ArticlePDF AvailableLiterature Review

Natural products and long noncoding RNA signatures in gallbladder cancer: a review focuses on pathogenesis, diagnosis, and drug resistance

Authors:
  • University of Hertfordshire hosted by Global Academic Foundation

Abstract and Figures

Gallbladder cancer (GBC) is an aggressive and lethal malignancy with a poor prognosis. Long noncoding RNAs (lncRNAs) and natural products have emerged as key orchestrators of cancer pathogenesis through widespread dysregulation across GBC transcriptomes. Functional studies have revealed that lncRNAs interact with oncoproteins and tumor suppressors to control proliferation, invasion, metastasis, angiogenesis, stemness, and drug resistance. Curcumin, baicalein, oleanolic acid, shikonin, oxymatrine, arctigenin, liensinine, fangchinoline, and dioscin are a few examples of natural compounds that have demonstrated promising anticancer activities against GBC through the regulation of important signaling pathways. The lncRNAs, i.e., SNHG6, Linc00261, GALM, OIP5-AS1, FOXD2-AS1, MINCR, DGCR5, MEG3, GATA6-AS, TUG1, and DILC, are key players in regulating the aforementioned processes. For example, the lncRNAs FOXD2-AS1, DILC, and HOTAIR activate oncogenes such as DNMT1, Wnt/β-catenin, BMI1, and c-Myc, whereas MEG3 and GATA6-AS suppress the tumor proteins NF-κB, EZH2, and miR-421. Clinically, specific lncRNAs can serve as diagnostic or prognostic biomarkers based on overexpression correlating with advanced TNM stage, metastasis, chemoresistance, and poor survival. Therapeutically, targeting aberrant lncRNAs with siRNA or antisense oligos disrupts their oncogenic signaling and inhibits GBC progression. Overall, dysfunctional lncRNA regulatory circuits offer multiple avenues for precision medicine approaches to improve early GBC detection and overcome this deadly cancer. They have the potential to serve as novel biomarkers as they are detectable in bodily fluids and tissues. These findings enhance gallbladder treatments, mitigating resistance to chemo- and radiotherapy.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
Naunyn-Schmiedeberg's Archives of Pharmacology
https://doi.org/10.1007/s00210-024-03279-1
REVIEW
Natural products andlong noncoding RNA signatures ingallbladder
cancer: areview focuses onpathogenesis, diagnosis, anddrug
resistance
HananElimam1 · NoraA.A.Alhamshry1· AbdulrahmanHatawsh2· NourhanElfar3,4· RewanMoussa5·
AbdullahF.Radwan6· MaiA.Abd‑Elmawla7· AkramM.Elkashlan1· MohamedBakrZaki1·
MustafaAhmedAbdel‑Reheim8,9 · OsamaA.Mohammed10· AhmedSDoghish11,12
Received: 28 May 2024 / Accepted: 2 July 2024
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2024
Abstract
Gallbladder cancer (GBC) is an aggressive and lethal malignancy with a poor prognosis. Long noncoding RNAs (lncRNAs)
and natural products have emerged as key orchestrators of cancer pathogenesis through widespread dysregulation across
GBC transcriptomes. Functional studies have revealed that lncRNAs interact with oncoproteins and tumor suppressors to
control proliferation, invasion, metastasis, angiogenesis, stemness, and drug resistance. Curcumin, baicalein, oleanolic acid,
shikonin, oxymatrine, arctigenin, liensinine, fangchinoline, and dioscin are a few examples of natural compounds that have
demonstrated promising anticancer activities against GBC through the regulation of important signaling pathways. The lncR-
NAs, i.e., SNHG6, Linc00261, GALM, OIP5-AS1, FOXD2-AS1, MINCR, DGCR5, MEG3, GATA6-AS, TUG1, and DILC,
are key players in regulating the aforementioned processes. For example, the lncRNAs FOXD2-AS1, DILC, and HOTAIR
activate oncogenes such as DNMT1, Wnt/β-catenin, BMI1, and c-Myc, whereas MEG3 and GATA6-AS suppress the tumor
proteins NF-κB, EZH2, and miR-421. Clinically, specific lncRNAs can serve as diagnostic or prognostic biomarkers based
on overexpression correlating with advanced TNM stage, metastasis, chemoresistance, and poor survival. Therapeutically,
targeting aberrant lncRNAs with siRNA or antisense oligos disrupts their oncogenic signaling and inhibits GBC progression.
Overall, dysfunctional lncRNA regulatory circuits offer multiple avenues for precision medicine approaches to improve early
GBC detection and overcome this deadly cancer. They have the potential to serve as novel biomarkers as they are detectable
in bodily fluids and tissues. These findings enhance gallbladder treatments, mitigating resistance to chemo- and radiotherapy.
Keywords LncRNA· Natural products· Gallbladder cancer· Drug resistance· Diagnosis· Prognosis
* Hanan Elimam
Hanan.Elimam@fop.usc.edu.eg
* Mustafa Ahmed Abdel-Reheim
m.ahmed@su.edu.sa
1 Department ofBiochemistry, Faculty ofPharmacy,
University ofSadat City, SadatCity32897, Egypt
2 Biotechnology School, 26th ofJuly Corridor, Sheikh Zayed
City, Nile University, Giza12588, Egypt
3 School ofLife andMedical Sciences, University
ofHertfordshire Hosted byGlobal Academic Foundation,
New Administrative Capital, Cairo11578, Egypt
4 Egyptian Drug Authority (EDA), Ministry ofHealth
andPopulation, Cairo11567, Egypt
5 Faculty ofMedicine, Helwan University, Cairo11795, Egypt
6 Department ofBiochemistry, Faculty ofPharmacy, Egyptian
Russian University, Cairo11829, Egypt
7 Department ofBiochemistry, Faculty ofPharmacy, Cairo
University, Cairo, Egypt
8 Department ofPharmaceutical Sciences, College
ofPharmacy, Shaqra University, 11961Shaqra, SaudiArabia
9 Department ofPharmacology andToxicology, Faculty
ofPharmacy, Beni-Suef University, BeniSuef62521, Egypt
10 Department ofPharmacology, College ofMedicine,
University ofBisha, 61922Bisha, SaudiArabia
11 Department ofBiochemistry, Faculty ofPharmacy, Badr
University inCairo (BUC), BadrCity, Cairo11829, Egypt
12 Faculty ofPharmacy (Boys), Al-Azhar University,
NasrCity11231, Cairo, Egypt
Naunyn-Schmiedeberg's Archives of Pharmacology
Abbreviations
GBC Gallbladder cancer
lncRNAs Long noncoding RNAs
EMT Epithelial–mesenchymal transition
PRC2 Polycomb repressive complex 2
HOTAIR HOX transcript antisense RNA
XIST X-inactive specific transcript
FOXM Forkhead box protein M1
AKT2 AKT serine/threonine kinase 2
MALAT1 Metastasis-associated lung adenocarci-
noma transcript 1
EZH2 Enhancer of zeste homolog 2
LATS2 Large tumor suppressor kinase 2
NF-κB Nuclear factor kappa-light-chain-enhancer
of activated B cells
CSCs Cancer stem cells
MEG3 Maternally expressed 3
CEA Carcinoembryonic antigen
MYLK-AS1 Myosin light-chain kinase antisense RNA
1
GBCDRlnc1 Gallbladder cancer drug resistance-associ-
ated lncRNA1
MMP Matrix metalloproteinase
Dox Doxorubicin
MAPK Mitogen-activated protein kinase
AFAP1-AS1 Actin filament–associated protein 1 anti-
sense RNA 1
ANRIL Antisense noncoding RNA in the INK4
locus
BMI1 B lymphoma Mo-MLV insertion region 1
homolog
GALM GBC associated with liver metastasis
HEGBC Highly expressed in gallbladder carcinoma
H19 Imprinted maternally expressed transcript
CBX4 Chromobox4
p38 MAPK p38 mitogen-activated protein kinases
JNK c-Jun N-terminal kinase
ERK Extracellular signal-regulated kinases
MINCR Myocardial infarction–associated
transcript
NEAT1 Nuclear paraspeckle assembly transcript 1
PVT1 Plasmacytoma variant translocation 1
HK2 Hexokinase 2
SSTR5-AS1 Somatostatin receptor 5 antisense RNA 1
NONO Non-POU domain containing octamer
binding protein
ROR Regulator of reprogramming
TUG1 Taurine upregulated gene 1
CCAT1 Colon cancer associated transcript 1
FIRRE Functional intergenic repeating RNA
element
DILC Downregulated in liver cancer stem cells
DGCR5 DiGeorge syndrome critical region gene 5
ZO-1 Zonula occludens 1
FOXD2-AS1 FOXD2 adjacent opposite strand RNA 1
DNMT1 DNA methyltransferase 1
MLH1 MutL homolog 1
HGBC Highly expressed in gallbladder carcinoma
HuR Hu antigen R
HOXA-AS2 HOXA cluster antisense RNA 2
LINC00152 Long intergenic non-protein coding RNA
152
PI3K Phosphoinositide 3-kinase
UCA1 Urothelial cancer associated 1
EZH2 Enhancer of zeste homolog 2
LET Low expression in tumor
PAGBC Prognosisassociated gallbladder cancer
SOC Suppressor of cytokine signaling
TGF Transformed growth factor
ZEB Zinc Finger E-Box Binding Homeobox
Introduction
The malignant tumor known as gallbladder cancer (GBC)
is characterized by aggressiveness and a high failure rate.
Globally, GBC is the most prevalent biliary tract cancer,
making up around two-thirds of all biliary tract cancer
cases (Krinsky 2004). Patients with advanced, incurable
GBC have a dismal median overall survival of less than 12
months and a 5-year survival rate of less than 5% due to
late-stage diagnosis and inherently aggressive disease biol-
ogy due to late-stage diagnosis, an aggressive course of the
disease, limited treatment options, and significant recurrence
rates (Hundal and Shaffer 2014, Shahin etal. 2023). Unfor-
tunately, in the early stages of the disease, the anatomical
location of the gallbladder causes non-specific symptom
presentation. More than 90% of GBC cases are diagnosed
at an advanced stage, making surgery impossible (Zou etal.
2013). Recently available cytotoxic chemotherapy regimens,
such as those involving gemcitabine and cisplatin, only mar-
ginally improve patient survival without raising quality of
life (Arslan and Yalcin 2014, Adamska etal. 2017). These
dark statistics highlight how urgently we must learn more
about the molecular pathophysiology of GBC to develop
targeted treatments.
Emerging evidence suggests that natural products from
medicinal plants and herbs may hold promise for treating
GBC by targeting key oncogenic pathways. Several natural
products such as curcumin, baicalein, oleanolic acid, shi-
konin, oxymatrine, arctigenin, liensinine, fangchinoline,
and dioscin have shown promising anticancer effects against
GBC by modulating key signaling pathways involved in pro-
liferation, apoptosis, metastasis, and drug resistance (Fig.1).
Moreover, long noncoding RNAs (lncRNAs) have become
increasingly important in the study of cancer because they
Naunyn-Schmiedeberg's Archives of Pharmacology
are complex regulators of gene expression that affect every
characteristic of malignant transformation and progression
(Tano and Akimitsu 2012, Worku etal. 2017, Wang etal.
2023). LncRNAs represent large and heterogeneous non-
protein coding transcripts exceeding 200 nucleotides gener-
ated from intergenic stretches or overlapping antisense tran-
scripts of protein-coding (Nobili etal. 2017, Mattick etal.
2023). Through dynamic interactions with DNA, RNAs, and
proteins, tens of thousands of lncRNAs—previously con-
sidered “junk DNA”—are known to participate in a variety
of biological processes (Scacalossi etal. 2019; Tyagi etal.
2018). Aberrant expression of lncRNAs is increasingly rec-
ognized as a hallmark of many cancers, orchestrating onco-
genesis through perturbed regulation of proto-oncogenes,
tumor suppressors, the cell cycle, and survival mediators
(Do and Kim 2018, Taniue and Akimitsu 2021).
Initial RNA sequencing analyses across independent
patient cohorts have demonstrated widespread dysregula-
tion of lncRNAs in GBC compared to normal gallbladder
tissue controls (Khandelwal etal. 2017, Niu etal. 2020,
Yang etal. 2023). Aberrant lncRNA expression manifests
at early preneoplastic stages and escalates with advancing
tumor progression. Functional studies have shown that lncR-
NAs interact with important signaling proteins to regulate
a variety of processes, including GBC growth, epithelial-
mesenchymal transition (EMT), invasion, metastasis, angio-
genesis, stemness, and therapeutic resistance (Jeon and Lee
2017, Hao etal. 2019, Niu etal. 2020, Hamidi etal. 2022).
Therefore, lncRNAs could serve as both attractive targets for
new GBC treatments and reliable biomarkers for diagnosis
and prognosis.
While tests for biomarkers like CEA, RCAS1, CA19-9,
CA125, CA242, and CA15-3 have been evaluated to diag-
nose GBC, they demonstrate limited sensitivity (61–87.5%).
As a result, the diagnosis and prognosis assessment of GBC
remains challenging, and it continues to have a poor prog-
nosis. Of the protein biomarkers studied, CA19-9 shows
the most promise and is advised as an important marker for
GBC diagnosis and disease monitoring. Still, more research
is warranted to improve the early detection and management
of this aggressive cancer (Shahin etal. 2023).
In this comprehensive review, we encapsulate current
concepts on lncRNA-mediated orchestration of GBC patho-
genesis, with particular emphasis on clinical translation.
Fig. 1 Widely investigated
chemical structures of natural
products
Naunyn-Schmiedeberg's Archives of Pharmacology
We outline lncRNA biogenesis, functional taxonomy, and
mechanisms enabling gene expression control in GBC cells.
Next, we detail major natural products and lncRNAs steer-
ing GBC growth, dissemination, and therapeutic resistance
through the regulation of pivotal onco- and tumor suppressor
proteins. Finally, we appraise the diagnostic, prognostic, and
therapeutic opportunities conferred by lncRNA signatures at
tissue and circulating levels to combat this lethal malignancy
through precision medicine approaches.
Natural products andGBC
Curcumin
Curcumin is an extract from turmeric and a naturally occur-
ring phytochemical. Clinical trials on curcumin have been
conducted on the basis of an array of invitro and invivo
evidence. Important signaling pathways involved in cellu-
lar functions are modulated by curcumin. Curcumin may
act as a multitarget medication because it can alter several
cellular pathways implicated in the development of cancer.
This has special relevance to the treatment of neoplastic
diseases (Irving etal. 2011, Gupta etal. 2013). Ono etal.
elucidated that curcumin exerts beneficial effects in GBC
through several mechanisms, including decreased Bcl-2
function, decreased AKT-mTOR activity, and increased
mitogen-activated protein (MAP) kinase activity, and pro-
motes G2/M arrest (Ono etal. 2013a). These findings offer
a molecular explanation for curcumin’s possible application
in the management of GBC. Another study investigated
that curcumin controls the Bcl-2/Bax ratio and triggers
the production of cleaved caspase-3, leading to apoptosis
in GBC-SD cells (Liu etal. 2013). Depending on the sig-
nal transduction mechanisms that are activated, curcumin
displays varying activities against apoptosis and cell cycle
progression. Additionally, curcumin can block downstream
signaling pathways that are activated by Src and Ras, indi-
cating that it may be useful in treating human cancers that
have activated Src or Ras (Ono etal. 2013b). Additionally,
curcumin lowered the expression of CXCR4 invitro and
invivo, which likely decreased metastasis by suppressing
the stromal cell–derived factor-1/CXCR4 signaling pathway
(Gu etal. 2019).
Baicalein
Baicalein is widely used and has a variety of pharmacologi-
cal actions. GBC cell metastasis was suppressed by baicalein
treatment. Moreover, baicalein inhibited GBC cell growth
and metastasis via downregulation of zinc finger protein
X-linked (ZFX) expression (Liu etal. 2015). By focusing on
the PTEN/PI3K/AKT signaling pathway, sinensetin flavone
has been shown to demonstrate strong anticancer efficacy
against drug-resistant human GBC through the former axis
(Huang etal. 2020). Song and his colleagues reported that
the flavone casticin causes G0/G1 arrest and apoptosis in
GBC and that casticin may be a new and useful treatment
for this disease (Song etal. 2017b). Concurrently, the fla-
vone hispidulin exhibits anti-malignant effects on GBC by
inhibiting HIF-1α signaling. Hispidulin attenuates the resist-
ance to chemotherapeutic drugs such as gemcitabine and
5-fluorouracil by downregulating HIF-1α (Gao etal. 2015).
The flavonoid icariin exhibits anticancer action and enhances
the antitumor activity of gemcitabine in GBC by inhibiting
NF-κB activity. For patients with GBC, the combination of
gemcitabine and icariin may provide a more effective course
of treatment (Zhang etal. 2013). It was documented that the
flavonoid isorhamnetin inhibited the growth and spread of
GBC cells, induced apoptosis, and stopped the G2/M phase
in GBC cells by blocking the PI3K/AKT signaling pathway
(Zhai etal. 2021).
Triterpenoids
Triterpenoids are natural compounds that are formed by the
cyclization of squalene, resulting in the production of either
3-deoxytriterpenes (hydrocarbons) or 3-hydroxytriterpenes.
Triterpenoids are derived from two categories of triterpenes
by the process of modifying their carbon framework to yield
oleanolic acid, pachymic acid, and shikonin. Triterpenoids
are extensively found in higher plants and are significant
due to their diverse structures and wide variety of biological
functions (Özdemir and Wimmer 2022, Zang etal. 2022).
