rev. Mol. Cell Biol. 22 (2) (2021) 96118.
[38] M. Entezari, A. Taheriazam, S. Orouei, S. Fallah, A. Sanaei, E.S. Hejazi, et al.,
LncRNA-miRNA axis in tumor progression and therapy response: an emphasis on
molecular interactions and therapeutic interventions, Biomed. Pharmacother.
154 (2022), 113609.
[39] M. Hashemi, M.S. Moosavi, H.M. Abed, M. Dehghani, M. Aalipour, E.A. Heydari,
et al., Long non-coding RNA (lncRNA) H19 in human cancer: from proliferation
and metastasis to therapy, Pharmacol. Res. 184 (2022), 106418.
[40] F. Kopp, J.T. Mendell, Functional classication and experimental dissection of
long noncoding RNAs, Cell 172 (3) (2018) 393407.
[41] S. Geisler, J. Coller, RNA in unexpected places: long non-coding RNA functions in
diverse cellular contexts, Nat. Rev. Mol. Cell Biol. 14 (11) (2013) 699712.
[42] K.A. Wharton, K.M. Johansen, T. Xu, S. Artavanis-Tsakonas, Nucleotide sequence
from the neurogenic locus notch implies a gene product that shares homology
with proteins containing EGF-like repeats, Cell 43 (3) (1985) 567581.
[43] R. Kopan, M.X.G. Ilagan, The canonical notch signaling pathway: unfolding the
activation mechanism, Cell 137 (2) (2009) 216233.
[44] S.J. Bray, Notch signalling: a simple pathway becomes complex, Nat. Rev. Mol.
Cell Biol. 7 (9) (2006) 678689.
[45] S. Majumder, J.S. Crabtree, T.E. Golde, L.M. Minter, B.A. Osborne, L. Miele,
Targeting notch in oncology: the path forward, Nat. Rev. Drug Discov. 20 (2)
(2021) 125144.
[46] I. Espinoza, L. Miele, Notch inhibitors for cancer treatment, Pharmacol. Ther. 139
(2) (2013) 95110.
[47] Y.-Y. Hu, M.-h. Zheng, R. Zhang, Y.-M. Liang, H. Han, Notch signaling pathway
and cancer metastasis, in: Notch Signaling in Embryology and Cancer, 2012,
pp. 186198.
[48] E.R. Andersson, U. Lendahl, Therapeutic modulation of notch signallingare we
there yet? Nat. Rev. Drug Discov. 13 (5) (2014) 357378.
[49] L. Song, Y. Peng, J. Yun, P. Rizzo, V. Chaturvedi, S. Weijzen, et al., Notch-1
associates with IKK
α
and regulates IKK activity in cervical cancer cells, Oncogene
27 (44) (2008) 58335844.
[50] T. Fukusumi, J. Califano, The NOTCH pathway in head and neck squamous cell
carcinoma, J. Dent. Res. 97 (6) (2018) 645653.
[51] L.W. Ellisen, J. Bird, D.C. West, A.L. Soreng, T.C. Reynolds, S.D. Smith, et al.,
TAN-1, the human homolog of the drosophila notch gene, is broken by
chromosomal translocations in T lymphoblastic neoplasms, Cell 66 (4) (1991)
649661.
[52] S.J. Bray, Notch signalling in context, Nat. Rev. Mol. Cell Biol. 17 (11) (2016)
722735.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1164
[53] X. Yuan, M. Zhang, H. Wu, H. Xu, N. Han, Q. Chu, et al., Expression of Notch1
correlates with breast cancer progression and prognosis, PloS one. 10 (6) (2015),
e0131689.
[54] T.-S. Yeh, C.-W. Wu, K.-W. Hsu, W.-J. Liao, M.-C. Yang, A.F.-Y. Li, et al., The
activated Notch1 signal pathway is associated with gastric cancer progression
through cyclooxygenase-2, Cancer Res. 69 (12) (2009) 50395048.
[55] V. Sriuranpong, M.W. Borges, R.K. Ravi, D.R. Arnold, B.D. Nelkin, S.B. Baylin, et
al., Notch signaling induces cell cycle arrest in small cell lung cancer cells, Cancer
Res. 61 (7) (2001) 32003205.
[56] J.-X. Wang, X. Zhang, L. Wang, H. Cheng, G. Yao, Inhibitory effects of Notch1
overexpression on proliferation and neuroendocrine marker expression in a small
cell lung cancer cell line, Zhonghua Zhong Liu Za Zhi 31 (5) (2009) 335339.
[57] H. Wael, R. Yoshida, S. Kudoh, K. Hasegawa, K. Niimori-Kita, T. Ito, Notch1
signaling controls cell proliferation, apoptosis and differentiation in lung
carcinoma, Lung Cancer 85 (2) (2014) 131140.
[58] W.A. Hassan, R. Yoshida, S. Kudoh, K. Hasegawa, K. Niimori-Kita, T. Ito, Notch1
controls cell invasion and metastasis in small cell lung carcinoma cell lines, Lung
Cancer 86 (3) (2014) 304310.
[59] Y. Chen, M.A. De Marco, I. Graziani, A.F. Gazdar, P.R. Strack, L. Miele, et al.,
Oxygen concentration determines the biological effects of NOTCH-1 signaling in
adenocarcinoma of the lung, Cancer Res. 67 (17) (2007) 79547959.
[60] S. Eliasz, S. Liang, Y. Chen, M.A. De Marco, O. Machek, S. Skucha, et al., Notch-1
stimulates survival of lung adenocarcinoma cells during hypoxia by activating the
IGF-1R pathway, Oncogene 29 (17) (2010) 24882498.
[61] A. Baumgart, S. Seidl, P. Vlachou, L. Michel, N. Mitova, N. Schatz, et al., ADAM17
regulates epidermal growth factor receptor expression through the activation of
Notch1 in non-small cell lung CancerADAM17 regulates EGFR expression through
Notch1, Cancer Res. 70 (13) (2010) 53685378.
[62] Y. Yang, Y.-H. Ahn, D.L. Gibbons, Y. Zang, W. Lin, N. Thilaganathan, et al., The
notch ligand Jagged2 promotes lung adenocarcinoma metastasis through a miR-
200dependent pathway in mice, J. Clin. Invest. 121 (4) (2011) 13731385.
[63] L. Guo, T. Zhang, Y. Xiong, Y. Yang, Roles of NOTCH1 as a therapeutic target and
a biomarker for lung cancer: controversies and perspectives, Dis. Markers 2015
(2015).
[64] W.S. Pear, J.C. Aster, T cell acute lymphoblastic leukemia/lymphoma: a human
cancer commonly associated with aberrant NOTCH1 signaling, Curr. Opin.
Hematol. 11 (6) (2004) 426433.
[65] M.A. Rice, E.-C. Hsu, M. Aslan, A. Ghoochani, A. Su, T. Stoyanova, Loss of Notch1
activity inhibits prostate cancer growth and metastasis and sensitizes prostate
cancer cells to antiandrogen TherapiesLoss of Notch1 as a therapeutic strategy for
prostate cancer, Mol. Cancer Ther. 18 (7) (2019) 12301242.
[66] D. Chu, Y. Li, W. Wang, Q. Zhao, J. Li, Y. Lu, et al., High level of Notch1 protein is
associated with poor overall survival in colorectal cancer, Ann. Surg. Oncol. 17
(5) (2010) 13371342.
[67] E.M. Rettig, C.H. Chung, J.A. Bishop, J.D. Howard, R. Sharma, R.J. Li, et al.,
Cleaved NOTCH1 expression pattern in head and neck squamous cell carcinoma
is associated with NOTCH1 mutation, HPV status, and high-risk FeaturesCleaved
NOTCH1 expression in head and neck cancer, Cancer Prev. Res. 8 (4) (2015)
287295.
[68] E.J. Seo, D.K. Kim, I.H. Jang, E.J. Choi, S.H. Shin, S.I. Lee, et al., Hypoxia-
NOTCH1-SOX2 signaling is important for maintaining cancer stem cells in
ovarian cancer, Oncotarget 7 (34) (2016) 55624.
[69] R.-H. Gan, H. Wei, J. Xie, D.-P. Zheng, E.-L. Luo, X.-Y. Huang, et al., Notch1
regulates tongue cancer cells proliferation, apoptosis and invasion, Cell Cycle 17
(2) (2018) 216224.
[70] M. Capulli, D. Hristova, Z. Valbret, K. Carys, R. Arjan, A. Maurizi, et al., Notch2
pathway mediates breast cancer cellular dormancy and mobilisation in bone and
contributes to haematopoietic stem cell mimicry, Br. J. Cancer 121 (2) (2019)
157171.
[71] Y.C. Tseng, Y.H. Tsai, M.J. Tseng, K.W. Hsu, M.C. Yang, K.H. Huang, et al.,
Notch2-induced COX-2 expression enhancing gastric cancer progression, Mol.
Carcinog. 51 (12) (2012) 939951.
[72] D. Chu, J. Zheng, W. Wang, Q. Zhao, Y. Li, J. Li, et al., Notch2 expression is
decreased in colorectal cancer and related to tumor differentiation status, Ann.
Surg. Oncol. 16 (12) (2009) 32593266.
[73] T. Hayashi, K.M. Gust, A.W. Wyatt, A. Goriki, W. J¨
ager, S. Awrey, et al., Not all
NOTCH is created equal: the oncogenic role of NOTCH2 in bladder cancer and its
implications for targeted TherapyTargeting NOTCH2 in bladder cancer, Clin.
Cancer Res. 22 (12) (2016) 29812992.
[74] P. Zhu, Y. Wang, Y. Du, L. He, G. Huang, G. Zhang, et al., C8orf4 negatively
regulates self-renewal of liver cancer stem cells via suppression of NOTCH2
signalling, Nat. Commun. 6 (1) (2015) 113.
[75] P.K. Mazur, H. Einw¨
achter, M. Lee, B. Sipos, H. Nakhai, R. Rad, et al., Notch2 is
required for progression of pancreatic intraepithelial neoplasia and development
of pancreatic ductal adenocarcinoma, Proc. Natl. Acad. Sci. 107 (30) (2010)
1343813443.
[76] M. Truong, M.R. Cook, S.N. Pinchot, M. Kunnimalaiyaan, H. Chen, Resveratrol
induces Notch2-mediated apoptosis and suppression of neuroendocrine markers
in medullary thyroid cancer, Ann. Surg. Oncol. 18 (5) (2011) 15061511.
