ArticlePDF AvailableLiterature Review

ZFAS1: A novel vital oncogenic lncRNA in multiple human cancers

Wiley
Cell Proliferation
Authors:

Abstract and Figures

Long noncoding RNAs (lncRNAs) are a class of noncoding, endogenous, single‐stranded RNAs longer than 200 nucleotides in length that are transcribed by RNA polymerase II. Mounting evidence has indicated that lncRNAs play key roles in several physiological and pathological processes by modifying gene expression at the transcriptional, posttranscriptional, epigenetic, and translation levels. Many reports have demonstrated that lncRNAs function as potential oncogene or tumour suppressors and thus play vital regulatory roles in tumourigenesis and tumour progression. ZNFX1 antisense RNA 1 (ZFAS1), a novel lncRNA transcribed in the antisense orientation of zinc finger NFX1‐type containing 1(ZNFX1), was found to be increased in multiple cancers, such as gastric cancer and hepatocellular carcinoma, contributing to cancer development and progression. In the present review, we summarized recent progression on study of the functions and underlying molecular mechanisms of ZFAS1 related to occurrence and development of multiple cancers.
This content is subject to copyright. Terms and conditions apply.
Cell Proliferation. 2019;52 :e12 513 . wileyonlinelibrar y.com/journal/cpr 
|
 1 of 9
https://doi.org/10.1111/cpr.12513
1 | INTRODUCTION
Recently, due to increasing incidence and mortality, cancer is one
of the leading public health problems all over the world and con-
tinues to capture our great attention.1 In the light of estimates in
a comprehensive overview called Cancer Statistics, 2017, there
were 1 688 780 new patients predicted to be diagnosed with can-
cers and 600 920 patients were predicted to die of cancers in the
United States are predicted to occur in 2017.2 In China, there were
an estimated 4 292 000 new cancer patients and 2 814 000 pa-
tients who died of cancers have occurred in 2015.3 Despite mul-
tiple treatments modalities being accessible for cancer patients,
including surgery, chemotherapy, radiation therapy, and targeted
therapy, the 3- and 5- year cancer- specific survival rates still remain
po or.1,4 Exploration of and revealing the molecular mechanisms im-
portant to cancer is therefore critical in order to identify new diag-
nostic and prognostic biomarkers as well as effective treatments.
Less than 2% of genes are associated with the production of
specific proteins according to eukaryote whole- genome sequenc-
ing.5 Furthermore, more than 70% of identified genes had no
protein- coding function.5 At first, such noncoding RNAs (ncRNAs)
were considered to be transcriptional noise with no specific biologi-
cal function.6 With further research of ncRNAs, however, they have
been identified as indispensable regulators of a variety of biological
processes, including epigenetics, cell cycle, posttranscriptional reg-
ulation and chromatin modification.6-8 Although we currently lack
satisfactory classifications for these ncRNAs, long ncRNAs are ar-
bitrarily considered to be longer than 200 nucleotides.6 ,9 Mounting
evidence has indicated that lncRNA plays key roles in physiolog-
ical and pathological processes by modifying gene expression at
the transcriptional, posttranscriptional, epigenetic, and translation
levels.6-8 Several reports have demonstrated that lncRNAs could
function as potential oncogenes or tumour suppressor genes in
order to play vital regulatory roles in tumourigenesis and tumour
progression.10,11
Received:6February2018 
|
Accepted:13June2018
DOI : 10.1111 /cpr.12 513
REVIEW
ZFAS1: A novel vital oncogenic lncRNA in multiple human
cancers
Anbang He | Shiming He | Xuesong Li | Liqun Zhou
Anban g He and Shi ming He are the aut hors who contr ibuted equall y.
Depar tment of Urology, The Institute
of Urolog y, Peking Universit y First
Hospital, Peking University, National
Urologi cal Cancer Centre, Beijing 100034,
China
Correspondence: Liqun Zhou and Xueson g
Li, Department of Urolog y, The Institute of
Urolog y, Peking Uni versit y First H ospita l,
Peking University, National Urological
Cancer Centre, B eijing 100034, China
(zhoulqmail@sina.com; pineneedle@sina.
com).