Oleanolic acid, a naturally derived triterpenoid, demon-
strates promising anticancer effects in many types of tumor
cell lines. Oleanolic acid effectively suppresses the growth
of GBC cells in a way that is influenced by both the dos-
age and duration of treatment. This study provided evidence
that oleanolic acid operates via the mitochondrial apoptotic
pathway. Furthermore, this medication suppressed the pro-
liferation of tumors in nude mice and hindered the growth
of GBC cells via controlling apoptosis and the cell cycle
process. Therefore, oleanolic acid shows potential as a ben-
eficial medication for adjuvant chemotherapy in gallblad-
der malignancies (Tang etal. 2022). In another study, the
impact of pachymic acid on tumors in human GBC cells
was investigated. This study demonstrated that pachymic
acid has the ability to suppress the development of GBC
via affecting the AKT and ERK signaling pathways. These
findings support pachymic acid as a potential treatment for
GBC (Chen etal. 2015). In addition, shikonin, a naturally
occurring compound obtained from the roots of Lithosper-
mum erythrorhizon, is known for its anticancer properties. It
was found that shikonin decreased the growth of GBC cells.
Additionally, it induced apoptosis and caused G0/G1 phase
Naunyn-Schmiedeberg's Archives of Pharmacology
arrest in the GBC cells through the JNK signaling pathway.
These findings demonstrate that shikonin has anticancer
properties on GBC cells through the stimulation of apopto-
sis and modulation of the cell cycle. Collectively, shikonin
has the potential to serve as a new and secure chemotherapy
drug for the management of GBC (Zhai etal. 2017, Wang
etal. 2020a).
Alkaloids
Alkaloids are chemical compounds that play an important
role in developing innovative medications since they provide
a rich resource for creating these new medicines. Studies
conducted on a variety of cancers, both invitro and invivo,
have shown promising outcomes for a number of alkaloids
that are derived from medicinal plants and herbs (Mondal
etal. 2019). Through research conducted both in the lab-
oratory and on animals, oxymatrine (OM) was shown to
have a potent anticancer effect on cells derived from GBC.
Additionally, it is believed that the anticancer benefits of
OM are likely derived from its ability to limit cell growth
and promote programmed cell death. The downregulation
of NF-κB expression and the activation of the mitochon-
dria-mediated apoptotic pathway are both necessary steps
in order to accomplish this (Wu etal. 2014b). Arctigenin,
which is derived from the seeds of Centaurea schischkinii,
was shown to have potent cytotoxic effects on CaCo-2 colon
cancer cell lines, according to the findings of an independent
analysis. Arctigenin was shown to be responsible for a sig-
nificant reduction in the expression of EGFR in GBC tissue,
which in turn led to the induction of cellular senescence. For
this purpose, the RAF-MEK-ERK signaling pathway was
altered. The results of these experiments showed that the
suppression of EGFR led to the cessation of cell division
during the G1/G0 phase and the induction of programmed
cell death (Zhang etal. 2017a).
Experiments conducted invitro and invivo demonstrated
that liensinine has a substantial impact on reducing GBC
cell proliferation. The effects of lisinine on cell growth and
proliferation invitro were dose- and time-dependent and
demonstrated a considerable reduction in both of these pro-
cesses. Lisinine was able to effectively suppress the forma-
tion of tumors invivo environments. A study conducted by
Shen etal. suggests that lisinine has the potential to cause
GBC cells to shut down in the G2/M phase. This is accom-
plished by increasing the quantities of CDK1 and Cyclin B1
protein in the body. Through the inhibition of the expression
of essential proteins such as AKT, p-AKT, PI3K, and ZFX,
Liensinine was able to have an impact on the advancement
of the GBC cell cycle and apoptosis rates (Shen etal. 2019).
Finally, it was shown by Li etal. that fangchinoline,
which is a bisbenzylisoquinoline alkaloid, inhibits the
growth and reproduction of GBC cells in a dose-dependent
manner. The results of flow cytometry, TUNEL staining,
and Hoechst staining gave further proof that fangchinoline
is capable of inducing cell death in GBCs. In GBC cells,
the anti-apoptotic PI3K/Akt/XIAP axis was significantly
suppressed by fangchinoline treatment, as shown by further
research (Li etal. 2022a).
Phenolic compounds
In the last 10 years, the antioxidant properties of dietary
plant polyphenols have garnered considerable attention for
their possible positive effects on health. Epidemiological
studies and meta-analyses have shown that a diet rich in
plant polyphenols reduces the risk of cancer, heart disease,
diabetes, osteoporosis, and neurological disorders when fol-
lowed over the long term. There is strong epidemiological
evidence that phenolics have anticarcinogenic, anti-inflam-
matory, and antioxidant properties, which make them pow-
erful tools in the fight against chronic diseases (Rahman
etal. 2022).
While both the crude and anthocyanin-rich Calafate fruit
extracts exhibited a reduction in the migratory potential and
invitro viability of gallbladder (G415) and stomach (AGS)
cancer cell lines, the anthocyanin-rich extract showed much
greater efficacy. The significant presence of delphinidin in
this extract, which is recognized for its potent antioxidant
properties in comparison to those of other phenolic and
anthocyanidin chemicals, is likely the primary factor con-
tributing to this discovery. Hence, the findings of this study
provide significant insights for future research, indicating
that Calafate fruits might serve as a valuable resource for
the creation of phytopharmaceutical compounds to address
human health concerns. To get a deeper understanding of
the positive impacts of Calafate fruits and their natural
components, such as phenolics or alkaloids that likely work
together in a favorable way, it is necessary to undertake a
comprehensive analysis of the secondary metabolites found
in the crude and anthocyanin-rich extracts of these fruits
(Calderón-Reyes etal. 2020).
Tea polyphenols (TPs) possess anti-inflammatory proper-
ties, combat cancer, inhibit blood clot formation, eradicate
microorganisms, and promote vasodilation. Wang etal. dis-
covered that TPs had a strong inhibitory effect on the prolif-
eration of GBC cells both invivo and invitro. Researchers
examined the impact of TPs on GBC cell apoptosis and cell
cycle arrest, which are two important indications of anti-
cancer activity. TPs may induce apoptosis that is associated
with mitochondria and halt the cell cycle of GBC cells in
the S phase. Furthermore, the research revealed that TPs
did not have any detrimental effects on healthy gallblad-
der cells. Based on their research, it has been shown that
TPs are harmless chemicals that have great promise for
treating GBC. The mitochondrial membrane potential of
Naunyn-Schmiedeberg's Archives of Pharmacology
NOZ-treated cells was also reduced after 48 h of TP treat-
ment, indicating that apoptosis linked to mitochondria con-
tributed to the demise of GBC cells. The expression of Bax
and Bcl2 was upregulated and downregulated, respectively,
after TP treatment. The release of cytochrome c into the
cytosol increased, whereas the membrane potential of the
mitochondria decreased, which was associated with altera-
tions in protein expression. Following treatment with TPs,
there was an observed rise in levels of cleaved caspase-3
and PARP. The Caspases family has a role in mitochondria-
induced apoptosis (Wang etal. 2018).
Numerous biological and pharmacological effects have
been shown for Dioscin (DSN), a plant glucoside sapo-
nin derived from Dioscorea zingiberensis Wright and
Dioscorea nipponica Makino. The anti-fungal, anti-viral,
and hepatoprotective effects of DSN have been shown in
earlier research. Because of its anticancer benefits on lung,
colon, breast, and stomach cancers, DSN has lately garnered
much interest. The review findings demonstrated that DSN
effectively halted the migration and proliferation of GBC
cells. In addition, DSN caused GBC cell death via means
of apoptotic signaling that relies on mitochondria. Tests
for glutathione (GSH) and reactive oxygen species (ROS)
showed that ROS scavengers entirely blocked DSN-induced
migration and apoptosis, suggesting that ROS are critical for
the development of gastric cancer. Analysis using Western
blotting demonstrated a significant decrease in AKT activity
after DSN administration. Furthermore, it was shown that
AKT apoptosis, but not migration, was boosted or abolished
when AKT was either inhibited or expressed ectopically.
Additionally, the research demonstrated that ROS are related
to the PI3K/AKT pathway and that DSN caused cell death
by controlling ROS-mediated PI3K/AKT signaling (Song
etal. 2017a).
The molecular choreography oflncRNA biogenesis
andfunction
LncRNAs are a diverse class of RNA transcripts longer than
200 nucleotides that are transcribed by RNA polymerase II
but do encode proteins (Mattick etal. 2023). The genomic
organization of lncRNA genes is similar to that of protein-
coding genes; both have 3 poly(A) tails and exons, introns,
and 5 caps (Ulitsky and Bartel 2013, Statello etal. 2020).
However, because they do not contain open reading frames,
lncRNAs avoid being translated into amino acid sequences.
Rather, complex molecular interactions involving proteins,
RNAs, and DNA enable lncRNAs to control the gene expres-
sion patterns that underpin tissue homeostasis, cell differ-
entiation, and embryonic development (Mangiavacchi etal.
2023). Aberrant lncRNA expression features prominently
across human cancer transcriptomes, with mutations in criti-
cal lncRNA loci implicated as driver events in tumorigenesis
(Jiang etal. 2019). Therefore, understanding the unique bio-
synthesis pathways and molecular mechanisms controlling
the functionality of lncRNAs can provide insights into both
healthy and diseased states of biology.
LncRNA biogenic pathways
LncRNA regulation has been shown to play important roles
in several classical pathways. The majority of these canoni-
cal pathways involve direct mechanisms connected to tran-
scriptional, post-transcriptional, and chromatin processes
(Fig.2). One major canonical pathway involves lncRNAs
regulating gene expression by interacting with chromatin
remodeling complexes to alter chromatin state and acces-
sibility. Chromosome accessibility and structure modula-
tion by lncRNAs enable them to affect subsequent genetic
programs. For example, in female placental mammals, the
lncRNA XIST coats and modifies one X chromosome’s
chromatin state to carry out the essential function of X
chromosome inactivation (Li etal. 2022b). The Polycomb
repressive complex 2 (PRC2), which sets down repressive
histone marks along chromosomes, is one of the chromatin
remodeling complexes that are recruited by XIST (Shi etal.
2017, Colognori etal. 2019).
Another example includes the lncRNA HOTAIR which
silences chromosomal domains by binding to and scaffold-
ing between PRC2 and LSD1 histone-modifying complexes,
enabling coordinated histone H3 lysine 27 trimethylation
and lysine 4 demethylation (Hajjari and Salavaty 2015,
Pawłowska etal. 2017). Additional illustrations of this
canonical pathway include lncRNAs that guide chroma-
tin modifiers, like PRC2, or histone-modifying enzymes
to specific genomic loci resulting in activation or silenc-
ing, respectively (Meseure etal. 2015, Zhang etal. 2021).
In addition to manipulating chromatin, lncRNAs are also
known to directly regulate gene expression at the transcrip-
tional level by interacting with key transcription factors and
other components of the basic transcriptional machinery.
For example, the lncRNA Evf-2 works cooperatively with
the homeodomain protein Dlx2 to form an activating com-
plex that induces transcription of downstream target genes
involved in important neural development pathways (Kaik-
konen etal. 2011). Lastly, another important mechanism
by which lncRNAs regulate gene expression is the complex
post-transcriptional regulation of RNA processing, transport,
stability, and translation (Kung etal. 2013, Doghish etal.
2024b).
In contrast to these widely described canonical inter-
actors and pathways, studies have also uncovered non-
canonical modes by which lncRNAs execute important
cellular functions. These non-traditional mechanisms dem-
onstrate the remarkable versatility of lncRNA signaling.
Through a variety of allosteric and stochastic interactions,
Naunyn-Schmiedeberg's Archives of Pharmacology
cytoplasmic and nuclear lncRNAs have emerged as impor-
tant modulators of protein activity instead of directly
affecting local chromatin or transcription (Guh etal.
2020). For instance, conformational changes induced upon
lncRNA binding can unlock inactive states of proteins, as
demonstrated by the lncRNA which binds to TLS, mak-
ing TLS undergo a conformational shift that enables its
amino terminus to block p300 and CBP’s histone acetyl-
transferase activity, which in turn suppresses gene expres-
sion (Fu 2014). Conversely, lncRNAs can obstruct active
sites or disrupt multimerization interfaces, as exemplified
by MEG3 binding and repressing p53 tetramerization to
attenuate downstream signaling (Zhang etal. 2023). Fur-
thermore, lncRNAs have emerged as critical controllers of
cellular pathways by acting as molecular decoys that soak
up protein or RNA regulators (Gao etal. 2020, Giuliani
etal. 2023). For example, NORAD sequesters PUMILIO
proteins to influence mitotic progression by acting as a
potent stoichiometric sponge (Schmitt and Chang 2016,
Liu and Chen 2022). By covering binding sites, cytoplas-
mic long noncoding RNAs (lncRNAs) like 1/2-sbsRNAs
can function as mRNA masks, preventing microRNA-
directed cleavage of transcripts associated with funda-
mental metabolism (Jafari-Raddani etal. 2022). Overall,
by titrating away regulators from intended targets, these
lncRNA decoys provide a customizable buffering system
to fine-tune pathways.
For example, cytoplasmic lncRNAs harbor binding sites
that sequester miRNAs away from their mRNA targets as
competitive “sponges” to impart an additional layer of tar-
get specificity (43). Other examples include lncRNAs that
interact with spliceosome machinery components to directly
modulate splicing and change the isoform expression of cru-
cial genes (44). Collectively, through interacting with vari-
ous RNA and protein regulators, lncRNAs hold vital roles in
cohesively ensuring proper mRNA and protein production.
LncRNA functions
LncRNAs carry out a multitude of gene regulatory functions
through diverse molecular mechanisms across numerous
cancers such as CRC, breast cancer, gastric cancer, and lung
cancer. They can serve as decoys to bind and sequester pro-
teins or microRNAs, act as scaffolds to bring together multi-
ple regulatory components, and guide chromatin and protein
complexes to target genes (Ratti etal. 2020). Through these
mechanisms, lncRNAs control transcriptional programs,
modulate chromatin architecture, and regulate down-
stream gene expression (Alvarez-Dominguez and Lodish
2017). The dysregulated expression of lncRNAs promotes
Fig. 2 LncRNAs have a crucial impact on the alteration and control
of gene expression. LncRNAs exert influence on mRNA stability
in various ways. Examples include direct binding to target mRNAs,
interactions with RNA-binding proteins, competition with miR-
NAs as competing endogenous RNAs, regulation of mRNA decay
pathways, and interference with transcription. LncRNAs can finely
regulate gene expression after transcription by either stabilizing or
degrading certain mRNAs.
Naunyn-Schmiedeberg's Archives of Pharmacology
tumorigenesis by stimulating proliferation, invasion, metas-
tasis, angiogenesis, immune evasion, and drug resistance
(Zhou etal. 2022). Cancer-specific lncRNA expression pat-
terns make them attractive biomarker candidates (Eptam-
initaki etal. 2022). Knockdown of oncogenic lncRNAs can
inhibit cancer growth and viability (Tsai etal. 2018). In
GBC, lncRNAs such as HOTAIR, H19, and MALAT1 are
overexpressed and drive oncogenesis by altering prolifera-
tion, chemoresistance, and metastasis (Pérez-Moreno etal.
2021, Schwerdtfeger etal. 2021). Overall, lncRNAs are criti-
cal gene regulators and play indispensable functional roles
in the initiation, progression, and metastasis of numerous
cancers, including GBC.
Role oflncRNAs inthepathogenesis ofGBC
Prior research has demonstrated the important involvement
of lncRNAs in the biology of cancer, and abnormalities
in lncRNAs are linked to several forms of human malig-
nancies. So far, a considerable number of lncRNAs have
been described to be involved with GBC (Pérez-Moreno
etal. 2021, Doghish etal. 2024a). Here the review article
demonstrated the effect of lncRNA expression alterations on
the pathogenesis of GBC.
Upregulated lncRNAs inGBC
Many lncRNAs have been identified to participate in GBC
pathogenesis through many pathways. Gao et. al found that
the lncRNA FOXD2-AS1 is capable of recruiting the meth-
yltransferase DNMT1 to the promoter region of the MLH1
gene, leading to the suppression of MLH1 transcription.
This process ultimately speeds up the advancement of GBC
(Gao etal. 2021b) (Fig.3). Another study revealed that gall-
bladder cancer stem cells (CSCs) and GBC tissues expressed
higher levels of DILC, leading to the enhancement of self-
generation, tumorigenicity, proliferation, and metastasis. In
addition, DILC facilitated the progression of GBC cells by
stimulating the Wnt/β-catenin signaling pathway. LncRNA
DILC enhances the Wnt/β-catenin signaling pathway by pro-
moting the stability and nuclear translocation of β-catenin.
This leads to increased transcriptional activity of β-catenin/
TCF complexes, driving the expression of genes that pro-
mote GBC cell proliferation, self-renewal, and metastasis
(Liang etal. 2019) (Fig.3).
Fig. 3 Regulatory mechanism involving lncRNA FOXD2-AS1,
DILC, and HGBC in GBC. lncRNA FOXD2-AS1 blocks MLH1 tran-
scription by binding the methyltransferase DNMT1 to its promoter.
This accelerates GBC. Gallbladder CSCs and GBC tissues produced
more DILC, enhancing self-generation, tumorigenicity, proliferation,
and metastasis. DILC also promoted GBC cell growth by activat-
ing the Wnt/β-catenin signaling pathway. LncRNA-HGBC sponging
miR-502-3p promotes GBC cell growth and metastasis. Blocking
miR-502-3p promotes SET expression, which activates AKT signal-
ing.
Naunyn-Schmiedeberg's Archives of Pharmacology
HOXA-AS2, a lncRNA located inside the HOXA cluster
between the HOXA3 and HOXA4 genes, has been identi-
fied as a promoter in many types of cancer, such as acute
promyelocytic leukemia and gastric cancer. Suppression
of HOXA-AS2 may hinder cell proliferation and trigger
apoptosis (Zhao etal. 2013). The HOXA-AS2 lncRNA
was shown to be highly upregulated in GBC tumor tissues.