[77] Z. Zhou, D. Cui, M.H. Sun, J.L. Huang, Z. Deng, B.M. Han, et al., CAFs-derived
MFAP5 promotes bladder cancer malignant behavior through NOTCH2/HEY1
signaling, FASEB J. 34 (6) (2020) 79707988.
[78] R.-H. Gan, L.-S. Lin, D.-P. Zheng, Y. Zhao, L.-C. Ding, D.-L. Zheng, et al., High
expression of Notch2 drives tongue squamous cell carcinoma carcinogenesis,
Experimental Cell Research 399 (1) (2021), 112452.
[79] X.Y. Ding, H.Y. Hu, K.N. Huang, R.Q. Wei, J. Min, C. Qi, et al., Ubiquitination of
NOTCH2 by DTX3 suppresses the proliferation and migration of human
esophageal carcinoma, Cancer Sci. 111 (2) (2020) 489501.
[80] V. Seran, L. Persano, L. Moserle, G. Esposito, M. Ghisi, M. Curtarello, et al.,
Notch3 signalling promotes tumour growth in colorectal cancer, J. Pathol. 224
(4) (2011) 448460.
[81] A.A. Leontovich, M. Jalalirad, J.L. Salisbury, L. Mills, C. Haddox, M. Schroeder, et
al., NOTCH3 expression is linked to breast cancer seeding and distant metastasis,
Breast Cancer Res. 20 (1) (2018) 119.
[82] N. Yamaguchi, T. Oyama, E. Ito, H. Satoh, S. Azuma, M. Hayashi, et al., NOTCH3
signaling pathway plays crucial roles in the proliferation of ErbB2-negative
human breast cancer cells, Cancer Res. 68 (6) (2008) 18811888.
[83] J.T. Park, X. Chen, C.G. Trop`
e, B. Davidson, I.-M. Shih, T.-L. Wang, Notch3
overexpression is related to the recurrence of ovarian cancer and confers
resistance to carboplatin, Am. J. Pathol. 177 (3) (2010) 10871094.
[84] W.A. Hassan, R. Yoshida, S. Kudoh, Y. Motooka, T. Ito, Evaluation of role of
Notch3 signaling pathway in human lung cancer cells, J. Cancer Res. Clin. Oncol.
142 (5) (2016) 981993.
[85] Y. Cui, Q. Li, W. Li, Y. Wang, F. Lv, X. Shi, et al., NOTCH3 is a prognostic factor
and is correlated with immune tolerance in gastric cancer, Front. Oncol. 10
(2021), 574937.
[86] G. Danza, C. Di Serio, M.R. Ambrosio, N. Sturli, G. Lonetto, F. Rosati, et al.,
Notch3 is activated by chronic hypoxia and contributes to the progression of
human prostate cancer, Int. J. Cancer 133 (11) (2013) 25772586.
[87] J. Yao, Cuijuan Qian, Inhibition of Notch3 enhances sensitivity to gemcitabine in
pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway, Med.
Oncol. 27 (3) (2010) 10171022.
[88] A. Sasnauskien ˙
e, V. Jonuˇ
sien˙
e, A. Krikˇ
staponien˙
e, S. Butkyt ˙
e, D. Dabkeviˇ
cien˙
e,
D. Kanopien ˙
e, et al., NOTCH1, NOTCH3, NOTCH4, and JAG2 protein levels in
human endometrial cancer, Medicina 50 (1) (2014) 1418.
[89] I. Nagamatsu, H. Onishi, S. Matsushita, M. Kubo, M. Kai, A. Imaizumi, et al.,
NOTCH4 is a potential therapeutic target for triple-negative breast cancer,
Anticancer Res. 34 (1) (2014) 6980.
[90] J.W. Wang, X.L. Wei, X.W. Dou, W.H. Huang, C.W. Du, G.J. Zhang, The
association between Notch4 expression, and clinicopathological characteristics
and clinical outcomes in patients with breast cancer, Oncol. Lett. 15 (6) (2018)
87498755.
[91] J. Zhang, Y. Kuang, Y. Wang, Q. Xu, Q. Ren, Notch-4 silencing inhibits prostate
cancer growth and EMT via the NF-κB pathway, Apoptosis 22 (6) (2017)
877884.
[92] C. Qian, F. Liu, B. Ye, X. Zhang, Y. Liang, J. Yao, Notch4 promotes gastric cancer
growth through activation of Wnt1/β-catenin signaling, Mol. Cell. Biochem. 401
(1) (2015) 165174.
[93] G. Wu, Z. Chen, J. Li, F. Ye, G. Chen, Q. Fan, et al., NOTCH4 is a novel prognostic
marker that correlates with colorectal cancer progression and prognosis,
J. Cancer 9 (13) (2018) 2374.
[94] T. Fukusumi, T.W. Guo, A. Sakai, M. Ando, S. Ren, S. Haft, et al., The
NOTCH4HEY1 pathway induces epithelial-mesenchymal transition in head and
neck squamous cell carcinomaNOTCH4HEY1 induces EMT in HNSCC, Clin.
Cancer Res. 24 (3) (2018) 619633.
[95] C.J. Qian, Y.Y. Chen, X. Zhang, F.Q. Liu, T.T. Yue, B. Ye, et al., Notch4 inhibition
reduces migration and invasion and enhances sensitivity to docetaxel by
inhibiting Akt/fascin in pancreatic cancer cells, Oncol. Lett. 12 (5) (2016)
34993505.
[96] L.-C. Ding, L. She, D.-L. Zheng, Q.-L. Huang, J.-F. Wang, F.-F. Zheng, et al., Notch-
4 contributes to the metastasis of salivary adenoid cystic carcinoma, Oncol. Rep.
24 (2) (2010) 363368.
[97] E. Bonyadi Rad, H. Hammerlindl, C. Wels, U. Popper, D. Ravindran Menon,
H. Breiteneder, et al., Notch4 signaling induces a MesenchymalEpitheliallike
transition in melanoma cells to suppress malignant behaviors, Cancer Res. 76 (7)
(2016) 16901697.
[98] E.S. Hibdon, N. Razumilava, T.M. Keeley, G. Wong, S. Solanki, Y.M. Shah, et al.,
Notch and mTOR signaling pathways promote human gastric cancer cell
proliferation, Neoplasia 21 (7) (2019) 702712.
[99] J. Hoarau-V´
echot, C. Touboul, N. Halabi, M. Blot-Dupin, R. Lis, C. Abi Khalil, et
al., Akt-activated endothelium promotes ovarian cancer proliferation through
notch activation, J. Transl. Med. 17 (1) (2019) 111.
[100] J. Zhang, K. Yu, X. Han, L. Zhen, M. Liu, X. Zhang, et al., Paeoniorin inuences
breast cancer cell proliferation and invasion via inhibition of the Notch-1
signaling pathway, Mol. Med. Rep. 17 (1) (2018) 13211325.
[101] J. Jiang, H. Ren, Y. Xu, M. Wudu, Q. Wang, Z. Liu, et al., TRIM67 promotes the
proliferation, migration, and invasion of non-small-cell lung cancer by positively
regulating the notch pathway, J. Cancer 11 (5) (2020) 1240.
[102] R.-H. Gan, L.-S. Lin, J. Xie, L. Huang, L.-C. Ding, B.-H. Su, et al., FLI-06 intercepts
notch signaling and suppresses the proliferation and self-renewal of tongue cancer
cells, OncoTargets and therapy 12 (2019) 7663.
[103] A. Baker, D. Wyatt, M. Bocchetta, J. Li, A. Filipovic, A. Green, et al., Notch-1-
PTEN-ERK1/2 signaling axis promotes HER2+breast cancer cell proliferation and
stem cell survival, Oncogene 37 (33) (2018) 44894504.
[104] Y. Jia, R. Lin, H. Jin, L. Si, W. Jian, Q. Yu, et al., MicroRNA-34 suppresses
proliferation of human ovarian cancer cells by triggering autophagy and
apoptosis and inhibits cell invasion by targeting notch 1, Biochimie 160 (2019)
193199.
[105] Z. Liu, T. Ma, J. Duan, X. Liu, L. Liu, MicroRNA-223-induced inhibition of the
FBXW7 gene affects the proliferation and apoptosis of colorectal cancer cells via
the notch and Akt/mTOR pathways, Mol. Med. Rep. 23 (2) (2021), 1-.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1165
[106] T. Iso, L. Kedes, Y. Hamamori, HES and HERP families: multiple effectors of the
notch signaling pathway, J. Cell. Physiol. 194 (3) (2003) 237255.
[107] K. Wu, J. Zou, C. Lin, Z.-G. Jie, MicroRNA-140-5p inhibits cell proliferation,
migration and promotes cell apoptosis in gastric cancer through the negative
regulation of THY1-mediated notch signaling, Biosci. Rep. 39 (7) (2019).
[108] B.-L. Pan, L. Wu, L. Pan, Y.-X. Yang, H.-H. Li, Y.-J. Dai, et al., Up-regulation of
microRNA-340 promotes osteosarcoma cell apoptosis while suppressing
proliferation, migration, and invasion by inactivating the CTNNB1-mediated
notch signaling pathway, Biosci. Rep. 38 (4) (2018).
[109] M. Sadrkhanloo, M. Entezari, S. Orouei, M. Ghollasi, S. Rezaei, E.S. Hejazi, et al.,
STAT3-EMT axis in tumors: modulation of cancer metastasis, stemness and
therapy response, Pharmacol. Res. 106311 (2022).
[110] M. Ashrazadeh, A. Zarrabi, K. Hushmandi, M. Kalantari, R. Mohammadinejad,
T. Javaheri, et al., Association of the epithelialmesenchymal transition (EMT)
with cisplatin resistance, Int. J. Mol. Sci. 21 (11) (2020) 4002.
[111] M. Ashrazadeh, S. Mirzaei, F. Hashemi, A. Zarrabi, A. Zabolian, H. Saleki, et al.,
New insight towards development of paclitaxel and docetaxel resistance in cancer
cells: EMT as a novel molecular mechanism and therapeutic possibilities, Biomed.
Pharmacother. 141 (2021), 111824.
[112] M. Sadrkhanloo, M. Entezari, M. Rashidi, M. Hashemi, R. Raesi, S. Saghari, et al.,
Non-coding RNAs in EMT regulation: association with tumor progression and
therapy response, Eur. J. Pharmacol. 175212 (2022).
[113] L. Li, P. Tang, S. Li, X. Qin, H. Yang, C. Wu, et al., Notch signaling pathway
networks in cancer metastasis: a new target for cancer therapy, Med. Oncol. 34
(10) (2017) 110.