Funding information
Nationa l Natural Science Fo undation of
China; Natural Science Foun dation of
Beijing, Grant/Award Number: 71772219
and 7152146; The Shenzhen Municipal
Government of China, Grant/Award
Number: ZDSYS201504301722174,
JCYJ2015033 0102720130and
GJHZ2015031615491 2494
Abstract
Long noncoding RNAs (lncRNAs) are a class of noncoding, endogenous, single-
stranded RNAs longer than 200 nucleotides in length that are transcribed by RNA
polymerase II. Mounting evidence has indicated that lncRNAs play key roles in sev-
eral physiological and pathological processes by modifying gene expression at the
transcriptional, posttranscriptional, epigenetic, and translation levels. Many reports
have demonstrated that lncRNAs function as potential oncogene or tumour suppres-
sors and thus play vital regulatory roles in tumourigenesis and tumour progression.
ZNFX1 antisense RNA 1 (ZFAS1), a novel lncRNA transcribed in the antisense orien-
tation of zinc finger NFX1- type containing 1(ZNFX1), was found to be increased in
multiple cancers, such as gastric cancer and hepatocellular carcinoma, contributing
to cancer development and progression. In the present review, we summarized re-
cent progression on study of the functions and underlying molecular mechanisms of
ZFAS1 related to occurrence and development of multiple cancers.
This is an op en access article under the terms of the Creati ve Commons Attribution Li cense, wh ich perm its use, distribution an d reproduction in any mediu m,
provide d the original work is properly cited.
© 2018 The Aut hors Cell ProliferationPublishedbyJohnW iley&SonsLtd
2 of 9 
|
   HE Et al.
ZNFX1 antisense RNA 1 (ZFAS1), a novel lncRNA transcribed
from the antisense orientation of zinc finger NFX1- type cont ain-
ing 1(ZNFX1), is located on chromosome 20q13.13 (Figure 1).12
Recent research has reported that ZFAS1 may act an emerging
regulatory factor in multiple diseases, such as acute myocardial
infarction,13,14 rheumatoid arthritis,15 and cancer.16 Accumulating
evidence has suggested that the abnormal expression of ZFAS1
contributes to the occurrence and development of various can-
cers, including gastric cancer, hepatocellular carcinoma, colorectal
cancer, glioma, osteosarcoma, ovarian cancer, acute myeloid leu-
kaemia, nonsmall cell lung cancer (NSCLC), oesophageal squamous
cell carcinoma, and breast cancer. In this review, we summarized
recent progression regarding study of the functions and underly-
ing mechanisms of ZFAS1 in the occurrence and development of
various cancers.
2 | DISCOVERY OF ZFAS1
ZFAS1, firstly studied in breast cancer by Marjan, is transcribed
from the gene ZNFX1 on the antisense DNA strand near the 5′
end. It is located in both cellular cytoplasmic and nuclear fractions
with a leng th of 17 561 bp, including five exons with five transcrip-
tional variations (NR _003604.3:1008 bp; NR_003605.2:689 bp;
NR_0 03606.3 :860 b p; NR_036658 .2: 946 b p; NR_036659.2:504 bp).