Furthermore, the upregulation of HOXA-AS2 was shown
to be linked to the advancement of GBC. Suppression of
HOXA-AS2 hampers the ability of GBC cell lines to invade
and metastasize via modulating the EMT pathway. In cells
overexpressing lncRNA HOXA-AS2, the expression of
E-cadherin was reduced, while the expression of vimentin
and N-cadherin was increased. This suggests that the effects
of HOXA-AS2 on cell migration and invasion are partially
related to the process of EMT in GBC (Zhang etal. 2017b).
According to Hu etal., LncRNA-HGBC stabilized by
HuR promotes the growth of GBC by controlling the miR-
502-3p/SET/AKT axis. LncRNA-HGBC acts by sponging
miR-502-3p to induce GBC cell proliferation and metasta-
sis. Blocking miR-502-3p increases the expression of SET,
which in turn triggers the activation of AKT signaling. This
ultimately leads to the advancement of cancer. The latest
research establishes that the lncRNA-HGBC/miR-502-3p/
SET/AKT pathway has a significant impact on the course
of GBC. This highlights lncRNA-HGBC as a promising tar-
get for therapeutic interventions in GBC (Hu etal. 2019b).
Another study that examined the function of the DiGeorge
syndrome critical region gene (DGCR5) in GBC revealed
that by sponging miR-3619-5p and activating the JNK/p38
MAPK and MEK/ERK1/2 pathways, DGCR5 promotes
GBC cell invasion, proliferation, and migration (Liu etal.
2020c) (Fig.4 and Table1).
In GBC tissues, there was a rise in CCAT1 expression.
Suppression of CCAT1 reduced the target gene Bmi1’s
expression, whereas overexpression of CCAT1 increased
its expression by competitively binding to miRNA-218-5p.
Downregulation of CCAT1 hindered the growth and inva-
sive properties of GBC cells, perhaps by interfering with the
miRNA-218-5p-mediated control of Bmi1 (Ma etal. 2015b).
Researchers found that lncRNA GBCDRlnc1 interacts with
PGK1 and prevents its depletion, which causes an upregu-
lation of ATG5-ATG12 expression, therefore promoting
autophagy and drug resistance in GBC cells. Hence, target-
ing the GBCDRlnc1/PGK1/ATG5-ATG12 conjugate signal-
ing pathway should serve as a promising treatment strategy
for chemotherapy in GBC (Cai etal. 2019a).
HOTAIR has higher expression levels in GBC tissues
compared to safety margin non-tumoral tissues, suggest-
ing that HOTAIR is often elevated in GBC. Furthermore,
HOTAIR is more prominently present in GBC classified as
T3 or T4, as opposed to tumors classified as T1 or T2. The
expression of HOTAIR is more pronounced in tumors that
have progressed to regional lymph nodes (N1) versus origi-
nal tumors. This suggests that HOTAIR expression might
serve as a diagnostic marker for the course and prognosis
of GBC patients. HOTAIR enhances the growth and move-
ment of GBC cell lines, showing a positive association with
c-Myc expression and a negative association with miRNA-
130a. This indicates that this mechanism may contribute to
the advancement of cancer in GBC patients (Ma etal. 2014).
Downregulated lncRNAs inGBC
The lncRNA MEG3 suppresses the proliferation and induces
apoptosis in human GBC cells invitro. Moreover, it plays
a crucial role in regulating the progression of GBC. Tar-
geting the lncRNA MEG3 might act as a novel treatment
approach for GBC. The lncRNA MEG3 interacts with the
tumor-inhibiting gene p53 and controls the production of
p53 target genes by directly modifying the activity of the
p53 promoter (Bao etal. 2020) (Table1).
The expression of GATA6-AS was considerably reduced
in GBC tissues, and the overexpression of GATA6-AS led
to the suppression of cancer cell migration and invasion.
Hence, in addition to promoting cell proliferation, GATA6-
AS may also modulate several cellular activities, contribut-
ing to cancer biology. GATA6-AS is a probable regulator
that acts before miR-421 to limit its activity in GBC cells.
There are reports indicating that the levels of miR-421 may
be controlled by certain lncRNAsduring the progression of
cancer (Li etal. 2019b).
A clear decrease in the expression of lncRNA-LET was
identified in GBC in comparison to the surrounding normal
tissues. Conversely, those who have low levels of lncRNA-
LET have a notably worse prognosis compared to those with
high levels. Furthermore, the upregulation of lncRNA-LET
was confirmed to effectively hinder the invasion of GBC
cells invitro, both in hypoxic and in normoxic conditions.
The overexpression of lncRNA-LET was shown to provide
a growth advantage to tumor cells in low-oxygen environ-
ments. Overexpression of lncRNA-LET resulted in the
enhancement of cell cycle arrest specifically, in the G0/
G1 phase, and the initiation of programmed cell death in
hypoxic environments. Overexpression of LncRNA-LET
prevented GBC from growing invivo (Ma etal. 2015a).
Role oflncRNAs intheprogression andmetastasis
ofGBC
In GBC tissues and cell lines, PVT1 upregulation and miR-
30d-5p downregulation were detected. Suppression of PVT1
led to a reduction in cellular proliferation and invasion in
GBC cells. The targeting of miR-30d-5p by PVT1 was
confirmed, and there was a negative correlation observed
between their expression in GBC tissues. Moreover, PVT1
Naunyn-Schmiedeberg's Archives of Pharmacology
sponges miR-30d-5p, which raises downstream factor
HDAC9 expression and helps cancer cells function more
easily. The inhibition of miR-30d-5p might counteract the
consequences of PVT1 knockdown. PVT1 contributed to the
development of GBC via stimulating cell growth and inva-
sion through the action of miR-30d-5p. This suggests that
PVT1 may be a useful biomarker for the identification and
treatment of GBC (Liu and Xu 2020) (Fig.5 and Table2).
Knocking down Linc-ITGB1 resulted in a considerable
decline in the growth of GBC cells and showed a substantial
decrease in cell migration and invasion, which was prob-
ably due to a rise in β-catenin and a fall in TCF8, slug, and
vimentin. The acceleration of EMT by inc-ITGB1 could
contribute to the invasion and spread of GBC. In advanced-
stage patients, RNA interference that targets linc-ITGB1
may be a viable therapeutic approach (Wang etal. 2015)
(Fig.5 and Table2).
The lncRNA-HGBC, which exhibited upregulation in
GBC tissue, was predictive of an unfavorable prognosis
regarding survival. In cell lines of GBC, overexpression
Fig. 4 Regulatory mechanism involving lncRNA DGCR5 in GBC. DiGeorge syndrome critical region gene (DGCR5) induces GBC cell inva-
sion, proliferation, and migration by sponging miR-3619-5p and activating the JNK/p38 MAPK and MEK/ERK1/2 pathways.
Naunyn-Schmiedeberg's Archives of Pharmacology
of lncRNA-HGBC led to a corresponding elevation in cel-
lular replication and infiltration in laboratory settings and
implanted malignancies. LncRNA-HGBC exhibited specific
binding to HuR, which served to stabilize HGBC. Addition-
ally, HGBC binds to miR-502-3p to generate a competitive
endogenous RNA, which inhibits (Hu etal. 2019a).
Suppression of KIAA0125 greatly reduced cellu-
lar migration and invasion. Moreover, after suppressing
KIAA0125, GBC-SD/M cells exhibited reduced vimentin
and increased β-catenin levels. Thus, the results indicate that
KIAA0125 might function as a potential target for treatment
in advanced GBC, as it promotes the migration and invasion
of GBC cells (Lv etal. 2015) (Fig.5 and Table2).
The expression of the lncRNA-ROR was increased in
GBC tissues and a robust relationship existed between
elevated expression of LncRNA-ROR and a worse progno-
sis in individuals with GBC. Furthermore, cellular prolif-
eration, migration, and invasion were hampered by ROR
suppression. The EMT phenotype caused by TGF-1 was
reversed in SGC-996 and Noz cells after lncRNA-ROR was
knocked down. LncRNA-ROR performs a critical function
in the advancement of GBC by facilitating the EMT in this
kind of cancer. LncRNA-ROR might be used as a target for
therapeutic interventions and a prognostic indicator in GBC
(Wang etal. 2016d).
SPRY4-IT1 was found to be overexpressed in GBC
tissues. Knocking down SPRY4-IT1 greatly reduced
GBC cell growth. Moreover, the suppressive impact of
SPRY4-IT1 on cellular migration and invasion has been
linked to the EMT process (Yang etal. 2017). Tissue
Table 1 Characteristics of lncRNAs in the pathogenesis of GBC
LncRNA Alteration Function Ref.
FOXD2-AS1 Upregulated Suppression of MLH1 transcription. Gao etal. (2021b)
DILC Stimulating Wnt/β-catenin. Liang etal. (2019)
HOXA-AS2 Modulating EMT. Zhang etal. (2017b)
LncRNA-HGBC Regulating miR-502-3p/SET/AKT axis. Hu etal. (2019b)
DGCR5 stimulating MEK/ ERK1/2 and JNK/p38 MAPK. Liu etal. (2020c)
CCAT1 competitively binding to miRNA-218-5p. Ma etal. (2015b)
GBCDRlnc1 GBCDRlnc1/PGK1/ATG5-ATG12 axis. Cai etal. (2019a)
HOTAIR Positive association with c-Myc expression and a negative association with miRNA-
130a.
Ma etal. (2014)
MEG3 Downregulated Directly modifying the activity of the p53 promoter. Bao etal. (2020)
GATA6-AS GATA6-AS is a probable regulator that acts before miR-421 to limit its activity in
GBC.
Li etal. (2019b)
LET Cell cycle arrest specifically, in the G0/G1 phase, and the initiation of programmed
cell death in hypoxic environments.
Ma etal. (2015a)
Fig. 5 Role of lncRNAs in the
progression and metastasis of
GBC
Naunyn-Schmiedeberg's Archives of Pharmacology
samples from GBC demonstrated a substantial rise in
TUG1 expression. Studies demonstrated that suppress-
ing TUG1 drastically decreased the proliferation and
spread of GBC cells. TUG1 is stimulated by TGF-β1.
Additionally, when TUG1 was suppressed, it resulted in
the prevention of EMT in GBC cells. Furthermore, it has
been shown that TUG1 exerts a deleterious impact on the
expression of miR-300, a molecule known to function as
a suppressor in a range of malignancies (Ma etal. 2017).
Although the precise lncRNA’s principal functions in
GBC have yet to be determined, researchers have shown
that they are important regulators in the growth and devel-
opment of GBC (Wang etal. 2016a). MALAT1 promotes,
by activating the ERK/MAPK pathway, the proliferation
and metastasis of GBC cells (Wu etal. 2014a) (Fig.5 and
Table2). Furthermore, lncRNA HOTAIR also contributes
to GBC metastasis and progression through c-myc activation
of malignancy and downregulation of miRNA-130a in GBC
(Gupta etal. 2010). Due to lncRNA-LET downregulation
when GBC spreads and metastasizes, it is considered a prog-
nostic biomarker in GBC (Ma etal. 2015d). Additionally,
by competitively sponging miRNA-218-5p, lncRNA
CCAT1 promotes the expression of Bmi1, the target gene
of miRNA-218-5p, which in turn promotes the formation
of GBC (Ma etal. 2015a). Moreover, through the pathway
of Wnt/β-catenin, lncRNA DILC accelerates the progres-
sion and metastasis of GBC (Li etal. 2019a). Along with
the previously mentioned lncRNAs, DGCR5 is the lncRNA
linked to invasion, growth, invasion, migration, and prolif-
eration in GBC invivo. It does this by downregulating ZO-1
and E-cadherin expression and upregulating N-cadherin,
vimentin, MMP-2, and MMP-9 expression levels via the
MEK/ERK1/2 and JNK/p38 MAPK axes (Liu etal. 2020b).
An additional lncRNA linked to the emergence of aggres-
sive characteristics in cancer is FOXD2-AS1, which has
been shown to induce proliferation and metastasis in GBC
because it has the ability to captivate DNMT1, a methyl-
transferase that subsequently methylates the MLH1 gene’s
promoter, thereby inhibiting MLH1 transcription (Gao etal.
2021a). H19 expression in GBC has been shown to be higher
in malignant tissue than in adjacent non-tumor tissue. By
sponging the miRNAs in the axes of H19/miR-342-3p/
Table 2 Role of lncRNAs in the progression and metastasis of GBC
LncRNA Expression Function Mechanism Ref.
PVT1 Up Stimulate cell growth and invasion miR-30d-5p downregulation Liu and Xu (2020)
Linc-ITGB1 Up Acceleration of EMT Decreased β-catenin and increased TCF8,
slug, and vimentin levels
Wang etal. (2015)
HGBC Up Elevation in cellular replication and infiltra-
tion
HGBC binds to miR-502-3p to generate a
competitive endogenous RNA
Hu etal. (2019a)
KIAA0125 Up Promotes the invasion and migration Reduced β-catenin and increased vimentin
levels
Lv etal. (2015)
Lnc-ROR Up Promotes cellular proliferation, migration,
invasion, and EMT
Not detected Wang etal. (2016d)
SPRY4-IT1 Up Promotes migration, invasion, and EMT
process
Not detected Yang etal. (2017)
TUG1 Up Promotes migration, invasion, and EMT
process
Exerts a deleterious impact on the expression
of miR-300
Ma etal. (2017)
MALAT1 Up Promotes the proliferation and metastasis Stimulating ERK/MAPK Wang etal. (2016a)
HOTAIR Up Contributes to metastasis and progression c-myc activation of malignancy and down-
regulation of miRNA-130a
Gupta etal. (2010)
LET Down It facilitates GBC to spread and metastasize Not determined Ma etal. (2015d)
CCAT1 Up Encourages the development and metastasis
of GBC
Escalates the expression of the Bmi1 gene by
miRNA-218-5p sponging
Ma etal. (2015a)
DILC Up Accelerates the progression and metastasis Wnt/β-catenin pathway activation Li etal. (2019a)
DGCR5 Up Promotes proliferation, migration, invasion,
and growth in GBC
Through enhancing the JNK/p38 MAPK and
MEK/ERK1/2 routes
Liu etal. (2020b)
FOXD2-AS1 Up Inhibiting MLH1 gene transcription Gao etal. (2021a)
H19 Up Associated with larger tumors, lymphatic
metastases, and a worse prognosis
Sponging the miRNAs in the axes of H19/
miR-342-3p/FOXM1 and H19/miR-194-5p/
AKT2
Wang etal. (2016b),
Wang etal.
(2016c)
GATA6-AS Down Induce proliferation and metastasis Sponging miR-421 Li etal. (2019a)
MEG3 Down Exacerbates the prognosis and metastasis By controlling the NF-κb pathway, ubiquit-
inating EZH2, and suppressing large tumor
suppressor 2 (LATS2)
Jin etal. (2018)
Naunyn-Schmiedeberg's Archives of Pharmacology
FOXM1 and H19/miR-194-5p/AKT2, its expression is
linked to bigger tumors, lymphatic metastases, and a worse
prognosis in patients with GBC (Wang etal. 2016b, Wang
etal. 2016c). LncRNAs that are downregulated during the
progression and metastasis of GBC are GATA6-AS and
MEG3. GATA6-AS has been shown to decrease progres-
sively in GBC advanced stages by downregulating miR-421
(Li etal. 2019a). On the other hand, poor MEG3 expression
exacerbates the prognosis and metastasis by controlling the
NF-κB pathway, ubiquitinating EZH2, and suppressing large
tumor suppressor 2 (LATS2) (Jin etal. 2018).
Role oflncRNAs indrug resistance
Since GBC is the most severe kind of cancer that may
affect the biliary tract, individuals with unresectable GBC
need intensive treatment. Nonetheless, physicians continue
to face difficulties with chemotherapy resistance (Lai etal.
2023). LncRNAs are involved not only in the etiology and
prognosis of GBC but also in the positive or negative reg-
ulation of resistance to various therapies (Eptaminitaki
etal. 2022). Studies revealed a considerable upregulation
of lncRNA myosin light-chain kinase antisense RNA 1
(MYLK-AS1) in GBC, which was found to be connected
with a worse prognosis and clinical features. Furthermore,
induced MYLK-AS1 expression increased GBC cell prolif-
eration and invitro gemcitabine resistance. In terms of its
underlying mechanisms, MYLK-AS1 released the inhibi-
tion of enhancer of zeste 2 polycomb repressive complex
2 (EZH2) subunit expression by acting as an effective
miR-217 sponge. The upregulation of EZH2 expression
by MYLK-AS1 was found to boost cell proliferation and
resistance to gemcitabine in GBC cells. Furthermore,
EZH2 was found to directly target miR-217 (Li etal. 2021).
It was found that lncRNA ENST00000425894, known as
GBC drug resistance-associated lncRNA1 (GBCDRlnc1),
expression is elevated in GBC tissues, and it functions as a
pivotal regulator in GBC chemoresistance. The sensitivity
of established GBC cells resistant to doxorubicin (Dox)
was improved by knocking down of GBCDRlnc1, which
was achieved by blocking autophagy at an early stage in
the cell culture. This effect was observed both invitro and
invivo. According to these findings, GBCDRlnc1 may be
a potential target for therapeutic intervention in advanced
GBC (Cai etal. 2019a) (Fig.6).
It was found that the lncRNA RP11-147L13.8 was recur-
rently downregulated in GBC tumor tissues. Furthermore,
there was a correlation between a poorer prognosis and a
lower expression level of lncRNA RP11-147L13.8 in GBC
patients. In addition, research conducted both invitro and
invivo has clarified that overexpression of the lncRNA
RP11-147L13.8 prevents GBC cells from migrating and
from becoming invasive, while at the same time it increases
the cells’ vulnerability to gemcitabine. As a result, RP11-
147L13.8 can be considered a promising combination of
treatments for the management of GBC that involves the
use of gemcitabine, as it effectively suppresses the metas-
tasis of GBC chemo-resistance and metastasis through its
Fig. 6 Role of lncRNAs in drug resistance. In GBC, lncRNA MYLK-
AS1 was upregulated and associated with a worse prognosis and
clinical characteristics. Invitro gemcitabine resistance and GBC cell
proliferation enhanced with MYLK-AS1 expression. By suppressing
autophagy early in cell culture, GBCDRlnc1 knockdown increased
the sensitivity of established GBC cells resistant to doxorubicin. With
these findings, GBCDRlnc1 may be a therapeutic target for advanced
GBC. Invitro and invivo studies have shown that overexpression of
the lncRNA RP11-147L13.8 stops GBC cells from migrating and
becoming invasive while increasing their gemcitabine susceptibility.