[114] C.-X. Wen, H.-L. Tian, E. Chen, J.-F. Liu, X.-X. Liu, MiRNA-873-5p acts as a
potential novel biomarker and promotes cervical cancer progression by regulating
ZEB1 via notch signaling pathway, Dose-Response 19 (1) (2021),
15593258211001255.
[115] J. Li, Q. Li, L. Lin, R. Wang, L. Chen, W. Du, et al., Targeting the Notch1 oncogene
by miR-139-5p inhibits glioma metastasis and epithelial-mesenchymal transition
(EMT), BMC Neurol. 18 (1) (2018) 113.
[116] Y. Chen, B. Li, J. Feng, Q. Fang, J. Cheng, W. Xie, et al., JAG1, regulated by
microRNA-424-3p, involved in tumorigenesis and epithelial-mesenchymal
transition of high proliferative potential-pituitary adenomas, Front. Oncol. 10
(2020), 567021.
[117] S. Mondal, N. Adhikari, S. Banerjee, S.A. Amin, T. Jha, Matrix metalloproteinase-
9 (MMP-9) and its inhibitors in cancer: a minireview, Eur. J. Med. Chem. 194
(2020), 112260.
[118] Y. Zhong, Y.-T. Lu, Y. Sun, Z.-H. Shi, N.-G. Li, Y.-P. Tang, et al., Recent
opportunities in matrix metalloproteinase inhibitor drug design for cancer, Expert
Opin. Drug Discovery 13 (1) (2018) 7587.
[119] C.M. Overall, C. L´
opez-Otín, Strategies for MMP inhibition in cancer: innovations
for the post-trial era, Nat. Rev. Cancer 2 (9) (2002) 657672.
[120] S.S. Ganguly, G. Hostetter, L. Tang, S.B. Frank, K. Saboda, R. Mehra, et al., Notch3
promotes prostate cancer-induced bone lesion development via MMP-3,
Oncogene 39 (1) (2020) 204218.
[121] Y.-T. Chen, C.-R. Huang, C.-L. Chang, J.Y. Chiang, C.-W. Luo, H.-H. Chen, et al.,
Jagged2 progressively increased expression from stage I to III of bladder cancer
and melatonin-mediated downregulation of Notch/Jagged2 suppresses the
bladder tumorigenesis via inhibiting PI3K/AKT/mTOR/MMPs signaling, Int. J.
Biol. Sci. 16 (14) (2020) 2648.
[122] B. Yu, J. Wei, X. Qian, D. Lei, Q. Ma, Y. Liu, Notch1 signaling pathway
participates in cancer invasion by regulating MMPs in lingual squamous cell
carcinoma, Oncol. Rep. 27 (2) (2012) 547552.
[123] Y. Shui, X. Yu, R. Duan, Q. Bao, J. Wu, H. Yuan, et al., miR-130b-3p inhibits cell
invasion and migration by targeting the notch ligand Delta-like 1 in breast
carcinoma, Gene 609 (2017) 8087.
[124] H. Kawasaki, T. Takeuchi, F. Ricciardiello, A. Lombardi, E. Biganzoli, M. Fornili,
et al., Denition of miRNA signatures of nodal metastasis in LCa: miR-449a
targets notch genes and suppresses cell migration and invasion, Mol. Ther.
Nucleic Acids 20 (2020) 711724.
[125] X. Zhang, F. Ai, X. Li, L. Tian, X. Wang, S. Shen, et al., MicroRNA-34a suppresses
colorectal cancer metastasis by regulating notch signaling, Oncol. Lett. 14 (2)
(2017) 23252333.
[126] W. Li, X. Xie, J. Bai, C. Wang, L. Zhao, D. Jiang, Increased expression of miR-1179
inhibits breast cancer cell metastasis by modulating notch signaling pathway and
correlates with favorable prognosis, Eur. Rev. Med. Pharmacol. Sci. 22 (23)
(2018) 83748382.
[127] L.-Z. Yang, C.-C. Lei, Y.-P. Zhao, H.-W. Sun, Q.-H. Yu, E.-J. Yang, MicroRNA-34c-
3p target inhibiting NOTCH1 suppresses chemosensitivity and metastasis of non-
small cell lung cancer, Journal of International Medical Research 48 (3) (2020),
0300060520904847.
[128] Y. Bae, H.C. Zeng, Y.T. Chen, S. Ketkar, E. Munivez, Z. Yu, et al., miRNA-34c
suppresses osteosarcoma progression in vivo by targeting notch and E2F, JBMR
plus. 6 (5) (2022), e10623.
[129] E.Y. Park, E. Chang, E.J. Lee, H.-W. Lee, H.-G. Kang, K.-H. Chun, et al., Targeting
of miR34aNOTCH1 Axis reduced breast cancer stemness and
ChemoresistancemiR34a regulates self-renewal through Notch signaling, Cancer
Res. 74 (24) (2014) 75737582.
[130] X. Ji, Z. Wang, A. Geamanu, A. Goja, F.H. Sarkar, S.V. Gupta, Delta-tocotrienol
suppresses Notch-1 pathway by upregulating miR-34a in nonsmall cell lung
cancer cells, Int. J. Cancer 131 (11) (2012) 26682677.
[131] H. Wei, X. Wang, X. Niu, R. Jiao, X. Li, S. Wang, miR-34c-5p targets Notch1 and
suppresses the metastasis and invasion of cervical cancer, Mol. Med. Rep. 23 (2)
(2021) 1.
[132] D.M. Valcourt, E.S. Day, Dual regulation of miR-34a and notch signaling in triple-
negative breast cancer by antibody/miRNA nanocarriers, Mol. Ther.Nucleic
Acids 21 (2020) 290298.
[133] N. Caporarello, G. Lupo, M. Olivieri, M. Cristaldi, M.T. Cambria, M. Salmeri, et
al., Classical VEGF, notch and ang signalling in cancer angiogenesis, alternative
approaches and future directions, Mol. Med. Rep. 16 (4) (2017) 43934402.
[134] A. Garcia, J.J. Kandel, Notch: a key regulator of tumor angiogenesis and
metastasis, Histol. Histopathol. 27 (2) (2012) 151.
[135] W. Zhou, G. Wang, S. Guo, Regulation of angiogenesis via notch signaling in
breast cancer and cancer stem cells. Biochimica et biophysica acta (BBA)-reviews
on, Cancer 1836 (2) (2013) 304320.
[136] A.O. Rehman, C.-Y. Wang, Notch signaling in the regulation of tumor
angiogenesis, Trends Cell Biol. 16 (6) (2006) 293300.
[137] T. Kangsamaksin, A. Murtomaki, N.M. Koer, H. Cuervo, R.A. Chaudhri, I.
W. Tattersall, et al., NOTCH decoys that selectively block DLL/NOTCH or JAG/
NOTCH disrupt angiogenesis by unique mechanisms to inhibit tumor
GrowthAntiangiogenic therapy via JAG/NOTCH blockade, Cancer Discov. 5 (2)
(2015) 182197.
[138] J. Dufraine, Y. Funahashi, J. Kitajewski, Notch signaling regulates tumor
angiogenesis by diverse mechanisms, Oncogene 27 (38) (2008) 51325137.
[139] A. Fischer, N. Schumacher, M. Maier, M. Sendtner, M. Gessler, The notch target
genes Hey1 and Hey2 are required for embryonic vascular development, Genes
Dev. 18 (8) (2004) 901911.
[140] Z. Jin, T. Zhan, J. Tao, B. Xu, H. Zheng, Y. Cheng, et al., MicroRNA-34a induces
transdifferentiation of glioma stem cells into vascular endothelial cells by
targeting notch pathway, Biosci. Biotechnol. Biochem. 81 (10) (2017)
18991907.
[141] S. Shi, Y. Jin, H. Song, X. Chen, MicroRNA-34a attenuates VEGF-mediated retinal
angiogenesis via targeting Notch1, Biochem. Cell Biol. 97 (4) (2019) 423430.
[142] H.-M. Jeon, S.-H. Kim, X. Jin, J.B. Park, S.H. Kim, K. Joshi, et al., Crosstalk
between glioma-initiating cells and endothelial cells drives tumor
ProgressionActivation of PDGFNOSID4miR129 Axis in glioma, Cancer Res. 74
(16) (2014) 44824492.
[143] X. Ji, Y. Lu, H. Tian, X. Meng, M. Wei, W.C. Cho, Chemoresistance mechanisms of
breast cancer and their countermeasures, Biomed. Pharmacother. 114 (2019),
108800.
[144] H.-C. Zheng, The molecular mechanisms of chemoresistance in cancers,
Oncotarget 8 (35) (2017) 59950.
[145] F. Guerra, A.A. Arbini, L. Moro, Mitochondria and cancer chemoresistance,
Biochim. Biophys. Acta, Bioenerg. 1858 (8) (2017) 686699.
[146] A. Nath, S. Mitra, T. Mistry, R. Pal, V.D. Nasare, Molecular targets and
therapeutics in chemoresistance of triple-negative breast cancer, Med. Oncol. 39
(1) (2022) 133.
[147] Y. Sun, D. Dong, Y. Xia, L. Hao, W. Wang, C. Zhao, YTHDF1 promotes breast
cancer cell growth, DNA damage repair and chemoresistance, Cell Death Dis. 13
(3) (2022) 111.
[148] A. Chimento, M. DAmico, V. Pezzi, F. De Amicis, Notch signaling in breast tumor
microenvironment as mediator of drug resistance, Int. J. Mol. Sci. 23 (11) (2022)
6296.
[149] Q. Wu, J. Guo, Y. Liu, Q. Zheng, X. Li, C. Wu, et al., YAP drives fate conversion
and chemoresistance of small cell lung cancer. Science, Advances (2021;7(40):
eabg1850.).
[150] T. Sun, D. Zhang, Z. Wang, B. Zhao, Y. Li, X. Sun, et al., Inhibition of the notch
signaling pathway overcomes resistance of cervical cancer cells to paclitaxel
through retardation of the epithelialmesenchymal transition process, Environ.
Toxicol. 36 (9) (2021) 17581764.
[151] S.S. Islam, K. Qassem, S. Islam, R.R. Parag, M.Z. Rahman, W.A. Farhat, et al.,
Genetic alterations of Keap1 confers chemotherapeutic resistance through
functional activation of Nrf2 and notch pathway in head and neck squamous cell
carcinoma, Cell Death Dis. 13 (8) (2022) 113.