ZFAS1 hosts three small nucleolar RNAs (snoRNAs), including
Snord12, Snord12b, and Snord12c.12 SnoRNAs, a class of RNA mol-
ecules with 60- 150 nt in length which may be involved in the chemi-
cal modifications of other RNAs, like transfer RNAs, ribosomal RNAs,
and small nuclear RNAs.17 Current research has indicated that upreg-
ulation of ZFAS1 is positively correlated with clinicopathologic al fea-
tures and prognosis, including TNM stage, lymph-node metastasis,
FIGURE1 ZFAS1 was located on chromosome 20q13.13 and transcribed in antisense orientation of ZNFX1 and also hosts three small
nucleolar RNAs, including Snord12, Snord12b, and Snord12c
TABLE1 Functional characterizations of ZFAS1 in multiple human cancers
Cancer t ypes Expression Role Functional role Related genes References
Colorectal cancer Upregulated Oncogenic Cell proliferation migration, invasive,
metastasis, cell cycle control, cell
antiapoptosis
miR- 484
miR- 590- 3p
p53 and EMT
signalling
pathway
21-24
Gastric cancer Upregulated Oncogenic Cell proliferation, cell antiapoptosis, cell
migration and metastasis, cell cycle control
EMT, KLF2, EZH2,
NKD2
25-27
Hepatocellular
carcinoma
Upregulated Oncogenic Cell invasion and tumour metastasis miR- 150, ZEB1,
MMP14, and
MMP16
28
Glioma Upregulated Oncogenic Cell proliferation, migration, and invasive Notch and EMT
signalling
pathway
30,31
Osteosarcoma Upregulated Oncogenic Cell proliferation, migration and invasion, cell
antiapoptosis, cell cycle control
miR- 200b/c,
miR- 486, BMI1,
ZEB2
32,33
Ovarian cancer Upregulated Oncogenic Cell proliferation migration, chemoresistance miR- 150 - 5p, Sp1 34,35
Acute myeloid
leukaemia
Upregulated Oncogenic Cell proliferation, cell antiapoptosis, cell cycle
control
36
Nonsmall cell lung
cancer
Upregulated Oncogenic 37
Oesophageal squamous
cell carcinoma
Upregulated Oncogenic 29
    
|
 3 of 9
HE Et al.
and overall survival. Several meta- analyses have indicated that
lncRNA ZFAS1 could be as a prognostic biomarker for patients with
various cancers.16,18-20 Upregulated ZFAS1 could function as an on-
cogene to promoting cell proliferation, migration, and invasion. The
relevant clinicopathological features and underlying molecular mech-
anisms of ZFAS1 in various cancers are summarized in Tables 1 and 2.
3 | ZFAS1 IN VARIOUS HUMAN CANCERS
Accumulating evidence has demonstrated the abnormal expression of
ZFAS1 in multiple cancers, including gastric cancer, hepatocellular car-
cinoma, colorectal cancer, glioma, osteosarcoma, ovarian cancer, acute
myeloid leukaemia, NSCLC, oesophageal squamous cell carcinoma, and
breast cancer. Specifically, the expression of ZFAS1 was found to be
upregulated in most cancers, except breast cancer. Upregulation of
ZFAS1 expression was positively associated with advanced TNM stage
and lymph-node metastasis in colorectal cancer, gastric cancer, ovar-
ian cancer, and NSCLC. Additionally, increased expression of ZFAS1
was closely correlated with poorer overall survival in colorec tal cancer,
gastric cancer, glioma, osteosarcoma, oesophageal squamous cell car-
cinoma, ovarian cancer, and NSCLC. The association between ZFAS1
expression and the relevant clinicopathological features of various can-
cers are summarized in Table 1. Overexpression of ZFAS1 was shown
to promote cell proliferation and induced cellular apoptosis in various
cancers, including colorectal cancer, gastric cancer, glioma, osteosar-
coma, acute myeloid leukaemia, and ovarian cancer. Interestingly,
knockdown of ZFAS1 inhibited cell migration and invasion in colorectal
cancer, gastric cancer, glioma, hepatocellular carcinoma, osteosarcoma.
The functions and underlying molecular mechanisms of ZFAS1 in vari-
ous cancers are summarized in Table 2 and detailed in the rest of this
review.