By blocking the proteasome, SSTR5-AS1 stabilizes the RNA-bind-
ing protein NONO. SSTR5-AS1-mediated gemcitabine resistance
requires NONO.
Naunyn-Schmiedeberg's Archives of Pharmacology
modulation of the c-Jun protein phosphorylation (Zheng
etal. 2021).
It was found that GBC samples and cell lines exhibit a
substantial rise in lncRNA SSTR5-AS1, especially gemcit-
abine-resistant cell lines, showing a significant increase in
lncRNA SSTR5-AS1. A worse survival rate was associated
with increased SSTR5-AS1 expression in GBC patients. It
has been discovered that via inhibiting apoptosis, the over-
expression of SSTR5-AS1 increases resistance to gemcit-
abine. Invitro, drug-resistant GBC cells were more sensitive
to gemcitabine when SSTR5-AS1 was knocked down, and
invivo, the development of drug-resistant GBC xenografts
was significantly reduced. These results are noteworthy
because they showed that SSTR5-AS1 binds to the RNA-
binding protein NONO and stabilizes it by preventing the
proteasome from breaking down NONO. Moreover, NONO
is necessary for gemcitabine resistance mediated by SSTR5-
AS1. The information provided in this work sheds light on
the function of lncRNAs in the emergence of chemothera-
peutic resistance. Additionally, these discoveries help in
developing efficient chemotherapy methods specifically tai-
lored for patients with unresectable GBC (Xue etal. 2020).
Clinical importance oflncRNAs inGBC
Diagnosis
Early GBC is currently identified incidentally in patients
with cholelithiasis since there are no specific signs or symp-
toms linked to this disease. A palpable mass, ascites, biliary
or gastrointestinal obstruction, and weight loss are typical
signs of advanced-stage GBC (Vijayakumar etal. 2013). In
these advanced stages, most GBCs are associated with poor
prognosis, short life expectancy, and metastasis, as well as
curative treatment is not possible. The 5-year survival rate
in advanced stages (T3 and T4 stages) is less than 5%; how-
ever, if the disease was discovered earlier in its course, the
rate rises to 75%. Thus, aggressive propagation and delayed
manifestations make it one of the most catastrophic tumors
with an unfavorable outcome. The molecular markers rou-
tinely used for diagnosis of GBC lack specificity where they
are increased in some non-carcinogenic diseases. Thus,
researchers are seeking more specific and reliable diagnos-
tic methods for early detection and intervention to mitsigate
tumor progression (Vijayakumar etal. 2013, Dilek etal.
2019, Lopes Vendrami etal. 2021, Yang etal. 2022).
Diverse studies have addressed that lncRNAs could act
as innovative and exciting molecular markers for the diag-
nosis of diverse diseases (Abd-Elmawla etal. 2020, Mehana
etal. 2022, Abd-Elmawla etal. 2023, Kortam etal. 2023).
Blood plasma and urine are examples of bodily fluids where
circulatory lncRNAs have been found to function as bio-
markers for the early detection of GBC. Tumor-derived
lncRNAs usually develop a very stable secondary structure
in the peripheral circulation which is resistant to ribonu-
clease activity, rendering them appropriate molecules for
quantitative detection (Zhong etal. 2019, Niu etal. 2020).
Circulating lncRNAs function as newly developed tumor-
specific molecular markers that are employed in early cancer
diagnosis (Zhou etal. 2016, Lu etal. 2017, Xie etal. 2021b).
There are a number of pros associated with using circulating
lncRNAs as diagnostic biomarkers, including the following:
(i) compared to traditional protein-based markers, they have
a better specificity and sensitivity; (ii) they have the abil-
ity to dynamically monitor the state of the illness, includ-
ing forecasting patient survival times and the likelihood of
tumor spread and relapse; (iii) in clinical practice, lncRNAs
get clinical acceptance as noninvasive tools, high repeat-
ability, reduced patient suffering from the biopsy, and can
save patients’ costs. Statistically, the relationship between
the diagnostic test’s sensitivity and specificity is reflected in
the receiver operating characteristic curve. The diagnostic
accuracy can be measured using the computed area under
the curve (AUC). Greater AUC values signify an enhanced
diagnostic precision of the test (Mandrekar 2010, Zhou etal.
2016, Xie etal. 2021b).
In this context, Liu etal. suggested that the lncRNA
SNHG6 could be a promising marker for the diagnosis
of GBC. The gene expression of SNHG6 was elevated in
GBC and correlated with an AUC greater than 0.8 (Liu
etal. 2020d) (Table3). Furthermore, the diagnostic value
of SNHG6 has been addressed in other types of cancers such
as esophageal squamous cell carcinoma and hepatocellular
carcinoma (Birgani etal. 2018, Zhang etal. 2019). Relat-
edly, Linc00261, which was downregulated in GBC tissues
when compared to normal ones, may serve as a diagnostic
biomarker in GBC. According to Niu and his colleagues,
Linc00261 possesses high diagnostic efficiency with high
specificity and sensitivity as well as an AUC=0.8 (Niu etal.
2020). Another lncRNA is DGCR5 which was reported to
participate in GBC cell invasion, migration, and prolif-
eration. The preceding study provides the first report that
DGCR5 could be a new biomarker for the early diagnosis
of GBC (Liu etal. 2020b) (Table3). Added to that, DGCR5
was nominated as a promising biomarker in ovarian cancer
as reported by Chen etal. (2019a).
The lncRNA in GBC linked to liver metastasis (lnc-
GALM) was substantially expressed in GBC patients and
associated with low survival rates, suggesting its utility as
a diagnostic marker as reported by Li etal. (2020a). More-
over, the noticed highly expressed levels of Linc01694
in GBC suggested that it could be a potential diagnostic
marker for GBC (Liu etal. 2020a). In tandem, ANRIL and
MEG3 were closely linked with GBC development and
recommended as noninvasive biomarkers. Similarly, com-
pared with paired normal ones collected from 84 patients,
Naunyn-Schmiedeberg's Archives of Pharmacology
Table 3 Theoretical basis underlying the utility of lncRNAs as diagnostic tools in GBC
LncRNAs Expression Diagnostic/prognostic Mechanism in GBC Effect Ref
SNHG6 Up Diagnostic Targets miR-26b-5p which influences the hedge-
hog signaling pathway
Promotes EMT, proliferation, and invasion of
GBC cells
Liu etal. (2020d)
Linc00261 Down Diagnostic Associated with GBC development and progres-
sion
Niu etal. (2020)
DGCR5 Up Diagnostic Sponges miR-3619-5p and activating MEK/
ERK1/2 and JNK/p38 MAPK pathways
Promotes proliferation, migration, invasion, and
induced apoptosis and cell cycle arrest
Liu etal. (2020b)
LncGALM Up Diagnostic/prognostic Sponges miR-200 family members, which
increase ZEB1, ZEB2, and N-cadherin.
Stimulate invasion-metastasis cascade by elevat-
ing EMT.
Li etal. (2020a), Li etal. (2020b)
Linc01694 Up Diagnostic Modulates miR-340-5p/Sox4 axis Progresses the proliferation and invasion in GBC
cells and attenuates the apoptosis
Liu etal. (2020a)
ANRIL
MEG3
Up
Low
Diagnostic/prognostic Reduces ki-67 and elevates caspase 3 levels ANRIL promotes the proliferation of gallbladder
cells and attenuates apoptosis.
MEG3 mitigates cell proliferation and promotes
apoptosis.
Xie etal. (2021a)
CCAT1 Up Diagnostic/prognostic Sponges miRNA-218-5p which stimulates Bmi1
gene expression
Promotes proliferation and invasiveness. Ma etal. (2015a), Ma etal. (2015b)
FOXD2-AS1 Up Diagnostic Stimulates MLH1 methylation by recruiting
DNMT1 to the MLH1 promoter, thus mitigat-
ing MLH1 transcription.
Stimulates cell proliferation, invasion, and
migration, whereas reduces cell apoptosis
Gao etal. (2021a)
GBCDRlnc1 Up Diagnostic Interacts with phosphoglycerate kinase 1 and
downregulates autophagy initiator ATG5-
ATG12 conjugate.
Enhances autophagy at the initial stage,
decreases the sensitivity of Dox-resistant in
GBC
Cai etal. (2019b)
HGBC Up Diagnostic/prognostic Sponges miR-502-3p and inhibit SET gene
expression
Acts via a positive regulatory loop consisting of
HEGBC, IL-11, and STAT3.
Stimulates cell proliferation and invasion invitro
and in xenografted tumors.
Hu etal. (2019a), Yang etal. (2018)
DILC Up Diagnostic Influences Wnt/β-catenin axis. Promotes the self-renewal, tumorigenicity, pro-
liferation, and metastasis
Liang etal. (2019)
LncROR Up Diagnostic Promotes EMT phenotype driven by TGF-β1 Activates the tumor cells proliferation, migra-
tion, and invasion
Wang etal. (2016d)
LncEPIC1 Up Diagnostic Represses lncRNA LET expression. Stimulates proliferation and inhibits apoptosis Liu etal. (2016b)
FIRRE Up Diagnostic Sponge miR-520a-3p via controlling YOD1 gene
expression
Stimulates proliferation and migration, and
inhibits apoptosis
Wang etal. (2021)
SPRY4-IT1 Up Diagnostic Enhances GBC cell proliferation Yang etal. (2017)
PVT1 Up Diagnostic PVT1 recruited DNMT1 via EZH2 to the miR-
18b-5p DNA promoter and suppressed the
transcription of miR-18b-5p through DNA
methylation.
Enhances GBC cell proliferation invitro and
invivo.
Jin etal. (2020b)
LncLET Down Diagnostic Promotes cell cycle arrest at G0/G1 phase and
the induction of apoptosis under hypoxic
conditions
Mitigates the invasion of GBC under hypoxic or
normoxic conditions invitro.
Ma etal. (2015d)
Naunyn-Schmiedeberg's Archives of Pharmacology
the level of ANRIL was highly upregulated and the level
of MEG3 was downregulated in GBC tissues (Liu etal.
2016a). The lncRNA CCAT1 was elevated in the tissues
of 40 patients with GBC in comparison to adjacent normal
tissues (Ma etal. 2015a). Gao etal. (2021a) reported that
the lncRNA FOXD2-AS1 was highly upregulated in GBC
tissues in comparison with the adjacent normal tissues.
Furthermore, a newly identified lncRNA is GBCDRlnc1
which was highly expressed in GBC tissues (Cai etal.
2019b). All of these aberrated lncRNAs may help to diag-
nose and treat GBC early.
The lncRNA which is referred to as highly expressed in
GBC (HGBC) was markedly elevated in GBC tissues than
adjacent benign tissues as reported by Hu and his colleagues
(2019a). Relevantly, the lncRNA DILC was highly expressed
in both GBC stem cells and patients’s tissues reflecting its
plausible role as a diagnostic molecule (Liang etal. 2019).
The lncRNA ROR which has been reported in several types
of malignancies, was also elevated in GBC and stimulated
the EMT in GBC (Wang etal. 2016d). FU etal. submitted
that the lncRNA EPIC1 which is named as epigenetically
induced lncRNA1 was highly differentiated in GBC giving a
theoretical basis for their utility in diagnosis (Fu etal. 2021).
In the same milieu, Wang etal. reports that the lncRNA
FIRRE which is referred to as a functional intergenic repeat-
ing RNA element could participate in the diagnostic strategy
of GBC. This may be attributed to its high expression pat-
tern in GBC and its close association with stimulated pro-
liferation and cancer invasion (Wang etal. 2021) (Table3).
Moreover, the lncRNAs SPRY4-IT1 and PVT1 exerted their
oncogenic role in GBC development suggesting its promis-
ing clinical marker tool (Yang etal. 2017, Jin etal. 2020b).
Another lncRNA is HOXA cluster antisense RNA2 (HOXA-
AS2) which was overexpressed in GBC tissues (Zhang etal.
2017b). On the opposite, Ma etal. (2015d) found that the
tumor suppressor lncRNA LET could be valuable in the
diagnosis of GBC where it was downregulated in GBC
in comparison to paracancerous normal tissues. Wu etal.
(2017) introduced the lncRNA PAGBC, which is referred to
as prognosisassociated GBC lncRNA as a diagnostic tool
in GBC based on its role in stimulating tumor growth and
metastasis.
Indeed, several differentially expressed lncRNAs were
identified in GBC that contribute to GBC development.
However, their clinical utility as diagnostic markers need
further investigations to fully interpret the molecular mecha-
nisms of GBC. Additionally, the mechanisms behind their
secretion and transport to the circulation should be reported
precisely to put forward their utility as plasma or serum
noninvasive diagnostic biomarkers. Quantitative real-time
PCR is regarded as the gold standard for lncRNA determi-
nation approaches. However, there are no unified standard
extraction techniques, endogenous controls, or quantification
Table 3 (continued)
LncRNAs Expression Diagnostic/prognostic Mechanism in GBC Effect Ref
PAGBC Up Diagnostic Sponges miR133b and miR511.
Stimulates AKT/mTOR
Promotes tumor growth and metastasis. Wu etal. (2017)
AFAP1-AS1 Up Prognostic Proliferation, invasion, epithelial-mesenchymal
transition
Yang etal. (2017)
HOTAIR Up Prognostic Enhances the expression of c-Myc via sponging
miR-130a
Migration, proliferation Ma etal. (2014)
H19 Up Prognostic Acts via the H19/miR-342-3p/FOXM1 and H19/
miR-194-5p/AKT2 axis.
Proliferation, tumorigenicity, migration, inva-
sion, epithelial-mesenchymal transition
Wang etal., (2016f), Wang etal. (2022)
MALAT1 Up Prognostic MiR-363-3p’s competing endogenous RNA,
MALAT1, controls the expression of MCL-1.
Proliferation tumorigenicity, migration, invasion,
metastasis
Wu etal. (2014a), Wang etal. (2016g)
MINCR Up Prognostic Alters miR-26a-5p’s capacity to bind to EZH2 Proliferation, migration, tumorigenicity, inva-
sion, epithelial-mesenchymal transition
Wang etal. (2016g)
OIP5-AS1 Up Prognostic Reduces the expression of miR-143-3p Proliferation, migration, invasion Li etal. (2020b)
SSTR5-AS1 Up Prognostic NONO/SSTR5_AS1 interaction stops NONO
from degrading
Drug resistance Xue etal. (2020)
MEG3 Up Prognostic Inhibit poor prognosis, inhibit proliferation,
inhibit migration, inhibit metastasis
Liu etal. (2016b), Jin etal. (2018), Bao
etal. (2020)
Naunyn-Schmiedeberg's Archives of Pharmacology
techniques (Yang etal. 2014, Bhan etal. 2017, Jiang etal.
2019).
Thereby, to introduce all of these circulating lncRNAs
into the clinical realm, additional points should be taken
into consideration such as the type and volume of sample
needed, the used anticoagulant, and the stored temperature.
The extraction techniques and endogenous regulation of
lncRNAs in bodily fluids ought to be uniform. Providing
standard techniques for evaluating the quality of lncRNA
and the accuracy of qPCR data should be more precise and
reliable in early diagnosis of GBC. Finally, a larger pool of
prospective patients is warranted to assess the performance
of these diagnostic tools in terms of sensitivity, specificity,
and diagnostic efficiency (Shi etal. 2016).
Prognosis
LncRNAs are increasingly being recognized as possible
biomarkers for disease development, metastasis, and patient
prognosis. In this discussion, we explore the function of
many lncRNAs as indicators for diagnosis and prognosis in
GBC. AFAP1-AS1, a lncRNA, is highly expressed in GBC
tissues and is linked to larger tumor growth and an unfavora-
ble prognosis in cancer patients (Li etal. 2019c). AFAP1-
AS1 knockdown may inhibit proliferation and invasion. Zhu
etal. found that the ceRNA AFAP1-AS1, which is overex-
pressed in GBC tissues and cell lines, controls the hsa-miR-
15a-5p/bcl-2 axis and promotes the growth and metastasis of
GBC cells. Zhu etal. found that AFAP1-AS1 overexpressed
in GBC tissues and cell lines is a ceRNA of hsa-miR-15a-5p,
which regulates the hsa-miR-15a-5p/bcl-2 axis and drives
GBC cell proliferation and metastasis. This may help treat
GBC clinically (Zhu etal. 2022). Also, ANRIL is upregu-
lated in GBC tissues and increases proliferation and tumor
size in a murine model, indicating its involvement in cancer
progression (Li etal. 2019c) (Table3).
CCAT1 is another lncRNA found in various cancers, such
as colorectal and gastric cancer (Liu etal. 2019). Gastric
and colorectal cancer tissues have increased expression of
it, particularly in advanced stages and lymph node metasta-
ses. The expression of CCAT1 is correlated with advanced
tumor node metastatic stage, invasion of lymph nodes, and
tumor status, indicating a poor prognosis for colorectal
and stomach cancer. Knocking down CCAT1 invitro and
invivo reduces the occurrence of S-phase, invasion, and the
growth of tumors. An RNA molecule of significant length,
GALM expression is elevated in tumor tissues from glio-
blastoma (GBM) and is linked to lymph node metastases,
poorly differentiated cells, and an advanced TNM stage. The
expression of GALM is associated with decreased survival
in GBC, suggesting a worse prognosis. The GALM gene
was artificially increased in GBC cell lines to investigate its
role, revealing enhanced migratory and invasive capabilities.