[152] S. Kumar, A. Nandi, S. Singh, R. Regulapati, N. Li, J.W. Tobias, et al., Dll1+
quiescent tumor stem cells drive chemoresistance in breast cancer through NF-κB
survival pathway, Nat. Commun. 12 (1) (2021) 113.
[153] V. Kumar, M. Vashishta, L. Kong, X. Wu, J.J. Lu, C. Guha, et al., The role of Notch,
Hedgehog, and Wnt signaling pathways in the resistance of tumors to anticancer
therapies, Frontiers in cell and developmental biology (2021) 857.
[154] L. Rios-Colon, J. Chijioke, S. Niture, Z. Afzal, Q. Qi, A. Srivastava, et al., Leptin
modulated microRNA-628-5p targets Jagged-1 and inhibits prostate cancer
hallmarks, Sci. Rep. 12 (1) (2022) 115.
[155] J. Huang, Y. Chen, J. Li, K. Zhang, J. Chen, D. Chen, et al., Notch-1 confers
chemoresistance in lung adenocarcinoma to taxanes through AP-1/microRNA-
451 mediated regulation of MDR-1, Mol. Ther.Nucleic Acids 5 (2016), e375.
[156] Z.-Y. Xie, F.-F. Wang, Z.-H. Xiao, S.-F. Liu, S.-L. Tang, Y.-L. Lai, Overexpressing
microRNA-34a overcomes ABCG2-mediated drug resistance to 5-FU in side
population cells from colon cancer via suppressing DLL1, J. Biochem. 167 (6)
(2020) 557564.
[157] L. Lan, Y. Wang, Z. Pan, B. Wang, Z. Yue, Z. Jiang, et al., Rhamnetin induces
apoptosis in human breast cancer cells via the miR-34a/Notch-1 signaling
pathway, Oncol. Lett. 17 (1) (2019) 676682.
[158] S. Roy, E. Levi, A.P. Majumdar, F.H. Sarkar, Expression of miR-34 is lost in colon
cancer which can be re-expressed by a novel agent CDF, J. Hematol. Oncol. 5 (1)
(2012) 16.
[159] W. Tang, Q. Ji, X. Hao, L. Li, D. Jeffrey, E.R. Fearon, et al., Tumor suppressor
microRNA miR-34 inhibits human pancreatic cancer stem cells, Cancer Res. 70 (8_
Supplement) (2010) 2084.
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1166
[160] X.-j. Li, M.-h. Ji, S.-l. Zhong, Q.-b. Zha, J.-j. Xu, J.-h. Zhao, et al., MicroRNA-34a
modulates chemosensitivity of breast cancer cells to adriamycin by targeting
Notch1, Arch. Med. Res. 43 (7) (2012) 514521.
[161] X. Yang, P. Shang, B. Yu, Q. Jin, J. Liao, L. Wang, et al., Combination therapy with
miR34a and doxorubicin synergistically inhibits dox-resistant breast cancer
progression via down-regulation of snail through suppressing Notch/NF-κB and
RAS/RAF/MEK/ERK signaling pathway, Acta Pharm. Sin. B 11 (9) (2021)
28192834.
[162] Y. Jin, M. Wang, H. Hu, Q. Huang, Y. Chen, G. Wang, Overcoming stemness and
chemoresistance in colorectal cancer through miR-195-5p-modulated inhibition
of notch signaling, Int. J. Biol. Macromol. 117 (2018) 445453.
[163] M.X. Liu, M.K. Siu, S.S. Liu, J.W. Yam, H.Y. Ngan, D.W. Chan, Epigenetic silencing
of microRNA-199b-5p is associated with acquired chemoresistance via activation
of JAG1-Notch1 signaling in ovarian cancer, Oncotarget 5 (4) (2014) 944.
[164] J.-Y. Jeong, H. Kang, T.H. Kim, G. Kim, J.-H. Heo, A.-Y. Kwon, et al., MicroRNA-
136 inhibits cancer stem cell activity and enhances the anti-tumor effect of
paclitaxel against chemoresistant ovarian cancer cells by targeting Notch3,
Cancer Lett. 386 (2017) 168178.
[165] T. Tian, X. Fu, J. Lu, Z. Ruan, K. Nan, Y. Yao, et al., MicroRNA-760 inhibits
doxorubicin resistance in hepatocellular carcinoma through regulating Notch1/
Hes1-PTEN/Akt signaling pathway, J. Biochem. Mol. Toxicol. 32 (8) (2018),
e22167.
[166] D. Zhu, J. Xia, C. Liu, C. Fang, Numb/Notch/PLK1 signaling pathway mediated
hyperglycemic memory in pancreatic cancer cell radioresistance and the
therapeutic effects of metformin, Cell. Signal. 93 (2022), 110268.
[167] A. Nandi, R. Chakrabarti, The many facets of notch signaling in breast cancer:
toward overcoming therapeutic resistance, Genes Dev. 34 (2122) (2020)
14221438.
[168] Y. Zhou, Y. Su, H. Zhu, X. Wang, X. Li, C. Dai, et al., Interleukin-23 receptor
signaling mediates cancer dormancy and radioresistance in human esophageal
squamous carcinoma cells via the Wnt/Notch pathway, J. Mol. Med. 97 (2)
(2019) 177188.
[169] J. Wang, T.P. Wakeman, J.D. Lathia, A.B. Hjelmeland, X.-F. Wang, R.R. White, et
al., Notch promotes radioresistance of glioma stem cells, Stem Cells 28 (1) (2010)
1728.
[170] J. Theys, S. Yahyanejad, R. Habets, P. Span, L. Dubois, K. Paesmans, et al., High
NOTCH activity induces radiation resistance in non small cell lung cancer,
Radiother. Oncol. 108 (3) (2013) 440445.
[171] Q. Zhao, A. Mao, J. Yan, C. Sun, C. Di, X. Zhou, et al., Downregulation of Nrf2
promotes radiation-induced apoptosis through Nrf2 mediated notch signaling in
non-small cell lung cancer cells, Int. J. Oncol. 48 (2) (2016) 765773.
[172] S. Yahyanejad, J. Theys, M. Vooijs, Targeting notch to overcome radiation
resistance, Oncotarget 7 (7) (2016) 7610.
[173] Z. Shen, Y. Wu, G. He, Long non-coding RNA PTPRG-AS1/microRNA-124-3p
regulates radiosensitivity of nasopharyngeal carcinoma via the LIM homeobox 2-
dependent notch pathway through competitive endogenous RNA mechanism,
Bioengineered 13 (4) (2022) 82088225.
[174] Z. Zhao, X. Shen, D. Zhang, H. Xiao, H. Kong, B. Yang, et al., miR-153 enhances
the therapeutic effect of radiotherapy by targeting JAG1 in pancreatic cancer
cells, Oncol. Lett. 21 (4) (2021) 18.
[175] B. Yang, C. Wang, H. Xie, Y. Wang, J. Huang, Y. Rong, et al., MicroRNA-3163
targets ADAM-17 and enhances the sensitivity of hepatocellular carcinoma cells
to molecular targeted agents, Cell Death Dis. 10 (10) (2019) 115.
[176] T. Pan, H. Ding, L. Jin, S. Zhang, D. Wu, W. Pan, et al., DNMT1-mediated
demethylation of lncRNA MEG3 promoter suppressed breast cancer progression
by repressing Notch1 signaling pathway, Cell Cycle 115 (2022).
[177] G. Yan, Z. Chang, C. Wang, Z. Gong, H. Xin, Z. Liu, LncRNA ILF3-AS1 promotes
cell migration, invasion and EMT process in hepatocellular carcinoma via the
miR-6285p/MEIS2 axis to activate the notch pathway, Dig. Liver Dis. 54 (1)
(2022) 125135.
[178] T. Zhang, X. Liu, K. Su, Q. Zheng, P. Liu, Z. Xu, et al., A novel mechanism of the
lncRNA PTTG3P/miR-142-5p/JAG1 axis modulating tongue cancer cell
phenotypes through the Notch1 signaling, Cells Dev. 169 (2022), 203762.
[179] S. Huang, S. Liang, J. Huang, P. Luo, D. Mo, H. Wang, LINC01806 mediated by
STAT1 promotes cell proliferation, migration, invasion, and stemness in non-
small cell lung cancer through notch signaling by miR-4428/NOTCH2 axis,
Cancer Cell Int. 22 (1) (2022) 115.
[180] M. He, Y. Wang, J. Cai, Y. Xie, C. Tao, Y. Jiang, et al., LncRNA DLEU2 promotes
cervical cancer cell proliferation by regulating cell cycle and NOTCH pathway,
Exp. Cell Res. 402 (1) (2021), 112551.
[181] G. Li, Z. Zhang, Z. Chen, B. Liu, H. Wu, LncRNA DLEU2 is activated by STAT1 and
induces gastric cancer development via targeting miR-23b-3p/NOTCH2 axis and
Notch signaling pathway, Life Sci. 277 (2021), 119419.
[182] J. Xu, W. Xu, X. Yang, Z. Liu, Y. Zhao, Q. Sun, LncRNA MIR99AHG mediated by
FOXA1 modulates NOTCH2/Notch signaling pathway to accelerate pancreatic
cancer through sponging miR-3129-5p and recruiting ELAVL1, Cancer Cell Int. 21
(1) (2021) 114.
[183] Y. Li, Y. Gao, X. Niu, M. Tang, J. Li, B. Song, et al., LncRNA BASP1-AS1 interacts
with YBX1 to regulate notch transcription and drives the malignancy of
melanoma, Cancer Sci. 112 (11) (2021) 4526.
[184] H. Yang, Z. Jiang, S. Wang, Y. Zhao, X. Song, Y. Xiao, et al., Long non-coding
small nucleolar RNA host genes in digestive cancers, Cancer Med. 8 (18) (2019)
76937704.
[185] W. Chen, L. Huang, J. Liang, Y. Ye, S. Yu, Y. Zhang, Long noncoding RNA small
nucleolar RNA host gene 15 deteriorates liver cancer via microRNA-18b-5p/LIM-
only 4 axis, IUBMB Life 73 (2) (2021) 349361.
[186] S. Shen, Y. Wang, Y. Zhang, Z. Dong, J. Xing, Long non-coding RNA small
nucleolar RNA host gene 14, a promising biomarker and therapeutic target in
malignancy, Front. Cell Dev. Biol. 9 (2021), 746714.
[187] Z. Yang, Q. Li, X. Zheng, L. Xie, Long noncoding RNA small nucleolar host gene: a
potential therapeutic target in urological cancers, Front. Oncol. 11 (2021),
638721.