3.1 | Colorectal cancer
ZFAS1 has been found in several studies to be overexpressed in
colorectal cancer tissues and cell lines compared with paired adja-
cent normal colorectal tissues and a human colonic epithelial cell
line, HCoEpiC. Fang et al21 demonstrated that upregulated ZFAS1
expression was positively associated with an advanced TNM stage,
vascular invasion, and lymph-node metastasis. Overexpression of
ZFAS1 promoted cell proliferation, invasion, and induced cell ap-
optosis in colorectal cancer (CRC). Furthermore, silencing of ZFAS1
could reduce Zinc Finger E- Box Binding Homeobox 1 (ZEB1) expres-
sion and increased the expression of epithelial markers, including
E- cadherin and ZO- 1, meanwhile decreasing the expression of mes-
enchymal markers, including vimentin and N- cadherin. Taking into
account these results, the researchers concluded that ZFAS1 could
function as an oncogene by promoting ZEB1 induction of epithelial-
to- mesenchymal transition (EMT).
Wang et al22 confirmed that the expression of ZFAS1 was higher
in CRC tissues than in adjacent normal colorectal tissues. Moreover,
compared with primary CRC tumours, ZFAS1 was upregulated in
metast atic tumours, suggesting ZFAS1 may have a specific role in
such cancer metastasis. Furthermore, ZFAS1 expression in CRC
was positively correlated with lymphatic invasion and TNM stage.
Increased ZFAS1 levels were determined to predict poor overall sur-
vival and short relapse- free sur vival and cox multivariate analyses
revealed that ZFAS1 expression is an independent prognostic factor
in CRC. Silencing ZFAS1 expression could suppress cell migration,
invasive, and metast asis ability of CRC cell in vitro and in vivo. Xie
et al23 also confirmed that elevated ZFAS1 expression was signifi-
cantly associated with advanced TNM stage, lymph-nodes met as-
tasis, and poor overall survival. Functional experiment s indicated
that downregulated ZFAS1 inhibited cell proliferation and invasion
Cancer t ypes Clinicopathological features References
Colorectal cancer Positive helicobacter pylori, advanced TNM
stage, positive lymph-node metastasis, poorer
overall survival, higher recurrence rate, positive
lymphatic invasion, and positive microvascular
invasion
21-24
Gastric cancer Advanced TNM stage, higher T s tage, positive
lymph-node metastasis , larger tumour size
25-27
Hepatocellular Carcinoma Positive microvascular invasion, shorter overall
survival, and higher recurrence rate
28
Oesophageal squamous cell
carcinoma
Poorer histological grade, shorter overall
survival
29
Glioma Higher clinical stage, poorer overall survival 30,31
Osteosarcoma Poorer overall survival 32,33
Ovarian cancer Poorer overall survival, advanced tumour stage,
larger tumour size, positive ly mph-node
metastasis
34,35
Nonsmall cell lung cancer Advanced TNM stage, positive lymph-node
metastasis, poor differentiation, poorer overall
survival
37
TABLE2 Clinical features of ZFAS1 in
multiple human cancers
4 of 9 
|
   HE Et al.
both in vitro and in vivo. Bioinformatics analysis and experiments
also proved that ZFAS1 directly interacted with miR- 484. Thorenoor
et al24 showed that silencing ZFAS1 decreased cyclin- dependent ki-
nase 1 (CDK1) to inhibit cell proliferation through G1- arrest of cell
cycle by sponging miR- 590- 3p in CRC cells. Decreased ZFAS1 ex-
pression resulted in reduction of p53 and cyclin B1 levels and pro-
moting poly ADP- ribose polymerase (PARP) cleavage to induce cell
apoptosis. These findings demonstrated that ZFAS1 could act an on-
cogenic lncRNA which may serve as a novel independent prognostic
biomarker for patient s with CRC.