The invivo experiments showed that GALM enhanced the
liver metastatic capacity, indicating its potential to enhance
EMT and metastasis in GBC. GALM interacts with miR-200
family members and IL-1β mRNA, functioning as molecular
decoys (Hu etal. 2019b).
HEGBC, a lncRNA, rises substantially in GBC cell
lines and tissues and is highly associated with lymph node
metastases and TNM stages. High HEGBC expression in
GBC patients is related to poor survival. Ectopic HEGBC
expression increases proliferation and migration, reducing
apoptosis. HEGBC overexpression in GBC cell lines in
nude mice enhances tumor growth, proliferation markers,
and liver metastasis. By binding to the promoter, HEGBC
promotes IL-11 transcription and activates STAT3 signal-
ing pathway cells. GBC tissues overexpress malignancy-
promoting lncRNA HOTAIR. Ma etal. analyzed HOTAIR
expression in 65 GBC tissue samples to determine its role.
In tumor tissues, HOTAIR expression was much greater than
in matching normal tissues. It may be a GBC progression
and prognosis marker since gallbladder tumors express it
more and disseminate it to regional lymph nodes. HOTAIR
increases GBC cell line proliferation and migration, corre-
lates positively with c-Myc expression, and negatively with
miRNA-130a, suggesting a role in GBC progression (Ma
etal. 2014).
It has been demonstrated that LncRNA H19, which is
expressed in a variety of malignancies, contributes to the
acquisition of different oncogenic characteristics (Ghafouri-
Fard etal. 2020). Patients with GBC had worse survival
rates and larger tumors due to the higher expression of H19
in malignant cells compared to non-cancerous gallbladder
tissue. The expression of the AKT2 gene and protein is
increased by H19, whereas the expression of miR-194-5p is
decreased. The modulation of the H19/miR-194-5p/AKT2
axis may contribute to the oncogenic properties of H19 in
GBC by influencing cell proliferation and EMT (Wang etal.
2016e, Wang etal. 2016f, Wang etal. 2022). Thus, treating
GBC in humans, H19 may therefore be regarded as a pos-
sible prognostic biomarker and/or therapeutic target.
Metastasis-associated lung adenocarcinoma transcript
1 (MALAT1) is a lncRNA that promotes the growth of
cancerous tumors and influences the process of alternative
splicing of pre-messenger RNA (Zhang etal. 1993). Interac-
tion with unmethylated CBX4 controls gene translocation
between the polycomb complex and interchromatin bodies.
Silencing MALAT1 reduces GBC cell growth but enhances
the G2/M cell cycle transition. MALAT1 inhibition reduces
tumor and xenograft growth invivo. MALAT1 knockdown
stops GBC cell invasion. MALAT1 is a downstream tar-
get of MMP9. MALAT1 deletion activates ERK/MAPK by
decreasing MMP9. These findings support MALAT1 as a
GBC therapeutic and prognostic marker (Wu etal. 2014a,
Wang etal. 2016g).
Naunyn-Schmiedeberg's Archives of Pharmacology
The expression of lncRNA MINCR is elevated in glio-
blastoma (GBM) tissues and is correlated with larger tumor
size, lymph node metastases, and a shorter overall survival
time. These findings indicate that MINCR is linked to a poor
prognosis in GBM patients. Knocking down MINCR leads
to a decrease in cell proliferation, migration, invasion, and
EMT under laboratory conditions. Tumor volume decreased
in mice that were injected with GBC si-MINCR cells.
MINCR might potentially influence the binding of miR-
26a-5p to EZH2 by interacting with miRNA ribonucleo-
protein complexes. NEAT1, a different lncRNA, is highly
expressed in GBC tissues and might potentially be down-
regulated to decrease the formation of colonies, migration,
invasion, and tumor growth (Wang etal. 2016g). LncRNA
OIP5-AS1 is a new lncRNA that plays a role in cancer devel-
opment (Wang etal. 2020b). It has been overexpressed in
GBC cell lines and is linked to proliferation, migration, and
invasion (Li etal. 2020b). PVT1 is an additional lncRNA
that its expression is increased in GBC tissues. It is linked
to a more advanced TNM stage, a distant spread of cancer,
and a poorer overall survival rate. PVT1 serves as a powerful
and autonomous prognostic marker for patients with GBC,
indicating an unfavorable prognosis. Suppression of PVT1
hampers cellular growth, the formation of colonies, move-
ment, and infiltration, indicating its potential to control EMT
and the advancement of cancer. The observed effects are
likely caused by the increased expression of HK2, which is
mediated by PVT1 through its ability to competitively bind
to miR-143, a kind of endogenous RNA (Chen etal. 2019b,
Jin etal. 2020a, Liu and Xu 2020).
Gemcitabine-resistant GBC cell lines express SSTR5-
AS1, a lncRNA involved in chemotherapy. Increased
SSTR5-AS1 expression reduces programmed cell death,
indicating drug resistance. It is also expressed at higher
levels in GBC tissues and cell lines and is associated with
worse survival. SSTR5-AS1 may predict GBC prognosis
(Yang etal. 2017). The expression of lncRNA-LET inde-
pendently predicts the metastasis and mortality of GBC
patients. Hypoxia facilitates the proliferation of tumors,
enhances their malignant properties, and contributes to their
resistance to treatment. Hypoxia decreases the expression of
lncRNA-LET in GBC EZ-GB2 and SGC-996 cells. When
GBC cells are exposed to low oxygen levels (hypoxia), the
overexpression of lncRNA-LET leads to an increase in the
levels of p21, Caspase-3, and the Bax/Bcl-2 ratio. These
findings indicate that lncRNA-LET plays a role in promot-
ing apoptosis (Ma etal. 2015a, Shen etal. 2015, Yan etal.
2017). The lncRNA-MEG3 is expressed at lower levels in
glioblastoma (GBM) tissues than in normal tissues, which is
associated with a worse prognosis and the spread of cancer
cells to the lymph nodes. MEG3 overexpression suppresses
cell growth, inhibits colony formation, and triggers apoptosis
in GBM cell lines. In an invivo setting, the overexpression
of a certain gene leads to a reduction in the formation of
tumors and a decrease in the presence of the Ki-67 marker in
the BALB/c nude mouse model. MEG3 suppresses the inva-
sion of NOZ cells and facilitates the degradation of EZH2 by
ubiquitination. This process regulates the LATS2 and NF-κb
pathways (Liu etal. 2016b, Jin etal. 2018, Bao etal. 2020).
Comparably, GCASPC is a noncoding RNA that is down-
expressed in GBC tissues and has been linked to decreased
overall survival (OS), disease-free survival (DFS) rates,
advanced disease stage, and tumor size (Ludwig etal. 2016).
Future prospective andconclusions
LncRNAs play a pivotal role in the control of gene expres-
sion at different levels, such as chromatin modification, tran-
scription, and post-transcriptional processing. Furthermore,
they have been identified to possess oncogenic and tumor-
suppressive functions. One of the most active areas in sci-
entific research is the use of lncRNAs for clinical practice.
GBC is an uncommon disease that has a poor prognosis.
It is characterized by a particular geographic distribution
and a distinct set of risk factors. Regretfully, there are few
treatment options available for GBC. Radiation, surgery,
and cytotoxic chemotherapy are among the limited sur-
vival benefits of GBC treatments. It is essential to under-
stand the molecular pathophysiology of GBC to advance
designed treatment strategies. Early diagnosis and prediction
of potential metastasis are key points for improving GBC
patient outcomes. For this purpose, diagnostic and prognos-
tic biomarkers are needed for the proper selection of the
best treatment option for preoperative and/or postoperative
GBC cases. The most important advantage of lncRNAs is
their presence in body fluids, making it possible to use them
as non-invasive biomarkers in clinics. So, more studies on
circulating lncRNAs should be performed for the identifica-
tion of lncRNAs that may be used practically as non-invasive
biomarkers for GBC. The low expression levels of several
lncRNAs present a challenge in this regard, necessitating the
use of procedures based on amplification and/or enrichment
of the circulating molecules. It is important to note that there
were no reliable internal controls for lncRNA in serum or
blood. In order to precisely estimate the circulating lncRNA
levels in the future, stable and reliable internal controls are
required. Overall, elucidating the mechanism of lncRNAs
aberrant expression and the downstream mechanism of lncR-
NAs in malignancies will help us better understand lncRNA
expression and will enable us to develop new tumor markers
for clinical diagnosis and prognosis evaluation of tumors.
In conclusion, lncRNAs have critical roles in GBC initia-
tion, progression, and metastasis through controlling various
cellular processes such as proliferation, apoptosis, angiogen-
esis, and EMT. In particular, SNHG6, Linc00261, GALM,
Naunyn-Schmiedeberg's Archives of Pharmacology
OIP5-AS1, MINCR, DGCR5, MEG3, TUG1, and DILC
are key players in regulating the aforementioned processes.
Dysregulation of lncRNAs in GBC can be observed in both
bodily fluids and tissues, offering the potential for identify-
ing innovative non-invasive biomarkers for diagnosing, pre-
dicting outcomes, and treating GBC. These markers could
eventually find clinical applications in the future.
Acknowledgements The authors are thankful to the Deanship of Grad-
uate Studies and Scientific Research at University of Bisha for sup-
porting this work through the Fast-Track Research Support Program.
The authors would like to thank the Deanship of Scientific Research at
Shaqra University for supporting this work.
Authors contributions Conception and design: H.E., O.A.M., R.M.,
M.B.Z., and A.S.D. Collection and/or assembly of the data: A.F.R.,
Mu.A.A., and N.E. Manuscript writing: N.A.A.A., A.H., Ma.A.A., and
A.M.E. All the authors have read and approved the published version
of the manuscript.The authors confirm that no paper mill and artificial
intelligence was used.
Data availability Not applicable.
Declarations
Ethical approval Not applicable.
Consent to participate Not applicable.
Consent for publication Not applicable.
Competing interests The authors declare no competing interests.
References
Abd-Elmawla MA, Hassan M, Elsabagh YA, Alnaggar ARL, Seno-
usy MA (2020) Deregulation of long noncoding RNAs ANCR,
TINCR, HOTTIP and SPRY4-IT1 in plasma of systemic scle-
rosis patients: SPRY4-IT1 as a novel biomarker of scleroderma
and its subtypes. Cytokine 133:155124
Abd-Elmawla MA, Elsabagh YA, Aborehab NM (2023) Association
of XIST/miRNA155/Gab2/TAK1 cascade with the pathogenesis
of anti-phospholipid syndrome and its effect on cell adhesion
molecules and inflammatory mediators. Sci Rep 13:18790
Adamska A, Domenichini A, Falasca M (2017) Pancreatic ductal
adenocarcinoma: current and evolving therapies. Int J Mol Sci
18:1338–1338
Alvarez-Dominguez JR, Lodish HF (2017) Emerging mechanisms
of long noncoding RNA function during normal and malignant
hematopoiesis. Blood 130:1965–1975
Arslan C, Yalcin S (2014) Current and future systemic treatment
options in metastatic pancreatic cancer. J Gastrointest Oncol
5:280–280
Bao D, Yuan RX, Zhang Y (2020) Effects of lncRNA MEG3 on pro-
liferation and apoptosis of gallbladder cancer cells through regu-
lating NF-κB signaling pathway. Eur Rev Med Pharmacol Sci
24:6632–6638
Bhan A, Soleimani M, Mandal SS (2017) Long noncoding RNA and
cancer: a new paradigm. Cancer Res 77:3965–3981
Birgani MT, Hajjari M, Shahrisa A, Khoshnevisan A, Shoja Z, Mota-
hari P, Farhangi B (2018) Long non-coding RNA SNHG6 as a
potential biomarker for hepatocellular carcinoma. Pathol Oncol
Res 24:329–337
Cai Q, Wang S, Jin L, Weng M, Zhou D, Wang J, Tang Z, Quan Z
(2019a) Long non-coding RNA GBCDRlnc1 induces chemore-
sistance of gallbladder cancer cells by activating autophagy. Mol
Cancer 18:82
Cai Q, Wang S, Jin L, Weng M, Zhou D, Wang J, Tang Z, Quan Z
(2019b) Long non-coding RNA GBCDRlnc1 induces chemore-
sistance of gallbladder cancer cells by activating autophagy. Mol
Cancer 18:1–16
Calderón-Reyes C, Pezoa RS, Leal P, Ribera-Fonseca A, Cáceres C,
Riquelme I, Zambrano T, Peña D, Alberdi M, Reyes-Díaz M
(2020) Anthocyanin-rich extracts of Calafate (Berberis micro-
phylla G. Forst.) fruits decrease invitro viability and migration
of human gastric and gallbladder cancer cell lines. J Soil Sci
Plant Nutr 20:1891–1903
Chen Y, Lian P, Liu Y, Xu K (2015) Pachymic acid inhibits tumorigen-
esis in gallbladder carcinoma cells. Int J Clin Exp Med 8:17781
Chen H, Tian X, Luan Y, Lu H (2019a) Downregulated long noncod-
ing RNA DGCR5 acts as a new promising biomarker for the
diagnosis and prognosis of ovarian cancer. Technol Cancer Res
Treat 18:1533033819896809
Chen J, Yu Y, Li H, Hu Q, Chen X, He Y, Xue C, Ren F, Ren Z, Li J,
Liu L, Duan Z, Cui G, Sun R (2019b) Long non-coding RNA
PVT1 promotes tumor progression by regulating the miR-143/
HK2 axis in gallbladder cancer. Mol Cancer 18:33
Colognori D, Sunwoo H, Kriz AJ, Wang CY, Lee JT (2019) Xist dele-
tional analysis reveals an interdependency between Xist RNA
and polycomb complexes for spreading along the inactive X. Mol
Cell 74:101-117.e110
Dilek ON, Karasu S, Dilek FH (2019) Diagnosis and treatment of gall-
bladder polyps: current perspectives. Eur J Hepato-Gastroenterol
9:40
Do H, Kim W (2018) Roles of oncogenic long non-coding RNAs in
cancer development. Genom Inform 16:e18–e18
Doghish AS, Radwan AF, Zaki MB, Elfar N, Moussa R, Walash Z,
Alhamshry NAA, Mohammed OA, Abdel-Reheim MA, Elimam
H (2024a) Decoding the role of long non-coding RNAs in gall-
bladder cancer pathogenesis: a review focus on signaling path-
ways interplay. Int J Biol Macromol 264:130426
Doghish AS, Zaki MB, Eldeib MG, Radwan AF, Moussa R, Abdel-
Wahab MM, Kizilaslan EZ, Alhamshry NAA, Ashour AE,
Elimam H (2024b) The potential relevance of long non-coding
RNAs in colorectal cancer pathogenesis and treatment: a review
focus on signaling pathways. Pathol Res Pract 253:155044
Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S (2022) Long non-
coding RNAs (lncRNAs) signaling in cancer chemoresistance:
from prediction to druggability. Drug Resist Updat 65:100866
Fu XD (2014) Non-coding RNA: a new frontier in regulatory biology.
Nat Sci Rev 1:190–190
Fu C, Nie L, Yin T, Xu X, Lu W (2021) LncRNA EPIC1 promotes
proliferation and inhibits apoptosis of gallbladder cancer cells
by interacting with LET. Annals Hepatol 26:100563
Gao H, Xie J, Peng J, Han Y, Jiang Q, Han M, Wang C (2015) His-
pidulin inhibits proliferation and enhances chemosensitivity
of gallbladder cancer cells by targeting HIF-1α. Exp Cell Res
332:236–246
Gao N, Li Y, Li J, Gao Z, Yang Z, Li Y, Liu H, Fan T (2020) Long non-
coding RNAs: the regulatory mechanisms, research strategies,
and future directions in cancers. Front Oncol 10:598817–598817
Gao J, Dai C, Yu X, Yin X-B, Liao W-J, Huang Y, Zhou F (2021a)
Silencing of long non-coding RNA FOXD2-AS1 inhibits the
progression of gallbladder cancer by mediating methylation of
MLH1. Gene Ther 28:306–318
Naunyn-Schmiedeberg's Archives of Pharmacology
Gao J, Dai C, Yu X, Yin XB, Liao WJ, Huang Y, Zhou F (2021b)
Silencing of long non-coding RNA FOXD2-AS1 inhibits the
progression of gallbladder cancer by mediating methylation of
MLH1. Gene Ther 28:306–318
Ghafouri-Fard S, Esmaeili M, Taheri M (2020) H19 lncRNA: roles
in tumorigenesis. Biomed Pharmacother Biomed Pharmacother.
123:109774
Giuliani B, Tordonato C, Nicassio F (2023) Mechanisms of long non-
coding RNA in breast cancer. Int J Mol Sci 24:4538–4538
Gu X, Zhang Q, Zhang W, Zhu L (2019) Curcumin inhibits liver metas-
tasis of gastric cancer through reducing circulating tumor cells.
Aging (Albany NY) 11:1501
Guh CY, Hsieh YH, Chu HP (2020) Functions and properties of
nuclear lncRNAs-from systematically mapping the interactomes
of lncRNAs. J Biomed Sci 27:1–14
Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai
M-C, Hung T, Argani P, Rinn JL (2010) Long non-coding RNA
HOTAIR reprograms chromatin state to promote cancer metas-
tasis. Nature 464:1071–1076
Gupta SC, Patchva S, Aggarwal BB (2013) Therapeutic roles of
curcumin: lessons learned from clinical trials. AAPS Journal
15:195–218
Hajjari M, Salavaty A (2015) HOTAIR: an oncogenic long non-coding
RNA in different cancers. Cancer Biol Med 12:1–1
Hamidi AA, Khalili-Tanha G, Nasrpour Navaei Z, Moghbeli M (2022)
Long non-coding RNAs as the critical regulators of epithelial
mesenchymal transition in colorectal tumor cells: an overview.