[188] Y. Wang, D. Bao, L. Wan, C. Zhang, S. Hui, H. Guo, Long non-coding RNA small
nucleolar RNA host gene 7 facilitates the proliferation, migration, and invasion of
esophageal cancer cells by regulating microRNA-625, J. Gastrointest. Oncol. 12
(2) (2021) 423.
[189] D. Cheng, J. Fan, K. Qin, Y. Zhou, J. Yang, Y. Ma, et al., LncRNA SNHG7 regulates
mesenchymal stem cell through the Notch1/Jagged1/Hes-1 signaling pathway
and inuences folrinox resistance in pancreatic cancer, Front. Oncol. 11 (2021),
719855.
[190] X. Sun, T. Huang, Z. Liu, M. Sun, S. Luo, LncRNA SNHG7 contributes to
tumorigenesis and progression in breast cancer by interacting with miR-34a
through EMT initiation and the Notch-1 pathway, Eur. J. Pharmacol. 856 (2019),
172407.
[191] G. Hu, J. Ma, J. Zhang, Y. Chen, H. Liu, Y. Huang, et al., Hypoxia-induced
lncHILAR promotes renal cancer metastasis via ceRNA for the miR-613/206/1-1-
3p/Jagged-1/Notch/CXCR4 signaling pathway, Mol. Ther. 29 (10) (2021)
29792994.
[192] C. Zou, S. Wu, H. Wei, H. Luo, Z. Tang, X. Li, et al., LINC01355 contributes to
malignant phenotype of oral squamous cell carcinoma and cytotoxic T cell
inltration via activating notch signaling pathway, Journal of
immunologyResearch 2021 (2021).
[193] J. Liu, Y. Shi, M. Wu, F. Zhang, M. Xu, Z. He, et al., JAG1 enhances angiogenesis
in triple-negative breast cancer through promoting the secretion of exosomal
lncRNA MALAT1, Genes Dis. (2022).
[194] Y. Deng, L. Zhang, LncRNA SNHG11 accelerates the progression of lung
adenocarcinoma via activating notch pathways, Pathol. Res. Pract. 234 (2022),
153849.
[195] L. Zhang, G. Li, X. Wang, Y. Zhang, X. Huang, H. Wu, lncRNA SNHG3 acts as
oncogene in ovarian cancer through miR-139-5p and Notch1, Oncol. Lett. 21 (2)
(2021), 1-.
[196] H. Jiang, X. Li, W. Wang, H. Dong, Long non-coding RNA SNHG3 promotes breast
cancer cell proliferation and metastasis by binding to microRNA-154-3p and
activating the notch signaling pathway, BMC Cancer 20 (1) (2020) 113.
[197] X. Mao, T. Ji, A. Liu, Y. Weng, ELK4-mediated lncRNA SNHG22 promotes gastric
cancer progression through interacting with EZH2 and regulating miR-200c-3p/
Notch1 axis, Cell Death Dis. 12 (11) (2021) 111.
[198] P. Cai, G. Li, M. Wu, B. Zhang, H. Bai, ZIC2 upregulates lncRNA SNHG12
expression to promote endometrial cancer cell proliferation and migration by
activating the notch signaling pathway, Mol. Med. Rep. 24 (3) (2021) 19.
[199] Z.-B. Liu, C. Tang, X. Jin, S.-H. Liu, W. Pi, Increased expression of lncRNA
SNHG12 predicts a poor prognosis of nasopharyngeal carcinoma and regulates
cell proliferation and metastasis by modulating notch signal pathway, Cancer
Biomark. 23 (4) (2018) 603613.
[200] L.K. Kim, S. Park, Y. Yang, Y.T. Kim, T.-H. Heo, H.J. Kim, LncRNA SRA mediates
cell migration, invasion, and progression of ovarian cancer via NOTCH signaling
and epithelialmesenchymal transition, Biosci. Rep. 41 (9) (2021).
[201] Q. Wu, S. Lu, L. Zhang, L. Zhao, LncRNA HOXA-AS2 activates the notch pathway
to promote cervical cancer cell proliferation and migration, Reprod. Sci. 28 (10)
(2021) 30003009.
[202] J. Xu, W. Xu, X. Yang, Z. Liu, Q. Sun, LncRNA HCG11/miR-579-3p/MDM2 axis
modulates malignant biological properties in pancreatic carcinoma via Notch/
Hes1 signaling pathway, Aging (Albany NY) 13 (12) (2021) 16471.
[203] L. Pan, M. Du, H. Liu, B. Cheng, M. Zhu, B. Jia, et al., LncRNA FTX promotes the
malignant progression of colorectal cancer by regulating the miR-214-5pJAG1
axis, Ann. Transl. Med. 9 (17) (2021).
[204] Q. Guo, Z. Qian, D. Yan, L. Li, L. Huang, LncRNA-MEG3 inhibits cell proliferation
of endometrial carcinoma by repressing notch signaling, Biomed. Pharmacother.
82 (2016) 589594.
[205] L. Gai, Y. Huang, L. Zhao, F. Li, Z. Zhuang, Long non-coding RNA HAGLROS
regulates the proliferation, migration, and apoptosis of esophageal cancer cells
via the HAGLROS-miR-206-NOTCH3 axis, J. Gastrointest. Oncol. 12 (5) (2021)
2093.
[206] Th. Zhang, Liang Lz, Wu. Liu Xl, Su.K. Jn, Chen Jy, et al., LncRNA UCA1/miR-124
axis modulates TGFβ1-induced epithelial-mesenchymal transition and invasion of
tongue cancer cells through JAG1/Notch signaling, J. Cell. Biochem. 120 (6)
(2019) 1049510504.
[207] Y. Zhu, Y. Tong, J. Wu, Y. Liu, M. Zhao, Knockdown of LncRNA GHET1 suppresses
prostate cancer cell proliferation by inhibiting HIF-1
α
/Notch-1 signaling pathway
via KLF2, Biofactors 45 (3) (2019) 364373.
[208] L. Luo, Y. Zhang, H. He, C. Chen, B. Zhang, M. Cai, LncRNA FEZF1-AS1 sponges
miR-34a to upregulate Notch-1 in glioblastoma, Cancer Manag. Res. 12 (2020)
1827.
[209] S. Huang, C. Li, J. Huang, P. Luo, D. Mo, H. Wang, LncRNA FEZF1-AS1 promotes
non-small lung cancer cell migration and invasion through the up-regulation of
NOTCH1 by serving as a sponge of miR-34a, BMC Pulm. Med. 20 (1) (2020) 17.
[210] X. Wang, Y. Yan, C. Zhang, W. Wei, X. Ai, Y. Pang, et al., Upregulation of lncRNA
PlncRNA-1 indicates the poor prognosis and promotes glioma progression by
activation of notch signal pathway, Biomed. Pharmacother. 103 (2018) 216221.
[211] X. Wang, G. Zhang, Z. Cheng, L. Dai, L. Jia, X. Jing, et al., Knockdown of LncRNA-
XIST suppresses proliferation and TGF-β1-induced EMT in NSCLC through the
M. Hashemi et al.
International Journal of Biological Macromolecules 222 (2022) 1151–1167
1167
Notch-1 pathway by regulation of miR-137, Genet. Test. Mol. Biomarkers 22 (6)
(2018) 333342.
[212] S. Li, H. Zhao, J. Li, A. Zhang, H. Wang, Downregulation of long non-coding RNA
LET predicts poor prognosis and increases notch signaling in non-small cell lung
cancer, Oncotarget 9 (1) (2018) 1156.
[213] Z. Liu, H. Li, S. Fan, H. Lin, W. Lian, STAT3-induced upregulation of long
noncoding RNA HNF1A-AS1 promotes the progression of oral squamous cell
carcinoma via activating notch signaling pathway, Cancer Biol. Ther. 20 (4)
(2019) 444453.
[214] S. Lu, W. Dong, P. Zhao, Z. Liu, lncRNA FAM83H-AS1 is associated with the
prognosis of colorectal carcinoma and promotes cell proliferation by targeting the
notch signaling pathway, Oncol. Lett. 15 (2) (2018) 18611868.
[215] M. Lee, H.J. Kim, S.W. Kim, S.-A. Park, K.-H. Chun, N.H. Cho, et al., The long non-
coding RNA HOTAIR increases tumour growth and invasion in cervical cancer by
targeting the notch pathway, Oncotarget 7 (28) (2016) 44558.
[216] H. Cai, J. Yao, Y. An, X. Chen, W. Chen, D. Wu, et al., LncRNA HOTAIR acts as
competing endogenous RNA to control the expression of Notch3 via sponging
miR-613 in pancreatic cancer, Oncotarget 8 (20) (2017) 32905.
[217] K. Gao, Z. Ji, K. She, Q. Yang, L. Shao, Long non-coding RNA ZFAS1 is an
unfavourable prognostic factor and promotes glioma cell progression by
activation of the notch signaling pathway, Biomed. Pharmacother. 87 (2017)
555560.
[218] J. Xu, G. Wu, Y. Zhao, Y. Han, S. Zhang, C. Li, et al., Long noncoding RNA
DSCAM-AS1 facilitates colorectal cancer cell proliferation and migration via miR-
137/Notch1 axis, J. Cancer 11 (22) (2020) 6623.
[219] H. Li, Y. Wang, Long noncoding RNA (lncRNA) MIR22HG suppresses gastric
cancer progression through attenuating NOTCH2 signaling, Med. Sci. Monit. 25
(2019) 656.
[220] S. Tao, Q. Chen, C. Lin, H. Dong, Linc00514 promotes breast cancer metastasis
and M2 polarization of tumor-associated macrophages via Jagged1-mediated
notch signaling pathway, J. Exp. Clin. Cancer Res. 39 (1) (2020) 117.
[221] Y. Wang, P. Wu, R. Lin, L. Rong, Y. Xue, Y. Fang, LncRNA NALT interaction with
NOTCH1 promoted cell proliferation in pediatric T cell acute lymphoblastic
leukemia, Sci. Rep. 5 (1) (2015) 110.
[222] J. Huang, B. Pan, G. Xia, J. Zhu, C. Li, J. Feng, LncRNA SNHG15 regulates EGFR-
TKI acquired resistance in lung adenocarcinoma through sponging miR-451 to
upregulate MDR-1, Cell Death Dis. 11 (7) (2020) 114.