3.2 | Gastric cancer
Current research trends have explored the function and molecular
mechanisms of ZFAS1in gastric cancer. Zhou et al, 25 Pan et al,26 and
Nie et al27 all demonstrated the elevated expression of ZFAS1 in
gastric cancer tissues and cell lines, compared to adjacent nontu-
mour tissues and normal gastric cell line (GES- 1). Increased expres-
sion of ZFAS1 in gastric cancer was also closely interrelated with
TNM stage, lymph- node metastasis, and tumour size. Kaplan- Meier
analysis revealed that GC patient s with higher ZFAS1 expression
levels had poorer overall survival (OS) and progression- free survival
(PFS) than those with lower ZFAS1 expression levels. Knockdown
of ZFAS1 suppressed cancer cells proliferation and promoted can-
cer cells apoptosis in vitro, and restrained tumourigenesis of gastric
cancer cells in vivo. Further research showed that ZFAS1 could si-
multaneously interact with the enhancer of zeste homolog 2 (EZH2)
and lysine demethylase 1A/histone demethyltransferase of REST
complex (L SD1/CoREST) to inhibit Kruppel Like Factor 2 (KLF2)
and Naked Cuticle Homolog 2 (NKD2) transcription. Moreover,
rescue experiments suggested that ZFAS1 par tially relied on sup-
pression of KLF2 and NKD2 to exert its oncogenic effect s. Zhou
et al25 found that circulating ZFAS1 was also overexpressed in GC
patients and that surgery operation could decrease subsequent ex-
pression in plasma. Finally, to elucidate the potential diagnostic value
of plasma ZFAS1, receiver operating characteristic (ROC) curve and
the Youden index were calculated. The area under the ROC cur ve
(AUC) was 0.727 (95% CI: 0.6 42- 0.813, P < 0.001), with a sensitivity
of 0.766 and specificity of 0.639. All of these results indicated that
circulating ZFAS1 had a relatively moderate accuracy when distin-
guishing gastric cancer patients from other individuals. Pan et al26
also found that silencing of ZFAS1 repressed cell proliferation and
migration by inhibiting cell cycle and EMT. Finally, ZFAS1 was ob-
served in exosomes and was transported by exosomes to promote
cell proliferation and migration of GC.
3.3 | Hepatocellular carcinoma
Li et al28 reported that ZFAS1 expression was signific antly amplified
in HCC tissues and cell lines compared with the paired nontumour
tissues and normal liver cells. Correlation analysis demonstrated that
high ZFAS1 levels were positively associated with microvascular in-
vasion and recurrence. Kaplan- Meier analysis suggested that HCC
patients with upregulated ZFAS1 experienced poorer overall sur-
vival (OS) and progression- free survival (PFS). Fur ther study showed
that amplified ZFAS1 could enhance HCC cell invasion and tumour
metast asis in vitro and in vivo. Subsequent bioinformatics analysis
and experimentation revealed that ZFAS1 directly interacted with
miR- 150. Moreover, miR- 150 could suppress HCC cell invasion and
metast asis by targeting ZEB1, matrix metallopeptidase 14 (MMP14),
and matrix metallopeptidase 16 (MMP16). From these results, the
authors concluded that ZFAS1 was acting in an oncogenic role by
binding miR- 150 and abrogating its tumour- suppressive function in
HCC progression.
3.4 | Oesophageal squamous cell carcinoma
Shi et al29 demonstrated that ZFAS1 expression was elevated in
oesophageal squamous cell carcinoma (ESCC) and high ZFAS1 was
positively correlated with histological grade. ESCC patient s with
high ZFAS1 expression had poorer OS than those with low ZFAS1
expression both in the primary cohort and validation cohor t. In
both cohorts, both univariate and multivariate analysis confirmed
that ZFAS1 expression, histological grade, and T stage as prognos-
tic factors. A nomogram then used clinicopathological factors and
ZFAS1 expression to predict the prognosis of lymph-node- negative
ESCC patients without preoperative chemoradiotherapy. The result-
ing decision- making curve suggested that the proposed nomogram
wasbetterthan8thAJCC-TNMstaging systemintermsof survival
prediction.