Cancer Cell Int 22:1–15
Hao Y, Baker D, Dijke PT (2019) TGF-β-mediated epithelial-
mesenchymal transition and cancer metastasis. Int J Mol Sci
20:2767–2767
Hu Y-P, Jin Y-P, Wu X-S, Yang Y, Li Y-S, Li H-F, Xiang S-S, Song
X-L, Jiang L, Zhang Y-J (2019a) LncRNA-HGBC stabilized by
HuR promotes gallbladder cancer progression by regulating miR-
502-3p/SET/AKT axis. Mol Cancer 18:1–18
Hu YP, Jin YP, Wu XS, Yang Y, Li YS, Li HF, Xiang SS, Song XL,
Jiang L, Zhang YJ, Huang W, Chen SL, Liu FT, Chen C, Zhu Q,
Chen HZ, Shao R, Liu YB (2019b) LncRNA-HGBC stabilized
by HuR promotes gallbladder cancer progression by regulating
miR-502-3p/SET/AKT axis. Mol Cancer 18:167
Huang B, Zhai M, Qin A, Wu J, Jiang X, Qiao Z (2020) Sinensetin
flavone exhibits potent anticancer activity against drug-resistant
human gallbladder adenocarcinoma cells by targeting PTEN/
PI3K/AKT signalling pathway, induces cellular apoptosis and
inhibits cell migration and invasion. J BUON 25:1251–1256
Hundal R, Shaffer EA (2014) Gallbladder cancer: epidemiology and
outcome. Clin Epidemiol 6:99–99
Irving GR, Karmokar A, Berry DP, Brown K, Steward WP (2011)
Curcumin: the potential for efficacy in gastrointestinal diseases.
Best Pract Res Clin Gastroenterol 25:519–534
Jafari-Raddani F, Davoodi-Moghaddam Z, Yousefi AM, Ghaffari SH,
Bashash D (2022) An overview of long noncoding RNAs: biol-
ogy, functions, therapeutics, analysis methods, and bioinformat-
ics tools. Cell Biochem Funct 40:800–825
Jeon HM, Lee J (2017) MET: roles in epithelial-mesenchymal transi-
tion and cancer stemness. Annals Trans Med 5:5–5
Jiang M-C, Ni J-J, Cui W-Y, Wang B-Y, Zhuo W (2019) Emerging
roles of lncRNA in cancer and therapeutic opportunities. Am J
Cancer Res 9:1354–1354
Jin L, Cai Q, Wang S, Wang S, Mondal T, Wang J, Quan Z (2018)
Long noncoding RNA MEG3 regulates LATS2 by promoting the
ubiquitination of EZH2 and inhibits proliferation and invasion in
gallbladder cancer. Cell Death Disease 9:1017
Jin L, Cai Q, Wang S, Wang S, Wang J, Quan Z (2020a) Long noncod-
ing RNA PVT1 promoted gallbladder cancer proliferation by
epigenetically suppressing miR-18b-5p via DNA methylation.
Cell Death Dis 11:871
Jin L, Cai Q, Wang S, Wang S, Wang J, Quan Z (2020b) Long noncod-
ing RNA PVT1 promoted gallbladder cancer proliferation by
epigenetically suppressing miR-18b-5p via DNA methylation.
Cell Death Disease 11:871
Kaikkonen MU, Lam MTY, Glass CK (2011) Non-coding RNAs as
regulators of gene expression and epigenetics. Cardiovasc Res
90:430–440
Khandelwal A, Malhotra A, Jain M, Vasquez KM, Jain A (2017) The
emerging role of long non-coding RNA in gallbladder cancer
pathogenesis. Biochimie 132:152–160
Kortam MA, Elfar N, Shaker OG, El-Boghdady NA, Abd-Elmawla MA
(2023) MAGI2-AS3 and miR-374b-5p as putative regulators of
multiple sclerosis via modulating the PTEN/AKT/IRF-3/IFN-β
axis: new clinical insights. ACS Chem Neurosci 14:1107–1118
Krinsky ML (2004) Cholangiocarcinoma. Encycloped Gastroenterol:
127:301–304.https:// linki nghub. elsev ier . com/ retri eve/ pii/ B0123
86860 20011 79
Kung JTY, Colognori D, Lee JT (2013) Long noncoding RNAs: past,
present, and future. Genetics 193:651–651
Lai J, Yang S, Lin Z, Huang W, Li X, Li R, Tan J, Wang W (2023)
Update on chemoresistance mechanisms to first-line chemother-
apy for gallbladder cancer and potential reversal strategies. Am
J Clin Oncol 46:131
Li K, Tang J, Hou Y (2019a) LncRNA GATA6-AS inhibits cancer cell
migration and invasion in gallbladder cancer by downregulating
miR-421. Onco Targets Ther 12:8047
Li K, Tang J, Hou Y (2019b) LncRNA GATA6-AS inhibits cancer cell
migration and invasion in gallbladder cancer by downregulating
miR-421. OncoTargets Ther 12:8047–8053
Li Z, Ding Z, Rong D, Tang W, Cao H (2019c) Overexpression of
lncRNA AFAP1-AS1 promotes cell proliferation and invasion
in gastric cancer. Oncol Lett 18:3211–3217
Li H, Hu Y, Jin Y, Zhu Y, Hao Y, Liu F, Yang Y, Li G, Song X, Ye Y
(2020a) Long noncoding RNA lncGALM increases risk of liver
metastasis in gallbladder cancer through facilitating N-cadherin
and IL-1β-dependent liver arrest and tumor extravasation. Clin
Trans Med 10:e201
Li J, Zhang H, Luo H (2020b) Long non-coding RNA OIP5-AS1 con-
tributes to gallbladder cancer cell invasion and migration by
miR-143-3p suppression. Cancer Manag Res 12:12983–12992
Li J, Cen W, Tong C, Wang L, Zhang W, Deng S, Yu J (2022a)
Fangchinoline induces gallbladder cancer cell apoptosis by sup-
pressing PI3K/Akt/XIAP axis. Plos One 17:e0266738
Li J, Ming Z, Yang L, Wang T, Liu G, Ma Q (2022b) Long noncoding
RNA XIST: mechanisms for X chromosome inactivation, roles
in sex-biased diseases, and therapeutic opportunities. Genes Dis-
eases 9:1478–1478
Li Y, Tian M, Zhang D, Zhuang Y, Li Z, Xie S, Sun K (2021) Long
non-coding RNA myosin light chain kinase antisense 1 plays an
oncogenic role in gallbladder carcinoma by promoting chem-
oresistance and proliferation. Cancer Manage Res, 6219-6230
Liang C, Yang P, Han T, Wang R-Y, Xing X-L, Si A-F, Ma Q-Y,
Chen Z, Li H-Y, Zhang B (2019) Long non-coding RNA DILC
promotes the progression of gallbladder carcinoma. Gene
694:102–110
Liu T-Y, Tan Z-J, Jiang L, Gu J-F, Wu X-S, Cao Y, Li M-L, Wu K-J,
Liu Y-B (2013) Curcumin induces apoptosis in gallbladder car-
cinoma cell line GBC-SD cells. Cancer Cell Int 13:1–9
Liu T-Y, Gong W, Tan Z-J, Lu W, Wu X-S, Weng H, Ding Q, Shu
Y-J, Bao R-F, Cao Y (2015) Baicalein inhibits progression of
gallbladder cancer cells by downregulating ZFX. PloS One
10:e0114851
Naunyn-Schmiedeberg's Archives of Pharmacology
Liu B, Shen E-D, Liao M-M, Hu Y-B, Wu K, Yang P, Zhou L, Chen
W-D (2016a) Expression and mechanisms of long non-coding
RNA genes MEG3 and ANRIL in gallbladder cancer. Tumor
Biol 37:9875–9886
Liu B, Shen ED, Liao MM, Hu YB, Wu K, Yang P, Zhou L, Chen WD
(2016b) Expression and mechanisms of long non-coding RNA
genes MEG3 and ANRIL in gallbladder cancer. Tumour Biol
37:9875–9886
Liu Z, Chen Q, Hann SS (2019) The functions and oncogenic roles of
CCAT1 in human cancer. Biomed Pharmacother Biomed Phar-
macoth 115:108943
Liu K, Xu Q (2020) LncRNA PVT1 regulates gallbladder cancer pro-
gression through miR-30d-5p. J Biol Reg Homeostat Agents
34:875–883
Liu X-F, Wang K, Du H-C (2020d) LncRNA SNHG6 regulating
Hedgehog signaling pathway and affecting the biological func-
tion of gallbladder carcinoma cells through targeting miR-
26b-5p. Eur Rev Med Pharmacol Sci 24:7598–7611
Liu L, Yan Y, Zhang G, Chen C, Shen W, Xing P (2020a) Knockdown
of LINC01694 inhibits growth of gallbladder cancer cells via
miR-340-5p/Sox4. Bioscience Reports 40: BSR20194444
Liu S, Chu B, Cai C, Wu X, Yao W, Wu Z, Yang Z, Li F, Liu Y, Dong
P (2020b) DGCR5 promotes gallbladder cancer by sponging
MiR-3619-5p via MEK/ERK1/2 and JNK/p38 MAPK pathways.
J Cancer 11:5466
Liu S, Chu B, Cai C, Wu X, Yao W, Wu Z, Yang Z, Li F, Liu Y, Dong
P, Gong W (2020c) DGCR5 promotes gallbladder cancer by
sponging MiR-3619-5p via MEK/ERK1/2 and JNK/p38 MAPK
pathways. J Cancer 11:5466–5477
Liu CX, Chen LL (2022) Circular RNAs: characterization, cellular
roles, and applications. Cell 185:2016–2034
Lopes Vendrami C, Magnetta MJ, Mittal PK, Moreno CC, Miller FH
(2021) Gallbladder carcinoma and its differential diagnosis at
MRI: what radiologists should know. Radiographics 41:78–95
Lu Q, Yu T, Ou X, Cao D, Xie T, Chen X (2017) Potential lncRNA
diagnostic biomarkers for early gastric cancer. Mol Med Rep
16:9545–9552
Ludwig N, Werner TV, Backes C, Trampert P, Gessler M, Keller A,
Lenhof H-P, Graf N, Meese E (2016) Combining miRNA and
mRNA expression profiles in Wilms tumor subtypes. Int J Mol
Sci 17:475
Lv W, Wang L, Lu J, Mu J, Liu Y, Dong P (2015) Long noncoding
RNA KIAA0125 potentiates cell migration and invasion in gall-
bladder cancer. BioMed Res Int 2015:1–9
Ma MZ, Li CX, Zhang Y, Weng MZ, Zhang MD, Qin YY, Gong W,
Quan ZW (2014) Long non-coding RNA HOTAIR, a c-Myc acti-
vated driver of malignancy, negatively regulates miRNA-130a in
gallbladder cancer. Mol Cancer 13:156
Ma MZ, Kong X, Weng MZ, Zhang MD, Qin YY, Gong W, Zhang
WJ, Quan ZW (2015a) Long non-coding RNA-LET is a posi-
tive prognostic factor and exhibits tumor-suppressive activity in
gallbladder cancer. Mol Carcinogen 54:1397–1406
Ma M, Chu B, Zhang Y, Weng M, Qin Y, Gong W, Quan Z (2015b)
Long non-coding RNA CCAT1 promotes gallbladder cancer
development via negative modulation of miRNA-218-5p. Cell
Death Disease 6:e1583–e1583
Ma F, Wang S-h, Cai Q, Jin L-y, Zhou D, Ding J, Quan Z-w (2017)
Long non-coding RNA TUG1 promotes cell proliferation and
metastasis by negatively regulating miR-300 in gallbladder car-
cinoma. Biomed Pharmacother 88:863–869
Mandrekar JN (2010) Receiver operating characteristic curve in diag-
nostic test assessment. J Thorac Oncol 5:1315–1316
Mangiavacchi A, Morelli G, Orlando V (2023) Behind the scenes: How
RNA orchestrates the epigenetic regulation of gene expression.
Front Cell Dev Biol 11:1123975–1123975
Mattick JS, Amaral PP, Carninci P, Carpenter S, Chang HY, Chen LL,
Chen R, Dean C, Dinger ME, Fitzgerald KA, Gingeras TR, Gutt-
man M, Hirose T, Huarte M, Johnson R, Kanduri C, Kapranov
P, Lawrence JB, Lee JT, Mendell JT, Mercer TR, Moore KJ,
Nakagawa S, Rinn JL, Spector DL, Ulitsky I, Wan Y, Wilusz
JE (2023) Wu M (2023) Long non-coding RNAs: definitions,
functions, challenges and recommendations. Nat Rev Mol Cell
Biol 24:430–447
Mehana NA, Ghaiad HR, Hassan M, Elsabagh YA, Labib S, Abd-
Elmawla MA (2022) LncRNA MEG3 regulates the interplay
between Th17 and Treg cells in Behçet’s disease and systemic
lupus erythematosus. Life Sci 309:120965
Meseure D, Alsibai KD, Nicolas A, Bieche I, Morillon A (2015) Long
noncoding RNAs as new architects in cancer epigenetics, prog-
nostic biomarkers, and potential therapeutic targets. BioMed Res
Int 2015:320214
Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A (2019)
Alkaloids for cancer prevention and therapy: current progress
and future perspectives. Eur J Pharmacol 858:172472
Niu J-Z, Liang X-C, Xu Z-W, Li Z-H, Li J, Meng Y, Sun Z-W (2020)
Long non-coding RNA Linc00261 as a novel potential diagnostic
and prognostic biomarker for gallbladder cancer. Trans Cancer
Res 9:6078
Nobili L, Ronchetti D, Taiana E, Neri A (2017) Long non-coding
RNAs in B-cell malignancies: a comprehensive overview. Onco-
target 8:60605–60605
Ono M, Higuchi T, Takeshima M, Chen C, Nakano S (2013a) Anti-
proliferative and apoptosis-inducing activity of curcumin against
human gallbladder adenocarcinoma cells. Anticancer Res
33:1861–1866
Ono M, Higuchi T, Takeshima M, Chen C, Nakano S (2013b) Differ-
ential anti-tumor activities of curcumin against apoptosis and cell
cycle progression in Src-and Ras-activated human gallbladder
carcinoma cells. Cancer Res 73:2575–2575
Özdemir Z, Wimmer Z (2022) Selected plant triterpenoids and their
amide derivatives in cancer treatment: A review. Phytochemistry
203:113340
Pawłowska E, Szczepanska J, Blasiak J (2017) The long noncoding
RNA HOTAIR in breast cancer: does autophagy play a role? Int
J Mol Sci 18:2317–2317
Pérez-Moreno P, Riquelme I, Brebi P, Roa JC (2021) Role of lncRNAs
in the development of an aggressive phenotype in gallbladder
cancer. J Clin Med 10:4206
Rahman MM, Rahaman MS, Islam MR, Rahman F, Mithi FM,
Alqahtani T, Almikhlafi MA, Alghamdi SQ, Alruwaili AS, Hos-
sain MS, Ahmed M, Das R, Emran TB, Uddin MS (2022) Role
of phenolic compounds in human disease: current knowledge and
future prospects. Molecules 27:233
Ratti M, Lampis A, Ghidini M, Salati M, Mirchev MB, Valeri N, Hahne
JC (2020) MicroRNAs (miRNAs) and long non-coding RNAs
(lncRNAs) as new tools for cancer therapy: first steps from bench
to bedside. Target Oncol 15:261–278
Scacalossi KR, van Solingen C, Moore KJ (2019) Long non-coding
RNAs regulating macrophage functions in homeostasis and dis-
ease. Vasc Pharmacol 114:122–122
Schmitt AM, Chang HY (2016) Long noncoding RNAs in cancer path-
ways. Cancer Cell 29:452–463
Schwerdtfeger M, Desiderio V, Kobold S, Regad T, Zappavigna S,
Caraglia M (2021) Long non-coding RNAs in cancer stem cells.