[223] Z.-h. Li, N.-s. Yu, Q. Deng, Y. Zhang, Y.-y. Hu, G. Liu, et al., Lncrna snhg7
mediates the chemoresistance and stemness of breast cancer by sponging mir-34a,
Front. Oncol. 10 (2020), 592757.
[224] Q. Hang, R. Sun, C. Jiang, Y. Li, Notch 1 promotes cisplatin-resistant gastric
cancer formation by upregulating lncRNA AK022798 expression, Anti-Cancer
Drugs 26 (6) (2015) 632640.
[225] Y. Ma, B. Fan, Z. Ren, B. Liu, Y. Wang, Long noncoding RNA DANCR contributes
to docetaxel resistance in prostate cancer through targeting the miR-34a-5p/JAG1
pathway, OncoTargets Ther. 12 (2019) 5485.
[226] L. Bai, A. Wang, Y. Zhang, X. Xu, X. Zhang, Knockdown of MALAT1 enhances
chemosensitivity of ovarian cancer cells to cisplatin through inhibiting the
Notch1 signaling pathway, Exp. Cell Res. 366 (2) (2018) 161171.
[227] Z. Bian, J. Zhang, M. Li, Y. Feng, S. Yao, M. Song, et al., Long non-coding RNA
LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in
colorectal cancer by inhibiting miR-139-5p, Oncogenesis 6 (11) (2017) 111.
[228] S. Zhang, S. Guo, C. Liang, M. Lian, Long intergenic noncoding RNA 00021
promotes glioblastoma temozolomide resistance by epigenetically silencing p21
through notch pathway, IUBMB Life 72 (8) (2020) 17471756.
[229] F. Zhang, Y. Sang, D. Chen, X. Wu, X. Wang, W. Yang, et al., M2 macrophage-
derived exosomal long non-coding RNA AGAP2-AS1 enhances radiotherapy
immunity in lung cancer by reducing microRNA-296 and elevating NOTCH2, Cell
Death Dis. 12 (5) (2021) 115.
[230] M. Yu, B. Lu, J. Zhang, J. Ding, P. Liu, Y. Lu, tRNA-derived RNA fragments in
cancer: current status and future perspectives, J. Hematol. Oncol. 13 (1) (2020)
114.
[231] L. Zhu, J. Ge, T. Li, Y. Shen, J. Guo, tRNA-derived fragments and tRNA halves: the
new players in cancers, Cancer Lett. 452 (2019) 3137.
[232] P. Zhu, J. Yu, P. Zhou, Role of tRNA-derived fragments in cancer: novel diagnostic
and therapeutic targets tRFs in cancer, Am. J. Cancer Res. 10 (2) (2020) 393.
[233] M. Zhang, F. Li, J. Wang, W. He, Y. Li, H. Li, et al., tRNA-derived fragment tRF-
03357 promotes cell proliferation, migration and invasion in high-grade serous
ovarian cancer, OncoTargets Ther. 12 (2019) 6371.
[234] X. Li, X. Liu, D. Zhao, W. Cui, Y. Wu, C. Zhang, et al., tRNA-derived small RNAs:
novel regulators of cancer hallmarks and targets of clinical application, Cell Death
Discov. 7 (1) (2021) 110.
[235] D. Yao, X. Sun, L. Zhou, M. Amanullah, X. Pan, Y. Liu, et al., OncotRF: an online
resource for exploration of tRNA-derived fragments in human cancers, RNA Biol.
17 (8) (2020) 10811091.
[236] B. Huang, H. Yang, X. Cheng, D. Wang, S. Fu, W. Shen, et al., tRF/miR-1280
suppresses stem Celllike cells and metastasis in colorectal CancertRF/miR-1280
represses colorectal cancer via notch signaling, Cancer Res. 77 (12) (2017)
31943206.
[237] I.L. Patop, S. Kadener, circRNAs in cancer, Curr. Opin. Genet. Dev. 48 (2018)
121127.
[238] L. Chen, G. Shan, CircRNA in cancer: fundamental mechanism and clinical
potential, Cancer Lett. 505 (2021) 4957.
[239] S. Meng, H. Zhou, Z. Feng, Z. Xu, Y. Tang, P. Li, et al., CircRNA: functions and
properties of a novel potential biomarker for cancer, Mol. Cancer 16 (1) (2017)
18.
[240] C. Xue, G. Li, Q. Zheng, X. Gu, Z. Bao, J. Lu, et al., The functional roles of the
circRNA/Wnt axis in cancer, Mol. Cancer 21 (1) (2022) 124.
[241] H. Xu, Y. Zhang, L. Qi, L. Ding, H. Jiang, H. Yu, NFIX circular RNA promotes
glioma progression by regulating miR-34a-5p via notch signaling pathway, Front.
Mol. Neurosci. 11 (2018) 225.
[242] Y. Li, S. Lin, N. An, Hsa_circ_0009910: oncogenic circular RNA targets microRNA-
145 in ovarian cancer cells, Cell Cycle 19 (15) (2020) 18571868.
[243] Y.-r. Shi, Z. Wu, K. Xiong, Q.-j. Liao, X. Ye, P. Yang, et al., Circular RNA circKIF4A
sponges miR-375/1231 to promote bladder cancer progression by upregulating
NOTCH2 expression, Front. Pharmacol. 11 (2020) 605.
[244] Y. Yao, X. Chen, H. Yang, W. Chen, Y. Qian, Z. Yan, et al., Hsa_circ_0058124
promotes papillary thyroid cancer tumorigenesis and invasiveness through the
NOTCH3/GATAD2A axis, J. Exp. Clin. Cancer Res. 38 (1) (2019) 118.
[245] H.-B. Wu, S.-S. Huang, C.-G. Lu, S.-D. Tian, M. Chen, CircAPLP2 regulates the
proliferation and metastasis of colorectal cancer by targeting miR-101-3p to
activate the notch signalling pathway, Am. J. Transl. Res. 12 (6) (2020) 2554.
[246] L.Y. Chen, Z. Zhi, L. Wang, Y.Y. Zhao, M. Deng, Y.H. Liu, et al., NSD2 circular
RNA promotes metastasis of colorectal cancer by targeting miR-199b-5p-
mediated DDR1 and JAG1 signalling, J. Pathol. 248 (1) (2019) 103115.
[247] Y. Yao, X. Li, L. Cheng, X. Wu, B. Wu, Circular RNA FAT atypical cadherin 1
(circFAT1)/microRNA-525-5p/spindle and kinetochore-associated complex
subunit 1 (SKA1) axis regulates oxaliplatin resistance in breast cancer by
activating the notch and wnt signaling pathway, Bioengineered 12 (1) (2021)
40324043.
M. Hashemi et al.
... Evanescent waves have attracted substantial attention in atom and nonlinear optics, optical trapping, sensing, and microscopy. [1][2][3][4][5][6][7] It has been shown that evanescent waves are able to translate colloidal-sized particles next to a flat plate in water, drive microparticles along a channel waveguide surface, alter the motion of micrometer-sized particles and neutral atoms, and trap small particles close to a dielectric interface. [8][9][10][11][12][13] Evanescent waves have been used to displace objects at nanometer and micrometer levels and excite fluorescence molecules on the surface of a cell. ...
... Among these applications, we can name evanescent based imaging and sensing. 4,7 The curves of the enhancement factor as a function of the incident angle have been plotted at three different wavelengths. To verify the obtained results, the transmittance curve is calculated as a function of the incident angle. ...
Article
We theoretically investigate the enhancement of the evanescent waves near the surface of a prism, coated with several layers of metals, graphene and double negative materials. The thicknesses of the metals and double negative material films along with the number of graphene layers are optimized to achieve the highest enhancement factor. We show that the intensity of the evanescent wave in the proposed resonance structure is about 109 times higher than that of a bare prism. A comparison between our results and those reported in the literature indicates a 104 times amplification of the evanescent field intensities. This amplification can open a new window to manipulate attached biological cells, design perfect atom mirrors and apply an effective force on nano- and micro- particles for the purpose of manipulation and trapping. The effect of the optimization of the involved parameters on the results shows the importance of these theoretical and simulation results, which leads to the reduction in time and costs of experimental works.
... It is worth mentioning that we have normalized the time-average power transported by each mode. Equations (28) and (29) are generalized equations that can be used to calculate the coupling coefficients of both doublepositive and double-negative waveguide gratings. In the case that both ε i and µ i (i = c , f , s ) are positive, they will reduce to the less general and simple equations in the literature [54]. ...
... For thicknesses far above the cutoff, where N is very close to n f due to the presence of c 2 ε f µ f − N 2 at the numerator of Eqs. (28) and (29), the coupling coefficients are very small. We see that the peak of ...
Article
Full-text available
The propagation of waves in waveguide gratings with a double-negative film but double-positive cover and substrate is studied. Perturbation theory is used to derive expressions that approximate the transverse and longitudinal coupling coefficients for coupling between various transverse-magnetic guided modes in a three-layer waveguide with a rectangular grating profile at the film–cover interface. An analytical method is then used to draw the coupling coefficients’ curves as functions of the film thickness. The curves of the waveguide gratings with double-negative material film differ significantly from that of the conventional waveguide gratings and those with a double-negative cover. Numerical results show that the obtained coupling coefficients are much higher than those of conventional waveguide gratings. Waveguide gratings with double-negative film possess a number of unusual properties. We show that the fundamental mode does not exist and the first-order mode is confined within a narrow film thickness range and cannot couple to any other modes. The coexistence of both the longitudinal and transverse coupling coefficients with the same order of magnitude, double degeneracy of the modes, and double degeneracy of the coupling coefficients near the cutoff are other unusual properties of the proposed structure.
... Evanescent waves have attracted considerable attention in nonlinear optics [23], optical sensors, and near-field optical microscopy [24][25][26][27]. The radiation force of an evanescent wave has been used to drive microparticles along the surface of dielectric prisms, channel waveguides, optical-planar waveguides, and optical nanofibers [28,29]. ...
Article
Full-text available
Evanescent waves of a guided mode carry both momentum and energy, which enables them to move small objects located on a waveguide surface. This optical force can be used for optical near-field manipulation, arrangement, and acceleration of particles. In this paper, using arbitrary beam theory, the optical force on a dielectric particle in the evanescent wave of a resonance waveguiding structure is investigated. Using Maxwell’s equations and applying the boundary conditions, all the field components and a generalized dispersion relation are obtained. An expression for the evanescent field is derived in terms of the spherical wave functions. Cartesian components of the radiation force are analytically formulated and numerically evaluated by ignoring the multiple scattering that occurs between the sphere and plane surface of the structure. Our numerical data show that both the horizontal and vertical force components and the forward particle velocity are enhanced significantly in the proposed resonance structure compared to those reported for three-layer conventional waveguides. Exerting stronger force on macro- and nanoparticles can be very useful to perform advanced experiments in solutions with high viscosity and experiments on biological cells. In addition, this resonance planar structure can be mounted on an inverted optical microscope stage for imaging the motion of nanoparticles especially when the particle collides and interacts with objects.