3.5 | Glioma
ZFAS1 expression was markedly overexpressed in glioma tissues and
cell lines in reports by Gao et al30 and Lv et al31 with high ZFAS1
expression in glioma tissues being closely associated with advanced
clinical stage and poor overall survival. Both univariate and multi-
variable Cox regression analysis revealed that upregulated ZFAS1
and the clinical stage were independent prognostic factors in the
overall sur vival of glioma patients. Silencing of ZFAS1 could sup-
press cell proliferation by arresting the cell cycle and inducing cell
apoptosis in glioma. Fur thermore, knockdown ZFAS1 inhibited cell
migration and invasion in glioma. Several EMT markers play indis-
pensable roles in epithelial- mesenchymal transition signalling path-
way, including MMP2, MMP9, E- cadherin, N- cadherin, Integrin β1,
ZEB1, Twist, and Snail. After silencing ZFAS1, the expression levels
of MMP2, MMP9, N- cadherin, Integrin β1, ZEB1, Twist, and Snail
were markedly downregulated, while E- cadherin expression was sig-
nificantly upregulated. Furthermore, the expression of Notch signal-
related proteins Hes family BHLH transcription fac tor 1 (Hes- 1) and
NICD were decreased by silencing ZFAS1. Notch signalling pathway
is believed to function as a vital regulator of embryo development
via the regulation of intercellular signal communication by modifying
cell proliferation and apoptosis. Therefore, these data indicated that
ZFAS1 may enhance glioma progression by activating the EMT and
Notch signalling pathways.
    
|
 5 of 9
HE Et al.
3.6 | Osteosarcoma
In osteosarcoma tissues and cell lines, Liu et al and Li et al32,33
showed that ZFAS1 was markedly upregulated. Kaplan- Meier analy-
sis suggested that osteosarcoma patients with high ZFAS1 levels had
shorter overall sur vival (OS) than those with low ZFAS1. Functional
experiments revealed that upregulated ZFAS1 promoted osteosar-
coma cell proliferation, migration, and invasion in vitro and in vivo.
Mechanistically, the researchers discovered that ZFAS1 regulated
malignant phenotypes by competitively binding the miR- 200b/c to
increase B Lymphoma Mo- MLV Insertion Region 1 Homolog (BMI1)
expression. Further RNA pull- down assays proved that ZFAS1 inter-
acted with ZEB2 to keep ZEB2 protein stability. Moreover, they found
that specificity protein 1 (SP1) acted as an upstream ac tivated factor
of ZFAS1. Li et al discovered that ZFAS1 sponges miR- 4 86 to promote
osteosarcoma cells progression and metastasis in vitro and vivo.
3.7 | Ovarian cancer
A report by Xia et al34 showed that ZFAS1 was upregulated in
epithelial ovarian cancer (EOC) tissues and cell lines. Amplified
expression ZFAS1 in EOC tissues was positively correlated with
advanced clinical stage, larger tumour size, and lymph- node metas-
tasis. Kaplan- Meier analysis suggested that EOC patients with high
ZFAS1 expression had poorer overall sur vival (OS) than those with
low ZFAS1. Cell experiments revealed that knockdown of ZFAS1
suppressed cell proliferation, migration, and chemoresistance in
EOC. Further research identified downregulated miR- 150- 5p as a
potential t arget of ZFAS1 in EOC tissue. Further luciferase assays
indicated miR- 150- 5p subsequently suppressed transcription fac-
tor SP1 expression. Subsequent rescue experiments revealed than
silencing miR- 150- 5p could partially rescue the suppressed prolif-
eration and migration induced by inhibition of ZFAS1 in EOC cells.
These dat a suggested that ZFAS1/miR- 150- 5p/SP1 axis may play a
critical role in enhancing cell proliferation, migration, and chemore-
sistance in EOC. Additionally, Liu et al35 identified eight lncRNA sig-
natures, which were remarkably correlated with chemosensitivity in
a multivariate logistic regression model and could precisely predict
the chemosensitivity of patients, by analysing the lncRNA expres-
sion profiles of 258 HGS- OvCa patients from The Cancer Genome
Atlas(TCGA). Similarly, data from the Gene Expression Omnibus
dataset s (GEO) confirmed the strong relation between ZFAS1 and
chemosensitivity. In vitro experiments suggested that the ZFAS1
expression was overexpressed with cisplatin treatment in several
EOC cell lines (A2008, HeyA8, and HeyC2). Taken together, these
findings suggested that ZFAS1 may play a vital role in platinum
resistance, which should be further explored.