Trans Oncol 14:101134
Shahin RK, Elkady MA, Abulsoud AI, Abdelmaksoud NM, Abdel
Mageed SS, El-Dakroury WA, Zewail MB, Elazazy M, Sobhy
MH, Nomier Y, Elazazy O, Elballal MS, Mohammed OA,
Midan HM, Elrebehy MA, Ziada BO, Doghish AS (2023) miR-
NAs orchestration of gallbladder cancer – particular emphasis
Naunyn-Schmiedeberg's Archives of Pharmacology
on diagnosis, progression and drug resistance. Pathol Res Pract
248:154684–154684
Shen XH, Qi P, Du X (2015) Long non-coding RNAs in cancer
invasion and metastasis. Modern Pathol Off J United States
Canadian Acad Pathol Inc 28: 4-13
Shen Y, Bian R, Li Y, Gao Y, Liu Y, Xu Y, Song X, Zhang Y (2019)
Liensinine induces gallbladder cancer apoptosis and G2/M
arrest by inhibiting ZFX-induced PI3K/AKT pathway. Acta
Biochimica Et Biophysica Sinica 51:607–614
Shi T, Gao G, Cao Y (2016) Long noncoding RNAs as novel bio-
markers have a promising future in cancer diagnostics. Disease
Mark 2016:1–10
Shi Y, Wang XX, Zhuang YW, Jiang Y, Melcher K, Xu HE (2017)
Structure of the PRC2 complex and application to drug discov-
ery. Acta Pharmacol Sinica 38:963–963
Song X, Wang Z, Liang H, Zhang W, Ye Y, Li H, Hu Y, Zhang Y,
Weng H, Lu J, Wang X, Li M, Liu Y, Gu J (2017a) Dioscin
induces gallbladder cancer apoptosis by inhibiting ROS-medi-
ated PI3K/AKT signalling. Int J Biol Sci 13:782–793
Song X-L, Zhang Y-J, Wang X-F, Zhang W-J, Wang Z, Zhang F,
Zhang Y-J, Lu J-H, Mei J-W, Hu Y-P (2017b) Casticin induces
apoptosis and G0/G1 cell cycle arrest in gallbladder cancer
cells. Cancer Cell Int 17:1–10
Statello L, Guo CJ, Chen LL, Huarte M (2020) Gene regulation by
long non-coding RNAs and its biological functions. Nat Rev
Mol Cell Biol 22:96–118
Tang ZY, Li Y, Tang YT, Ma XD, Tang ZY (2022) Anticancer activ-
ity of oleanolic acid and its derivatives: Recent advances in
evidence, target profiling and mechanisms of action. Biomed
Pharmacother 145:112397
Taniue K, Akimitsu N (2021) The functions and unique features of
LncRNAs in cancer development and tumorigenesis. Int J Mol
Sci 22:632–632
Tano K, Akimitsu N (2012) Long non-coding RNAs in cancer pro-
gression. Front Gen 3:219
Tsai KW, Lo YH, Liu H, Yeh CY, Chen YZ, Hsu CW, Chen WS,
Wang JH (2018) Linc00659, a long noncoding RNA, acts as
novel oncogene in regulating cancer cell growth in colorectal
cancer. Mol Cancer 17:72
Tyagi S, Sharma A, Upadhyay SK, Tyagi S, Sharma A, Upadhyay
SK (2018) Role of next-generation RNA-Seq data in discovery
and characterization of long non-coding RNA in plants. Next
Gen Plant Breed 6:111–128
Ulitsky I, Bartel DP (2013) lincRNAs: genomics, evolution, and
mechanisms. Cell 154:26–26
Vijayakumar A, Vijayakumar A, Patil V, Mallikarjuna MN, Shivas-
wamy B (2013) Early diagnosis of gallbladder carcinoma: an
algorithm approach. ISRN Radiology 2013(1–6):239424
Wang L, Zhang Y, Lv W, Lu J, Mu J, Liu Y, Dong P (2015) Retracted:
long non-coding RNA Linc-ITGB 1 knockdown inhibits cell
migration and invasion in GBC-SD/M and GBC-SD gallblad-
der cancer cell lines. Chem Biol Drug Des 86:1064–1071
Wang D, Li Z, Zhang Y, Wang G, Wei M, Hu Y, Ma S, Jiang Y, Che
N, Wang X (2016a) Targeting of micro RNA-199a-5p protects
against pilocarpine-induced status epilepticus and seizure dam-
age via SIRT 1–p53 cascade. Epilepsia 57:706–716
Wang S-H, Ma F, Tang Z-h, Wu X-C, Cai Q, Zhang M-D, Weng
M-Z, Zhou D, Wang J-D, Quan Z-W (2016b) Long non-coding
RNA H19 regulates FOXM1 expression by competitively bind-
ing endogenous miR-342-3p in gallbladder cancer. J Exp Clin
Cancer Res 35:1–12
Wang S-H, Wu X-C, Zhang M-D, Weng M-Z, Zhou D, Quan Z-W
(2016c) Long noncoding RNA H19 contributes to gallbladder
cancer cell proliferation by modulated miR-194-5p targeting
AKT2. Tumor Biol 37:9721–9730
Wang S-H, Zhang M-D, Wu X-C, Weng M-Z, Zhou D, Quan Z-W
(2016d) Overexpression of LncRNA-ROR predicts a poor
outcome in gallbladder cancer patients and promotes the
tumor cells proliferation, migration, and invasion. Tumor Biol
37:12867–12875
Wang SH, Wu XC, Zhang MD, Weng MZ, Zhou D, Quan ZW (2016e)
Long noncoding RNA H19 contributes to gallbladder cancer cell
proliferation by modulated miR-194-5p targeting AKT2. Tumour
Biol J Int Socr Oncodevel Biol Med 37:9721–9730
Wang SH, Wu XC, Zhang MD, Weng MZ, Zhou D, Quan ZW (2016f)
Upregulation of H19 indicates a poor prognosis in gallbladder
carcinoma and promotes epithelial-mesenchymal transition. Am
J Cancer Res 6:15–26
Wang SH, Yang Y, Wu XC, Zhang MD, Weng MZ, Zhou D, Wang
JD, Quan ZW (2016g) Long non-coding RNA MINCR pro-
motes gallbladder cancer progression through stimulating EZH2
expression. Cancer Lett 380:122–133
Wang J, Pan Y, Hu J, Ma Q, Xu Y, Zhang Y, Zhang F, Liu Y (2018) Tea
polyphenols induce S phase arrest and apoptosis in gallbladder
cancer cells. Brazil J Med Biol Res 51:e6891
Wang F, Mayca Pozo F, Tian D, Geng X, Yao X, Zhang Y, Tang J
(2020a) Shikonin inhibits cancer through P21 upregulation and
apoptosis induction. Front Pharmacol 11:861
Wang Y, Dou L, Qin Y, Yang H, Yan P (2020b) OIP5-AS1 contributes
to tumorigenesis in hepatocellular carcinoma by miR-300/YY1-
activated WNT pathway. Cancer Cell Int 20:440
Wang S, Wang Y, Wang S, Tong H, Tang Z, Wang J, Zhang Y, Ou J,
Quan Z (2021) Long non-coding RNA FIRRE acts as a miR-
520a-3p sponge to promote gallbladder cancer progression via
mediating YOD1 expression. Front Genet 12:674653
Wang SH, Ma F, Tang ZH, Wu XC, Cai Q, Zhang MD, Weng MZ,
Zhou D, Wang JD, Quan ZW (2022) Correction to: long non-
coding RNA H19 regulates FOXM1 expression by competitively
binding endogenous miR-342-3p in gallbladder cancer. J Exp
Clin Cancer Res CR 41:55
Wang Y, Bai H, Jiang M, Zhou C, Gong Z (2023) Emerging role of
long non-coding RNA JPX in malignant processes and potential
applications in cancers. Chin Med J 136:757–757
Worku T, Bhattarai D, Ayers D, Wang K, Wang C, Rehman ZU, Tal-
pur HS, Yang L (2017) Long non-coding RNAs: the new hori-
zon of gene regulation in ovarian cancer. Cell Physiol Biochem
44:948–966
Wu X-S, Wang X-A, Wu W-G, Hu Y-P, Li M-L, Ding Q, Weng H, Shu
Y-J, Liu T-Y, Jiang L (2014a) MALAT1 promotes the prolifera-
tion and metastasis of gallbladder cancer cells by activating the
ERK/MAPK pathway. Cancer Biol Ther 15:806–814
Wu X-S, Yang T, Gu J, Li M-L, Wu W-G, Weng H, Ding Q, Mu J-S,
Bao R-F, Shu Y-J (2014b) Effects of oxymatrine on the apoptosis
and proliferation of gallbladder cancer cells. Anti-cancer Drugs
25:1007–1015
Wu XS, Wang F, Li HF, Hu YP, Jiang L, Zhang F, Li ML, Wang XA,
Jin YP, Zhang YJ (2017) Lnc RNA-PAGBC acts as a micro RNA
sponge and promotes gallbladder tumorigenesis. EMBO Reports
18:1837–1853
Xie L, Zhang Q, Mao J, Zhang J, Li L (2021a) The roles of lncRNA
in myocardial infarction: molecular mechanisms, diagnosis
biomarkers, and therapeutic perspectives. Front Cell Dev Biol
9:680713
Xie L, Zhang Q, Mao J, Zhang J, Li L (2021b) The roles of lncRNA
in myocardial infarction: molecular mechanisms, diagnosis bio-
markers, and therapeutic perspectives. Front Cell Devel Biol
9:680713
Xue Z, Yang B, Xu Q, Zhu X, Qin G (2020) Long non-coding RNA
SSTR5-AS1 facilitates gemcitabine resistance via stabilizing
NONO in gallbladder carcinoma. Biochem Biophys Res Com-
mun 522:952–959
Naunyn-Schmiedeberg's Archives of Pharmacology
Yan M, Zhang L, Li G, Xiao S, Dai J, Cen X (2017) Long noncoding
RNA linc-ITGB1 promotes cell migration and invasion in human
breast cancer. Biotechnol Appl Biochem 64:5–13
Yang L, Cheng X, Ge N, Guo W, Feng F, Wan F (2017) Long non-
coding RNA SPRY4-IT1 promotes gallbladder carcinoma pro-
gression. Oncotarget 8:3104
Yang L, Gao Q, Wu X, Feng F, Xu K (2018) Long noncoding RNA
HEGBC promotes tumorigenesis and metastasis of gallbladder
cancer via forming a positive feedback loop with IL-11/STAT3
signaling pathway. J Exp Clin Cancer Res CR 37:186
Yang P, Song F, Yang X, Yan X, Huang X, Qiu Z, Wen Z, Liang C,
Xin X, Lei Z (2022) Exosomal MicroRNA signature acts as an
efficient biomarker for non-invasive diagnosis of gallbladder car-
cinoma. Iscience 25:104868
Yang S, Qin L, Wu P, Liu Y, Zhang Y, Mao B, Yan Y, Yan S, Tan F,
Yue X, Liu H, Xue H (2023) RNA sequencing revealed the multi-
stage transcriptome transformations during the development of
gallbladder cancer associated with chronic inflammation. PloS
One 18:e0283770–e0283770
Yang G, Lu X, Yuan L (2014) LncRNA: a link between RNA and
cancer. Biochimica et Biophysica Acta (BBA)-Gene Regulatory
Mechanisms 1839: 1097-1109
Zang L, Xu H, Huang C, Wang C, Wang R, Chen Y, Wang L, Wang H
(2022) A link between chemical structure and biological activity
in triterpenoids. Recent Pat Anticancer Drug Discov 17:145–161
Zhai T, Hei Z, Ma Q, Liang H, Xu Y, Zhang Y, Jin L, Han C, Wang
J (2017) Shikonin induces apoptosis and G0/G1 phase arrest of
gallbladder cancer cells via the JNK signaling pathway. Oncol
Rep 38:3473–3480
Zhai T, Zhang X, Hei Z, Jin L, Han C, Ko AT, Yu X, Wang J (2021)
Isorhamnetin inhibits human gallbladder cancer cell proliferation
and metastasis via PI3K/AKT signaling pathway inactivation.
Front Pharmacol 12:628621
Zhang Y, Shields T, Crenshaw T, Hao Y, Moulton T, Tycko B (1993)
Imprinting of human H19: allele-specific CpG methylation, loss
of the active allele in Wilms tumor, and potential for somatic
allele switching. Am J Human Genet 53:113–124
Zhang D-c, Liu J-l, Ding Y-b, Xia J-g, Chen G-y (2013) Icariin potenti-
ates the antitumor activity of gemcitabine in gallbladder cancer
by suppressing NF-κB. Acta Pharmacologica Sinica 34:301–308
Zhang M, Cai S, Zuo B, Gong W, Tang Z, Zhou D, Weng M, Qin Y,
Wang S, Liu J (2017a) Arctigenin induced gallbladder cancer
senescence through modulating epidermal growth factor receptor
pathway. Tumor Biol 39:1010428317698359
Zhang P, Cao P, Zhu X, Pan M, Zhong K, He R, Li Y, Jiao X, Gao
Y (2017b) Upregulation of long non-coding RNA HOXA-AS2
promotes proliferation and induces epithelial-mesenchymal tran-
sition in gallbladder carcinoma. Oncotarget 8:33137
Zhang Y, Li R, Ding X, Zhang K, Qin W (2019) Upregulation of long
non-coding RNA SNHG6 promote esophageal squamous cell
carcinoma cell malignancy and its diagnostic value. Am J Trans
Res 11:1084
Zhang C, Niu K, Lian P, Hu Y, Shuai Z, Gao S, Ge S, Xu T, Xiao Q,
Chen Z (2021) Pathological bases and clinical application of
long noncoding RNAs in cardiovascular diseases. Hypertension
78:16–29
Zhang Z, Shi S, Li J, Costa M (2023) Long non-coding RNA MEG3
in metal carcinogenesis. Toxics 11:157–157
Zhao H, Zhang X, Frazão JB, Condino-Neto A, Newburger PE (2013)
HOX antisense lincRNA HOXA-AS2 is an apoptosis repressor in
all trans retinoic acid treated NB4 promyelocytic leukemia cells.
J Cell Biochem 114:2375–2383
Zheng B, Wang J, Fan K, Sun W, Wan W, Gao Z, Ni X, Zhang D, Ni
X, Suo T (2021) lncRNA RP11-147L13. 8 suppresses metasta-
sis and chemo-resistance by modulating the phosphorylation of
c-Jun protein in GBC. Mol Ther-Oncol 23:124–137
Zhong Y, Wu X, Li Q, Ge X, Wang F, Wu P, Deng X, Miao L (2019)
Long noncoding RNAs as potential biomarkers and therapeu-
tic targets in gallbladder cancer: a systematic review and meta-
analysis. Cancer Cell Int 19:1–10
Zhou M, Diao Z, Yue X, Chen Y, Zhao H, Cheng L, Sun J (2016)
Construction and analysis of dysregulated lncRNA-associated
ceRNA network identified novel lncRNA biomarkers for early
diagnosis of human pancreatic cancer. Oncotarget 7:56383
Zhou W, Xu X, Cen Y, Chen J (2022) The role of lncRNAs in the
tumor microenvironment and immunotherapy of melanoma.
Front Immunol 13:1085766
Zhu X, Shi C, Hou C (2022) AFAP1-AS1/Hsa-miR-15a-5p/Bcl-2 axis
is a potential regulator of cancer cell proliferation and apoptosis
in gallbladder carcinoma. Nutr Cancer 74:3363–3374
Zou Q, Yang Z-l, Yuan Y, Li J-h, Liang L-f, Zeng G-x, Chen S-l (2013)
Clinicopathological features and CCT2 and PDIA2 expression in
gallbladder squamous/adenosquamous carcinoma and gallblad-
der adenocarcinoma. World J Surg Oncol 11:1–11
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
... The mechanism of exerting its action is carried out primarily by antioxidation, working with the principle of free radicals' action being responsible for tissue oxidation that leads to the development of tumors. This reducing property of curcumin is facilitated by the methoxy group, 1,3 β-diketone moiety, and phenolic hydroxyl group [79,80]. Studies show that Curcumin inhibits nuclear factor-kB (NF-kB), initially responsible for activating inflammatory cytokines and chemokines, leading to several inflammatory conditions [81]. ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is a leading liver cancer that significantly impacts global life expectancy and remains challenging to treat due to often late diagnoses. Despite advances in treatment, the prognosis is still poor, especially in advanced stages. Studies have pointed out that investigations into the molecular mechanisms underlying HCC, including mitochondrial dysfunction and epigenetic regulators, are potentially important targets for diagnosis and therapy. Mitoepigenetics, or the epigenetic modifications of mitochondrial DNA, have drawn wide attention for their role in HCC progression. Besides, molecular biomarkers such as mitochondrial DNA alterations and non-coding RNAs showed early diagnosis and prognosis potential. Additionally, natural compounds like alkaloids, resveratrol, curcumin, and flavonoids show promise in HCC show promise in modulating mitochondrial and epigenetic pathways involved in cancer-related processes. This review discusses how mitochondrial dysfunction and epigenetic modifications, especially mitoepigenetics, influence HCC and delves into the potential of natural products as new adjuvant treatments against HCC.
Article
Full-text available
Gallbladder cancer is among the sixth most common gastrointestinal malignancies, with a meager prognosis. The progression of the disease is influenced by factors like chronic inflammation and geographical locations. Current treatment options are limited and often ineffective, emphasizing the need for novel therapeutic approaches. This paper explores potential multi-targeted natural compounds by targeting key signaling proteins associated with various hallmarks of Gallbladder cancer. In silico methods, including virtual screening, molecular docking, and molecular dynamics simulations, were utilized to assess the interactions of natural compounds with five critical targets: PD-L1, VEGFR, EGFR, HER2, and c-MET. To identify potential inhibitors, a library of anticancer natural compounds was screened against each target protein. The top ten compounds for each target were then selected for precise molecular docking. A common, promising compound was identified based on the lowest binding energy. Furthermore, DFT, bioavailability, and toxicity profiles of the selected compound were analyzed, and it was subsequently subjected to molecular dynamics simulations. Among the compounds studied, 13-beta, 21-Dihydroxyeurycomanol was a common and promising compound for each protein target, exhibiting strong binding affinities and favorable interactions. DFT analysis predicted high reactivity and strong binding interactions. Furthermore, ADMET analysis showed that it was non-toxic and safe. Molecular dynamics simulation analysis revealed that 13-beta, 21-Dihydroxyeurycomanol maintains stable complexes with all the protein targets. These findings indicate that it has the potential to be an effective multi-targeted therapeutic agent for gallbladder cancer and may aid in the development of conventional medicine-based treatments for this disease.
Article
Full-text available
Globally, the incidence and death rates associated with cancer persist in rising, despite considerable advancements in cancer therapy. Although some malignancies are manageable by a mix of chemotherapy, surgery, radiation, and targeted therapy, most malignant tumors either exhibit poor responsiveness to early identification or endure post-treatment survival. The prognosis for prostate cancer (PCa) is unfavorable since it is a perilous and lethal malignancy. The capacity of phytochemical and nutraceutical chemicals to repress oncogenic lncRNAs and activate tumor suppressor lncRNAs has garnered significant attention as a possible strategy to diminish the development, proliferation, metastasis, and invasion of cancer cells. A potential technique to treat cancer and enhance the sensitivity of cancer cells to existing conventional therapies is the use of phytochemicals with anticancer characteristics. Functional studies indicate that lncRNAs modulate drug resistance, stemness, invasion, metastasis, angiogenesis, and proliferation via interactions with tumor suppressors and oncoproteins. Among them, numerous lncRNAs, such as HOTAIR, PlncRNA1, GAS5, MEG3, LincRNA-21, and POTEF-AS1, support the development of PCa through many molecular mechanisms, including modulation of tumor suppressors and regulation of various signal pathways like PI3K/Akt, Bax/Caspase 3, P53, MAPK cascade, and TGF-β1. Other lncRNAs, in particular, MALAT-1, CCAT2, DANCR, LncRNA-ATB, PlncRNA1, LincRNA-21, POTEF-AS1, ZEB1-AS1, SChLAP1, and H19, are key players in regulating the aforementioned processes. Natural substances have shown promising anticancer benefits against PCa by altering essential signaling pathways. The overexpression of some lncRNAs is associated with advanced TNM stage, metastasis, chemoresistance, and reduced survival. LncRNAs possess crucial clinical and transitional implications in PCa, as diagnostic and prognostic biomarkers, as well as medicinal targets. To impede the progression of PCa, it is beneficial to target aberrant long non-coding RNAs using antisense oligonucleotides or small interfering RNAs (siRNAs). This prevents them from transmitting harmful messages. In summary, several precision medicine approaches may be used to rectify dysfunctional lncRNA regulatory circuits, so improving early PCa detection and eventually facilitating the conquest of this lethal disease. Due to their presence in biological fluids and tissues, they may serve as novel biomarkers. Enhancing PCa treatments mitigates resistance to chemotherapy and radiation.