... On the other hand, in sensing/biosensing applications, low-loss ion-exchanged planar waveguides with embedded metal ions represented the core of integrated optical sensors for the analytical determination of biomolecules at the interfaces by an evanescent wave interrogation mechanism, exploiting different optical detection approaches such as those based on fluorescence or absorption [82][83][84][85][86][87][88][89][90]. Furthermore, these ion-exchange optical waveguides have been used as excitation systems for surface plasmon resonance (SPR) or localized surface plasmon resonance (LSPR) in noble metal thin films or NP arrays selectively deposited on them. ...
Article
Full-text available
Glasses, in their different forms and compositions, have special properties that are not found in other materials. The combination of transparency and hardness at room temperature, combined with a suitable mechanical strength and excellent chemical durability, makes this material indispensable for many applications in different technological fields (as, for instance, the optical fibres which constitute the physical carrier for high-speed communication networks as well as the transducer for a wide range of high-performance sensors). For its part, ion-exchange from molten salts is a well-established, low-cost technology capable of modifying the chemical-physical properties of glass. The synergy between ion-exchange and glass has always been a happy marriage, from its ancient historical background for the realisation of wonderful artefacts, to the discovery of novel and fascinating solutions for modern technology (e.g., integrated optics). Getting inspiration from some hot topics related to the application context of this technique, the goal of this critical review is to show how ion-exchange in glass, far from being an obsolete process, can still have an important impact in everyday life, both at a merely commercial level as well as at that of frontier research.
... Imaging applications include fluorescence in the range of optical signal detected as well. Waveguide evanescent field fluorescence microscopy [64,67] was utilized to investigate plasma membranes of living osteoblast cells, imaging plasma membranes of the cells with high axial resolution, and allowing time-lapse imaging to investigate cell morphology, the effect of membrane disrupters (Triton X-100), resulting membrane solubilization was visualized, a three-stage solubilization model was proposed for liposomes and supported lipid bilayers [66,69], the method was used on HEK293 cells [68] and to measure cell-substratum separation distance [63,65]. ...
Chapter
Novel optical biosensors in direct interaction with living cells open new avenues in the detection of given cell types, including pathogenic microorganisms, and in the measurement of cytotoxicity of given xenobiotics, such as biothreat agents, on suitably selected cell types or lines. Evanescent optical field based biosensors emerging for such applications are reviewed, covering surface plasmon and waveguide based formats, imaging setup, employing optical waveguides, and digital holographic microscopy. Commercial technologies, along with main pathogenic microorganisms (Bacillus anthracis, Escherichia coli, Listeria monocytogenes, Campylobacter jejuni, etc.) as analytes and cell lines (native tissue and tumor cells) as cytotoxicity effect subjects are highlighted.
... We carried out experiments to study the Au nano-disk arrays on top of waveguides. The waveguides were fabricated using ion-exchange technique on BK7 glass substrates, 32,33 and n eff of the TE 0 mode was tuned to match the simulated waveguide structure. The details of the experiment are presented in the supplemental material. ...
Article
We study and analyze various spectral features of gold nano-disk arrays coupled with the evanescent field of an optical waveguide using finite difference time domain simulations for comparison to our previous work on this topic [Jiang et al., J. Lightwave Technol. 27(13), 2264–2270 (2009)]. We find that the quadrupolar and dipolar peaks are both heavily dependent on the coherent interactions of the periodic array and show comparable performance for refractive index sensing applications. We also find sharp extinction peaks and dips in the simulated spectra, but they are not sensitive to the index change of the surface-bound layer. A detailed analysis on these sharp features reveals that the sharp extinction peaks are grating-induced quadrupolar modes and the dips represent the suppressed plasmon resonances caused by the photonic band gap.
Chapter
In the last few years Waveguide Evanescent Field Fluorescence (WEFF) and Scattering (WEFS) microscopy were developed which are alternatives to TIR and TIRF microscopy. Both technologies implement a slab waveguide-microscopy chip with a coupling grating. The technologies are described and compared to TIR and TIRF microscopy. The advantages of the waveguide method are clearly addressed. A brief history of the technology’s development and similar activities in the field are discussed. Application examples from both WEF microscopies follow: static distance mapping with a multimode waveguide, dynamic solubilisation studies of cell plasma membranes and the kinetic response of osteoblasts to trypsin (WEFF); bacteria sterilization as well as cell adhesion and granularity studies (WEFS). The combination of both methods is discussed and found not suitable. In order to mass fabricate the necessary waveguide chips with the grating an all-polymer-waveguide chip was developed. This should allow to bring the new microscopy methods to the interested scientific community.
Article
Advancement in the understanding of biomolecular interactions has benefited greatly from the development of surface-sensitive bionalaytical sensors. To further increase their broad impact, significant efforts are presently being made to enable label-free and specific biomolecule detection with high sensitivity, allowing for quantitative interpretation and general applicability at low cost. In this work, we have addressed this challenge by developing a waveguide chip consisting of a flat silica core embedded in a symmetric organic cladding with a refractive index matching that of water. This is shown to reduce stray light (background) scattering and allow for label-free detection of faint objects, such as individual 18 nm gold nanoparticles as well as sub-100 nm lipid vesicles. Measurements and theoretical analysis revealed that light-scattering signals originating from single surface-bound lipid vesicles enable characterization of their sizes without employing fluorescent lipids as labels. The concept is also demonstrated for label-free measurements of protein binding to and enzymatic digestion of individual lipid vesicles, enabling an analysis of the influence on the measured kinetics of the dye-labeling of lipids required in previous assays. Further, diffraction-limited imaging of cells (platelets) binding to a silica surface showed that distinct sub-cellular features could be visualized and temporally resolved during attachment, activation and spreading. Taken together, these results underscore the versatility and general applicability of the method, which due to its simplicity and compatibility with conventional microscopy setups may reach a wide spread in life science and beyond.
Article
Photoacoustic microscopy is being established not only for the in vivo imaging but for the in vitro cellular imaging. Especially, to obtain of the internal information of 3D cell culture cluster by using photoacoustic microscopy should be useful tools for in vitro imaging study. In this article, we summarize recent reports regarding to photoacoustic microscopy for cell imaging and future perspects.
Article
Previous studies have measured the distance between cells and the substratum at sites of adhesion via the emission of a fluorescent dye and waveguide methods. Here, we demonstrate a novel approach to measure the position of fluorescent dyes above a waveguide surface in the 10–200 nm distance range throughout an entire area, yielding a 2D dye distance map or a 3D contour plot. The dye is located in a multilayered Langmuir Blodgett (LB) film or in the plasma membrane of a cell. Waveguide evanescent field fluorescence (WEFF) images obtained using two different waveguide modes are employed allowing, with a simple mathematical approach, the calculation of dye distance maps. Ultra‐thin steps made using LB technology, adhesion distances and the bending of the plasma membrane between focal adhesions of osteoblastic cells are shown as examples. The errors are discussed. False color representation of a dye distance map with four osteoblasts. The inset represents an overexposed WEFF image of the same field of view. magnified image False color representation of a dye distance map with four osteoblasts. The inset represents an overexposed WEFF image of the same field of view.
Article
Full-text available
Keap1 mutations regulate Nrf2 activity and lead to chemoresistance in cancers. Yet the underlying molecular mechanisms of chemoresistance are poorly explored. By focusing and genotyping head and neck squamous cell carcinoma (HNSCC) that had available pathologic and clinical data, we provide evidence that Keap1 displays frequent alterations (17%) in HNSCC. Functional loss of Keap1 results in significant activation of Nrf2 and promotes cancer cell growth, proliferation, and elevated cancer stem cell (CSCs) self-renewal efficiency and resistance to oxidative stress. Furthermore, decreased Keap1 activity in these cells increased nuclear accumulation of Nrf2 and activation of the Notch pathway, causing enhanced transcriptional alterations of antioxidants, xenobiotic metabolism enzymes, and resistance to chemotherapeutic treatment. Limiting the Nrf2 activity by either Keap1 complementation or by Nrf2 silencing increased the sensitivity to chemotherapy in Keap1-mutated cells and repressed the CSC self-renewal activity. Our findings suggest that Keap1 mutations define a distinct disease phenotype and the Keap1-Nrf2 pathway is one of the leading molecular mechanisms for clinical chemotherapeutic resistance. Targeting this pathway may provide a potential and attractive personalized treatment strategy for overcoming chemotherapeutic resistance conferred by Keap1 mutations.
Article
Full-text available
Despite significant improvements in five-year survival rates due to early diagnosis and combination therapy, triple-negative breast cancer (TNBC) treatment remains a major challenge. Finding new and effective targets for diagnosis and drug therapy is urgent for TNBC patients. Jagged-1 (JAG1), one of the canonical ligands of the Notch signaling pathway, is involved in vascular budding and is a poor prognostic factor of TNBC. In this study, combined with quantitative real-time PCR, database analysis, animal experiments, and other means, JAG1 was confirmed to be related to the poor prognosis of TNBC patients. JAG1 was highly expressed in MDA-MB-231Bone (231B) cells, with stronger invasion and metastasis ability than MDA-MB-231 (231) cells. Treatment of human vascular endothelial cells (HUVEC) with TNBC conditioned medium showed that TNBC JAG1 promoted the angiogenesis of HUVEC. Next, we detected the exosomes extracted from TNBC conditioned medium and found that JAG1 promoted the exosome secretion from 231 cells via ALIX-RAB11A/RAB35. In addition, we also found that the exosomes from JAG1 overexpressed TNBC cells contained more long non-coding RNA (lncRNA) MALAT1, and MALAT1 promoted angiogenesis of HUVEC by targeting miR-140-5p. Finally, the angiogenesis-promoting effect of JAG1 in TNBC was further investigated by matrix gel assay. In conclusion, we reveal that JAG1 has a pro-invasion effect on TNBC and is involved in microenvironment angiogenesis by promoting exosome secretion and the MALAT1-miR-140-5p-JAG1/VEGFA pathway.