3.8 | Acute myeloid leukaemia
When compared with normal cell lines, Guo et al30 revealed that the
expression of ZFAS1 was increased in all four human acute myeloid
leukaemia (AML) cell lines. The silencing of ZFAS1 suppressed cell
proliferation in HL-60 and SKNO-1 cell lines according to CCK-8
assay and induced AML cell cycle G1 phase arrest and promoted
cell apoptosis by flow cy tometr y. These findings demonstrated that
upregulated ZFAS1 enhanced cell proliferation and suppressed cell
apoptosis in AML.
3.9 | Nonsmall cell lung cancer
Tian et al36 demonstrated that the expression of ZFAS1 was in-
creased in nonsmall cell lung cancer (NSCLC) compared with adja-
cent noncancerous tissues. A gain, increased ZFAS1 was positively
correlated with advanced TMN stage, lymph-node metastasis, and
poor differentiation. Kaplan- Meier analysis indicated that NSCLC
patients with upregulated ZFAS1 had poorer OS than those with
downregulated ZFAS1. Both univariate and multivariable Cox re-
gression analysis suggested that high ZFAS1 expression TMN stage,
lymph-node metastasis, and differentiation level were independent
prognostic factors for overall survival of NSCLC patients.
3.10 | Breast cancer
Marjan et al12 identified that ZFAS1 as highly expressed in stage
of lactation in mouse mammary gland tissues and was reduced in
human invasive ductal breast carcinoma compared with normal
breast tissues. On the contrary, Hansji et al37 showed that ZFAS1
expression was not significantly different in breast cancer tissues
from normal tissues according to RNAseq data set (HiSeqV2- 2015-
02- 24) from TCGA. However, ZFAS1 expression was significantly
downregulated in basal and HER2 breast cancer subtypes compared
to normal breast tissue. Impor tantly, ER+ breast tumours had higher
expressionofZFAS1thanER−(negative)breasttumours.Thesetwo
studies had different results largely due to their limited number of
samples and testing of different breast cancer subtypes. Next, there
was no significant found between the expression level of ZFAS1 and
ZNFX1. They also found that ZFAS1 was localized to both the cy-
toplasm and the nucleus while ZNFX1 was enriched in the nucleus.
Further research has indicated that ZFAS1 is associated with only
part of the ribosomes, being enriched in 18S rRNA fractions com-
pared to those containing 28S rRNA. The expression of ZFAS1 is
associated with the expression of genes which were involved in ri-
bosome biogenesis. A series of experiments have also demonstrated
that ZFAS1 is induced upon ribosome biogenesis, revealing a role in
the synthesis or assembly of ribosomes.
4 | UNDERLYING MOLECULAR
MECHANISMS OF ZFAS1
4.1 | The molecular level
4.1.1| Function of ZFAS1 as a ceRNA
Competing endogenous RNAs (ceRNAs), a class of noncoding RNA,
have been shown to modify the mRNA expression of genes by
6 of 9 
|
   HE Et al.
acting as miRNA sponges competing for mutual microRNAs, which
simultaneously target both noncoding RNA and genes.38, 39 Several
articles have demonstrated that ZFAS1 could function as a ceRNA
to regulate the expression of specific genes by competing for spe-
cific microRNAs, exerting its oncogenic function in various cancers.