Article
Full-text available
Prostate cancer (PC) ranks among the most prevalent cancers in males. Recent studies have highlighted intricate connections between long non-coding RNAs (lncRNAs), natural products, and cellular signaling in PC development. LncRNAs, which are RNA transcripts without protein-coding function, influence cell growth, programmed cell death, metastasis, and resistance to treatments through pathways like PI3K/AKT, WNT/β-catenin, and androgen receptor signaling. Certain lncRNAs, including HOTAIR and PCA3, are associated with PC progression, with potential as diagnostic markers. Natural compounds, such as curcumin and resveratrol, demonstrate anticancer effects by targeting these pathways, reducing tumor growth, and modulating lncRNA expression. For instance, curcumin suppresses HOTAIR levels, hindering PC cell proliferation and invasion. The interaction between lncRNAs and natural compounds may open new avenues for therapy, as these substances can simultaneously impact multiple signaling pathways. These complex interactions offer promising directions for developing innovative PC treatments, enhancing diagnostics, and identifying new biomarkers for improved prevention and targeted therapy. This review aims to map the multifaceted relationship among natural products, lncRNAs, and signaling pathways in PC pathogenesis, focusing on key pathways such as AR, PI3K/AKT/mTOR, WNT/β-catenin, and MAPK, which are crucial in PC progression and therapy resistance. Regulation of these pathways by natural products and lncRNAs could lead to new insights into biomarker identification, preventive measures, and targeted PC therapies.
Article
Full-text available
Glioblastoma (GBM) is one of the most aggressive types of brain cancer, characterized by its poor prognosis and low survival rate despite current treatment modalities. Because GBM is lethal, clarifying the pathogenesis’s underlying mechanisms is important, which are still poorly understood. Recent discoveries in the fields of molecular genetics and cancer biology have demonstrated the critical role that non-coding RNAs (ncRNAs), especially long non-coding RNAs (lncRNAs), play in the molecular pathophysiology of GBM growth. LncRNAs are transcripts longer than 200 nucleotides that do not encode proteins. They are significant epigenetic modulators that control gene e expression at several levels. Their dysregulation and interactions with important signaling pathways play a major role in the malignancy and development of GBM. The increasing role of lncRNAs in GBM pathogenesis is thoroughly examined in this review, with particular attention given to their regulation mechanisms in key signaling pathways such as PI3K/AKT, Wnt/β-catenin, and p53. It also looks into lncRNAs’ potential as new biomarkers and treatment targets for GBM. In addition, the study discusses the difficulties in delivering lncRNA-based medicines across the blood-brain barrier and identifies areas that need more research to advance lncRNA-oriented treatments for this deadly cancer. Graphical abstract
Article
Full-text available
Gastric cancer (GC) remains a major public health challenge worldwide. Long non-coding RNAs (lncRNAs) play important roles in the development, progression, and resistance to the treatment of GC, as shown by recent developments in molecular characterization. Still, an in-depth investigation of the lncRNA landscape in GC is absent. However, The objective of this systematic review is to evaluate our present understanding of the role that lncRNA dysregulation plays in the etiology of GC and treatment resistance, with a focus on the underlying mechanisms and clinical implications. Research that described the functions of lncRNA in angiogenesis, stemness, epigenetics, metastasis, apoptosis, development, and resistance to key treatments was given priority. In GC, it has been discovered that a large number of lncRNAs, including MALAT1, HOTAIR, H19, and ANRIL, are aberrantly expressed and are connected with disease-related outcomes. Through various methods such as chromatin remodeling, signal transduction pathways, and microRNA sponging, they modulate hallmark cancer capabilities. Through the activation of stemness programs, epithelial-mesenchymal transition (EMT), and survival signaling, LncRNAs also control resistance to immunotherapy, chemotherapy, and targeted therapies. By clarifying their molecular roles further, we may be able to identify new treatment targets and ways to overcome resistance. This article aims to explore the interplay between lncRNAs, and GC. Specifically, the focus is on understanding how lncRNAs contribute to the etiology of GC and influence treatment resistance in patients with this disease.
Article
Full-text available
Colorectal cancer (CRC) is recognized as one of the most prevalent malignancies, both in terms of incidence and mortality rates. Current research into CRC has shed light on the molecular mechanisms driving its development. Several factors, including lifestyle, environmental influences, genetics, and diet, play significant roles in its pathogenesis. Natural compounds such as curcumin, tanshinone, lycorine, sinomenine, kaempferol, verbascoside, quercetin, berberine, and fisetin have shown great promise in the prevention and treatment of CRC. Research has also highlighted the significance of non-coding RNAs (ncRNAs) as biomarkers and therapeutic targets in CRC. Among these, long non-coding RNAs (lncRNAs) have been found to regulate the transcription of genes involved in cancer. LncRNAs contribute to cancer stem cell (CSC) proliferation, angiogenesis, epithelial-mesenchymal transition (EMT), and chemoresistance. Specific lncRNAs, including GAS5, LNC00337, HOTAIR, TPT1-AS1, cCSC1, BCAR4, TUG1, and Solh2, play crucial roles in these processes. They hold potential as novel biomarkers, detectable in bodily fluids and tissues, and could serve as therapeutic targets due to their involvement in drug resistance and sensitivity. These insights could improve CRC treatment strategies, addressing resistance to chemotherapy and radiotherapy. This review article aims to provide a comprehensive analysis of the current knowledge regarding the effectiveness of natural anti-cancer agents in CRC treatment. Additionally, it offers an in-depth evaluation of lncRNAs in CRC, their role in the disease’s progression, and their potential applications in its management. Graphical Abstract
Article
Full-text available
Anti-phospholipid syndrome (APS) is an autoimmune disease characterized by thrombosis and miscarriage events. Still, the molecular mechanisms underlying APS, which predisposes to a wide spectrum of complications, are being explored. Seventy patients with primary and secondary APS were recruited, in addition to 35 healthy subjects. Among APS groups, the gene expression levels of XIST, Gab2, and TAK1 were higher along with declined miRNA155 level compared with controls. Moreover, the sera levels of ICAM-1, VCAM-1, IL-1ꞵ, and TNF-α were highly elevated among APS groups either primary or secondary compared with controls. The lncRNA XIST was directly correlated with Gab2, TAK1, VCAM-1, ICAM-1, IL-1ꞵ, and TNF-α. The miRNA155 was inversely correlated with XIST, Gab2, and TAK1. Moreover, ROC curve analyses subscribed the predictive power of the lncRNA XIST and miRNA155, to differentiate between primary and secondary APS from control subjects. The lncRNA XIST and miRNA155 are the upstream regulators of the Gab2/TAK1 axis among APS patients via influencing the levels of VCAM-1, ICAM-1, IL1ꞵ, and TNF-α which propagates further inflammatory and immunological streams. Interestingly, the study addressed that XIST and miRNA155 may be responsible for the thrombotic and miscarriage events associated with APS and provides new noninvasive molecular biomarkers for diagnosing the disease and tracking its progression.
Article
Full-text available
Long non-coding RNAs (lncRNAs) reportedly function as important modulators of gene regulation and malignant processes in the development of human cancers. The lncRNA JPX is a novel molecular switch for X chromosome inactivation and differentially expressed JPX has exhibited certain clinical correlations in several cancers. Notably, JPX participates in cancer growth, metastasis, and chemoresistance, by acting as a competing endogenous RNA for microRNA, interacting with proteins, and regulating some specific signaling pathways. Moreover, JPX may serve as a potential biomarker and therapeutic target for the diagnosis, prognosis, and treatment of cancer. The present article summarizes our current understanding of the structure, expression, and function of JPX in malignant cancer processes and discusses its molecular mechanisms and potential applications in cancer biology and medicine.
Article
Full-text available
Gallbladder cancer (GBC) is a highly malignant tumor with extremely poor prognosis. Previous studies have suggested that the carcinogenesis and progression of GBC is a multi-stage and multi-step process, but most of them focused on the genome changes. And a few studies just compared the transcriptome differences between tumor tissues and adjacent noncancerous tissues. The transcriptome changes, relating to every stage of GBC evolution, have rarely been studied. We selected three cases of normal gallbladder, four cases of gallbladder with chronic inflammation induced by gallstones, five cases of early GBC, and five cases of advanced GBC, using next-generation RNA sequencing to reveal the changes in mRNAs and lncRNAs expression during the evolution of GBC. In-depth analysis of the sequencing data indicated that transcriptome changes from normal gallbladder to gallbladder with chronic inflammation were distinctly related to inflammation, lipid metabolism, and sex hormone metabolism; transcriptome changes from gallbladder with chronic inflammation to early GBC were distinctly related to immune activities and connection between cells; and the transcriptome changes from early GBC to advanced GBC were distinctly related to transmembrane transport of substances and migration of cells. Expression profiles of mRNAs and lncRNAs change significantly during the evolution of GBC, in which lipid-based metabolic abnormalities play an important promotive role, inflammation and immune activities play a key role, and membrane proteins are very highlighted molecular changes.
Article
Full-text available
Objective: Gallbladder cancer (GBC) mortality remains high and chemoresistance is increasing. This review consolidates what is known about the mechanisms of chemoresistance to inform and accelerate the development of novel GBC-specific chemotherapies. Methods: Studies related to GBC-related chemoresistance were systematically screened in PubMed using the advanced search function. Search terms included GBC, chemotherapy, and signaling pathway. Results: Analysis of existing studies showed that GBC has poor sensitivity to cisplatin, gemcitabine (GEM), and 5-fluorouracil. DNA damage repair-related proteins, including CHK1, V-SCR, and H2AX, are involved in tumor adaptation to drugs. GBC-specific chemoresistance is often accompanied by changes in the apoptosis and autophagy-related molecules, BCL-2, CRT, and GBCDRlnc1. CD44+ and CD133+ GBC cells are less resistant to GEM, indicating that tumor stem cells are also involved in chemoresistance. In addition, glucose metabolism, fat synthesis, and glutathione metabolism can influence the development of drug resistance. Finally, chemosensitizers such as lovastatin, tamoxifen, chloroquine, and verapamil are able improve the therapeutic effect of cisplatin or GEM in GBC. Conclusions: This review summarizes recent experimental and clinical studies of the molecular mechanisms of chemoresistance, including autophagy, DNA damage, tumor stem cells, mitochondrial function, and metabolism, in GBC. Information on potential chemosensitizers is also discussed. The proposed strategies to reverse chemoresistance should inform the clinical use of chemosensitizers and gene-based targeted therapy for this disease.
Article
Full-text available
The landscape of pervasive transcription in eukaryotic genomes has made space for the identification of thousands of transcripts that are difficult to frame in a specific functional category. A new class has been broadly named as long non-coding RNAs (lncRNAs) and shortly defined as transcripts that are longer than 200 nucleotides with no or limited coding potential. So far, about 19,000 lncRNAs genes have been annotated in the human genome (Gencode 41), nearly matching the number of protein-coding genes. A key scientific priority is the functional characterization of lncRNAs, a major challenge in molecular biology that has encouraged many high-throughput efforts. LncRNA studies have been stimulated by the enormous clinical potential that these molecules promise and have been based on the characterization of their expression and functional mechanisms. In this review, we illustrate some of these mechanisms as they have been pictured in the context of breast cancer.
Article
Full-text available
Most transcripts from human genomes are non-coding RNAs (ncRNAs) that are not translated into proteins. ncRNAs are divided into long (lncRNAs) and small non-coding RNAs (sncRNAs). LncRNAs regulate their target genes both transcriptionally and post-transcriptionally through interactions with proteins, RNAs, and DNAs. Maternally expressed gene 3 (MEG3), a lncRNA, functions as a tumor suppressor. MEG3 regulates cell proliferation, cell cycle, apoptosis, hypoxia, autophagy, and many other processes involved in tumor development. MEG3 is downregulated in various cancer cell lines and primary human cancers. Heavy metals, such as hexavalent chromium (Cr(VI)), arsenic, nickel, and cadmium, are confirmed human carcinogens. The exposure of cells to these metals causes a variety of cancers. Among them, lung cancer is the one that can be induced by exposure to all of these metals. In vitro studies have demonstrated that the chronic exposure of normal human bronchial epithelial cells (BEAS-2B) to these metals can cause malignant cell transformation. Metal-transformed cells have the capability to cause an increase in cell proliferation, resistance to apoptosis, elevated migration and invasion, and properties of cancer stem-like cells. Studies have revealed that MEG is downregulated in Cr(VI)-transformed cells, nickel-transformed cells, and cadmium (Cd)-transformed cells. The forced expression of MEG3 reduces the migration and invasion of Cr(VI)-transformed cells through the downregulation of the neuronal precursor of developmentally downregulated protein 9 (NEDD9). MEG3 suppresses the malignant cell transformation of nickel-transformed cells. The overexpression of MEG3 decreases Bcl-xL, causing reduced apoptosis resistance in Cd-transformed cells. This paper reviews the current knowledge of lncRNA MEG3 in metal carcinogenesis.
Article
Colorectal cancer (CRC) is one of the most frequent cancers in incidence and mortality. Despite advances in cancer biology, molecular genetics, and targeted treatments, CRC prognosis and survival have not kept pace. This is usually due to advanced staging and metastases at diagnosis. Thus, great importance has been placed upon understanding the molecular pathophysiology behind the development of CRC, which has highlighted the significance of non-coding RNA’s role and associated intracellular signaling pathways in the pathogenesis of the disease. According to recent studies, long non-coding RNAs (lncRNA), a subtype of ncRNAs whose length exceeds 200 nucleotides, have been found to have regulatory functions on multiple levels. Their actions at the transcription, post-transcriptional, translational levels, and epigenetic regulation have made them prime modulators of gene expression. Due to their role in cellular cancer hallmarks, their dysregulation has been linked to several illnesses, including cancer. Furthermore, their clinical relevance has expanded due to their possible detection in blood which has cemented them as potential future biomarkers and thus, potential targets for new therapy. This review will highlight the importance of lncRNAs and related signaling pathways in the development of CRC and their subsequent clinical applications.
Article
Gallbladder cancer (GBC) is characterized by a highly invasive nature and a poor prognosis, with adenocarcinoma being the main histological subtype. According to statistical data, patients diagnosed with advanced GBC have a survival rate of less than 5% for 5 years. Despite the novel therapeutic techniques, the unsatisfactory results could be related to the underlying biology of tumor cells and resistance to chemotherapy. Early diagnosis is more important than clinical therapy as it assists in determining the pathological stage of cancer and facilitates the selection of appropriate medication. Hence, it is very important to understand the precise pathogenesis of GBC and to discover potential novel biomarkers for early diagnosis of GBC. Non-coding RNAs, such as microRNAs, long non-coding RNAs, and circular RNAs, have been found to influence the transcriptional regulation of target genes associated with cancer, either directly or indirectly. microRNAs are a group of small, non-coding, single-stranded RNAs that are expressed endogenously. miRNAs play significant roles in various fundamental cellular processes. Therefore, miRNAs have the potential to serve as valuable biomarkers and therapeutic targets for GBC.
Article
Multiple sclerosis (MS) is a chronic disease and one of the leading causes of disability in young adults. The current study aims to investigate the pathogenesis of MS via studying the regulatory role of novel lncRNA MAGI2-AS3 in miR-374b-5p and their downstream targets PTEN/AKT/IRF-3/IFN-β and the relationship of this pathway with disease severity. Moreover, it aims to assess the role of MAGI2-AS3/miR-374b-5p as diagnostic and/or prognostic biomarkers for MS. Overall, 150 contributors were recruited: 100 patients with MS and 50 healthy volunteers. Gene expression of MAGI2-AS3, miR-374b-5p, PTEN, AKT, and IRF-3 were assessed using RT-qPCR, and IFN-β was measured by ELISA. Compared with the healthy control group, serum MAGI2-AS3 and PTEN were downregulated in MS patients, whereas miR-374b-5p, PI3K, AKT, IRF-3, and IFN-β were upregulated in MS patients. Furthermore, MAGI2-AS3 was downregulated, while miR-374b-5p was upregulated in MS patients with an expanded disability status scale (EDSS) ≥3.5, compared to patients with an EDSS <3.5. Receiver-operating-characteristic curve analysis revealed that MAGI2-AS3 and miR-374b-5p can be used in the diagnosis of MS. Remarkably, multivariate logistic analysis revealed that MAGI2-AS3, miR-374b-5p, PTEN, and AKT act as independent variables in MS. Moreover, MAGI2-AS3 was directly correlated with PTEN and inversely correlated with miR-374b-5p, AKT, and EDSS. Regarding miR-374b-5p, it was positively correlated with AKT and EDSS. In conclusion, the study showed for the first time that the crosstalk between MAGI2-AS3 and miR-374b-5p could affect the AKT/IRF3/IFN-β axis in MS. Interestingly, MAGI2-AS3 and miR-374b-5p could be genetic noninvasive biomarkers for MS.