Article
Full-text available
Background: One of the most malignant tumors in men is prostate cancer that is still incurable due to its heterogenous and progressive natures. Genetic and epigenetic changes play significant roles in its development. The RNA molecules with more than 200 nucleotides in length are known as lncRNAs and these epigenetic factors do not encode protein. They regulate gene expression at transcriptional, post-transcriptional and epigenetic levels. LncRNAs play vital biological functions in cells and in pathological events, hence their expression undergoes dysregulation. Aim of review: The role of epigenetic alterations in prostate cancer development are emphasized here. Therefore, lncRNAs were chosen for this purpose and their expression level and interaction with other signaling networks in prostate cancer progression were examined. Key scientific concepts of review: The aberrant expression of lncRNAs in prostate cancer has been well-documented and progression rate of tumor cells are regulated via affecting STAT3, NF-κB, Wnt, PI3K/Akt and PTEN, among other molecular pathways. Furthermore, lncRNAs regulate radio-resistance and chemo-resistance features of prostate tumor cells. Overexpression of tumor-promoting lncRNAs such as HOXD-AS1 and CCAT1 can result in drug resistance. Besides, lncRNAs can induce immune evasion of prostate cancer via upregulating PD-1. Pharmacological compounds such as quercetin and curcumin have been applied for targeting lncRNAs. Furthermore, siRNA tool can reduce expression of lncRNAs thereby suppressing prostate cancer progression. Prognosis and diagnosis of prostate tumor at clinical course can be evaluated by lncRNAs. The expression level of exosomal lncRNAs such as lncRNA-p21 can be investigated in serum of prostate cancer patients as a reliable biomarker.
Article
Full-text available
MicroRNAs (miRNAs) are single-stranded non-coding RNA molecules that play a regulatory role in gene expression and cancer cell signaling. We previously identified miR-628-5p (miR-628) as a potential biomarker in serum samples from men with prostate cancer (PCa) (Srivastava et al. in Tumour Biol 35:4867–4873, 10.1007/s13277-014-1638-1, 2014). This study examined the detailed cellular phenotypes and pathways regulated by miR-628 in PCa cells. Since obesity is a significant risk factor for PCa, and there is a correlation between levels of the obesity-associated hormone leptin and PCa development, here we investigated the functional relationship between leptin and miR-628 regulation in PCa. We demonstrated that exposure to leptin downregulated the expression of miR-628 and increased cell proliferation/migration in PCa cells. We next studied the effects on cancer-related phenotypes in PCa cells after altering miR-628 expression levels. Enforced expression of miR-628 in PCa cells inhibited cell proliferation, reduced PCa cell survival/migration/invasion/spheroid formation, and decreased markers of cell stemness. Mechanistically, miR-628 binds with the JAG1-3′UTR and inhibits the expression of Jagged-1 (JAG1). JAG1 inhibition by miR-628 downregulated Notch signaling, decreased the expression of Snail/Slug, and modulated epithelial-mesenchymal transition and invasiveness in PC3 cells. Furthermore, expression of miR-628 in PCa cells increased sensitivity towards the drugs enzalutamide and docetaxel by induction of cell apoptosis. Collectively our data suggest that miR-628 is a key regulator of PCa carcinogenesis and is modulated by leptin, offering a novel therapeutic opportunity to inhibit the growth of advanced PCa.
Article
Full-text available
Notch signaling dysregulation encourages breast cancer progression through different mechanisms such as stem cell maintenance, cell proliferation and migration/invasion. Furthermore, Notch is a crucial driver regulating juxtracrine and paracrine communications between tumor and stroma. The complex interplay between the abnormal Notch pathway orchestrating the activation of other signals and cellular heterogeneity contribute towards remodeling of the tumor microenvironment. These changes, together with tumor evolution and treatment pressure, drive breast cancer drug resistance. Preclinical studies have shown that targeting the Notch pathway can prevent or reverse resistance, reducing or eliminating breast cancer stem cells. In the present review, we will summarize the current scientific evidence that highlights the involvement of Notch activation within the breast tumor microenvironment, angiogenesis, extracellular matrix remodeling, and tumor/stroma/immune system interplay and its involvement in mechanisms of therapy resistance.
Article
RNA molecules lacking capacity in protein translation, are known as non-coding RNAs (ncRNAs). Growth, differentiation and migration are influenced by ncRNAs in cells. The abnormal expression of ncRNAs contributes to development of diseases, especially cancer. On the other hand, EMT is a vital mechanism for cancer invasion and diffusion in body. In this manuscript, role of ncRNAs in EMT regulation and subsequent effect on cancer progression is investigated. The miRNAs regulate EMT by affecting signaling pathways including PI3K/Akt and PTEN to modulate cancer metastasis. Furthermore, miRNA and EMT interaction has close association with drug sensitivity of tumor cells. LncRNAs can affect EMT via targeting ZEB1/2, Twist and Snail among others and similarly, based on the impact on EMT, sensitivity of cancer cells to therapy increases or decreases. CircRNAs regulate both drug sensitivity and metastasis of cancers by affecting EMT mechanism. Noteworthy, circRNAs and lncRNAs are capable of sponging miRNAs in modulating EMT mechanism. Exosomes belong to extracellular vesicles with low size that can be secreted by cells in transferring genetic materials. The transfer of ncRNAs by exosomes is performed and they can also regulate EMT in cancer progression. Finally, ncRNAs regulating EMT mechanism are used for cancer diagnosis and prognosis.
Article
Initiation and development of cancer depend on multiple factors that mutations in genes and epigenetic level can be considered as important drivers. Epigenetic factors include a large family of members and understanding their function in cancer has been a hot topic. LncRNAs are RNA molecules with no capacity in synthesis of proteins, and they have regulatory functions in cells. LncRNAs are localized in nucleus and cytoplasm, and their abnormal expression is related to development of tumor. This manuscript emphasizes on the role of lncRNA H19 in various cancers and its association with tumor hallmarks. The function of lncRNA H19 in most tumors is oncogenic and therefore, tumor cells increase its expression for promoting their progression. LncRNA H19 contributes to enhancing growth and cell cycle of cancers and by EMT induction, it is able to elevate metastasis rate. Silencing H19 induces apoptotic cell death and disrupts progression of tumors. LncRNA H19 triggers chemo- and radio-resistance in cancer cells. miRNAs are dually upregulated/down-regulated by lncRNA H19 in increasing tumor progression. Anti-cancer agents reduce lncRNA H19 in impairing tumor progression and increasing therapy sensitivity. A number of downstream targets and molecular pathways for lncRNA H19 have been detected in cancers including miRNAs, RUNX1, STAT3, β-catenin, Akt2 and FOXM1. Clinical studies have revealed potential of lncRNA H19 as biomarker and its association with poor prognosis. LncRNA H19 can be transferred to cancer cells via exosomes in enhancing their progression.
Article
Epigenetic factors are critical regulators of biological and pathological mechanisms and they could interact with different molecular pathways. Targeting epigenetic factors has been an idea approach in disease therapy, especially cancer. Accumulating evidence has highlighted function of long non-coding RNAs (lncRNAs) as epigenetic factors in cancer initiation and development and has focused on their association with downstream targets. microRNAs (miRNAs) are the most well-known targets of lncRNAs and present review focuses on lncRNA-miRNA axis in malignancy and therapy resistance of tumors. LncRNA-miRNA regulates cell death mechanisms such as apoptosis and autophagy in cancers. This axis affects tumor metastasis via regulating EMT and MMPs. Besides, lncRNA-miRNA axis determines sensitivity of tumor cells to chemotherapy, radiotherapy and immunotherapy. Based on the studies, lncRNAs can be affected by drugs and genetic tools in cancer therapy and this may affect expression level of miRNAs as their downstream targets, leading to cancer suppression/progression. LncRNAs have both tumor-promoting and tumor-suppressor functions in cancer and this unique function of lncRNAs has complicated their implication in tumor therapy. LncRNA-miRNA axis can also affect other signaling networks in cancer such as PI3K/Akt, STAT3, Wnt/β-catenin and EZH2 among others. Notably, lncRNA/miRNA axis can be considered as a signature for diagnosis and prognosis in cancers.
Article
Breast carcinoma is one of the common causes of cancer-related mortality in women. Maternally expressed gene 3 (MEG3), a lncRNA located at 14q32, can be involved in carcinogenesis. In this study, we discovered that MEG3 was downregulated by CpG hypermethylation within its gene promoter. Functionally, treatment of breast cancer cells with the DNA methylation inhibitor 5-AzadC as well as silencing of DNA methyltransferase-1 (DNMT1) could decrease the abnormal hypermethylation of the MEG3 promoter, reverse MEG3 expression, inhibit cell proliferation and promote cell apoptosis. In addition, we found that MEG3 expression was negatively correlated with DNMT1. Mechanistically, MEG3 knockdown combined with 5-AzadC or sh-DNMT1 treatment restored the expression of Notch1 receptor, leading to the Notch1 pathway activation, and promoted the progression of epithelial mesenchymal transformation (EMT). Finally, the mice tumor model experiments showed that DNMT1 knockdown can increase MEG3 expression and inhibit tumor growth. Collectively, our findings uncovered that DNMT1-mediated MEG3 demethylation leads to MEG3 upregulation, which in turn inhibits the Notch1 pathway and EMT process in breast cancer.
Article
Epithelial-to-mesenchymal transition (EMT) mechanism is responsible for metastasis of tumor cells and their spread to various organs and tissues of body, providing undesirable prognosis. In addition to migration, EMT increases stemness and mediates therapy resistance. Hence, pathways involved in EMT regulation should be highlighted. STAT3 is an oncogenic pathway that can elevate growth rate and migratory ability of cancer cells and induce drug resistance. The inhibition of STAT3 signaling impairs cancer progression and promotes chemotherapy-mediated cell death. Present review focuses on STAT3 and EMT interaction in modulating cancer migration. First of all, STAT3 is an upstream mediator of EMT and is able to induce EMT-mediated metastasis in brain tumors, thoracic cancers and gastrointestinal cancers. Therefore, STAT3 inhibition significantly suppresses cancer metastasis and improves prognosis of patients. EMT regulators such as ZEB1/2 proteins, TGF-β, Twist, Snail and Slug are affected by STAT3 signaling to stimulate cancer migration and invasion. Different molecular pathways such as miRNAs, lncRNAs and circRNAs modulate STAT3/EMT axis. Furthermore, we discuss how STAT3 and EMT interaction affects therapy response of cancer cells. Finally, we demonstrate targeting STAT3/EMT axis by anti-tumor agents and clinical application of this axis for improving patient prognosis.