Li et al and Xia et al showed that ZFAS1 directly interacted with
FIGURE2 Underlying molecular mechanisms of ZFAS1 in multiple human cancers. A, ZFAS1 functioned as a ceRNA to directly interac ted
with miR- 150 to increase the expression of ZEB1, MMP14, MMP16, and SP1. B, silencing ZFAS1 could decrease the expression of CDK1
by competing for miR- 590 - 3p. C, ZFAS1 may act as a ceRNA to upregulate the expression of BMI1 by competitively binding miR- 20 0b/c. D,
After Silencing ZFAS1, the expression levels of mesenchymal markers, including MMP2, MMP9, N- cadherin, Integrin β1, ZEB1, Twist, and
Snail, were markedly downregulated, while the expression of epithelial markers including E- cadherin and ZO1 significantly upregulated. E,
ZFAS1 could regulate the expression of Hes- 1 and NICD to activate Notch signalling pathway. F, decreased ZFAS1 contributed to cell-
cycle arrest and inducement of apoptosis by reducing the expression of p53 and cyclin B1 and promoting PARP cleavage. G, ZFAS1 could
simultaneously interact with EZH2 and LSD1/CoREST to inhibit KLF2 and NKD2 transcription. H, SP1 could activate the expression of
ZFAS1
    
|
 7 of 9
HE Et al.
miR- 150 to increase the expression of ZEB1, MMP14, MMP16, and
transcription factor SP1 (Figure 2A). Thorenoor et al then showed
that silencing of ZFAS1 could decrease the expression of CDK1 in
order to inhibit cell proliferation via G1 arrest of the cell cycle by
competing for miR- 590- 3p in CRC cells (Figure 2B). Liu et al found
that ZFAS1 promoted malignant phenotypes by competitively bind-
ing miR- 200b/c to upregulate the expression of BMI1 (Figure 2C).
The ring finger protein encoded by BMI1 is major component of
PRC1, which acts as an indispensable epigenetic repressor of a va-
riety of regulatory genes involved in self- renewal and embryonic
development by remodelling the chromatin of stem cells. Finally,
Xie et al proved that ZFAS1 could directly interact with miR- 484.
Previous study identified miR- 484 as an inhibitor of cell prolifera-
tion and invasion by targeting ZEB1 and SMAD2 in cervical cancer
cells, revealing that ZFAS1 may promote cell proliferation and inva-
sion via sponging miR- 484 to modulate the expression of ZEB1 and
SMAD2.
4.1.2 | Transcriptional regulation
Further research has demonstrated that ZFAS1 could simultaneously
interact with EZH2 and LSD1/CoREST to inhibit KLF2 and NKD2
transcription. Rescue experiments revealed that ZFAS1 was partly re-
lied on suppressing KLF2 and NKD2 expression to exert its oncogenic
effects (Figure 2G). SP1 was also demonstrated to be an upstream
factor for the activation of ZFAS1 expression (Figure 2H). Moreover,
Hansji et al showed that ZFAS1 was induced upon ribosome biogen-
esis, revealing a role in synthesis or assembly of ribosomes.
4.2 | ZFAS1 is involved in several signalling
pathways
4.2.1 | EMT signalling pathway
EMT plays an essential role in both physiological and pathological
processes, such as embryonic development as well as the occurrence
and development of tumours.40 ,41 Several EMT markers are indis-
pensable in the EMT signalling pathway, including MMP2, MMP9,
E- cadherin, N- cadherin, Integrin β1, ZEB1/2, Twist, and Snail.41
After silencing of ZFAS1, the expression levels of MMP2, MMP9, N-
cadherin, Integrin β1, ZEB1, Twist, and Snail were all markedly down-
regulated, while E- cadherin expression significantly upregulated. In
conclusion, Liu et al also showed that ZFAS1 interacted with ZEB2 to
protect ZEB2 protein stability in the activation of the EMT signalling
pathw ay.
4.2.2 | Notch signalling pathway
The Notch signalling pat hway functions as a vital regulator in embr yo
development via the regulation of intercellular signal communication
by modifying cell proliferation and apoptosis42-4 4; both Hes- 1 and
NICD have vital functions in Notch signalling pathway.44,45 Gao et al
found that the expression Notch signal- related proteins Hes- 1 and
NICD were decreased by silencing ZFAS1in glioma cell, suggesting
ZFAS1 could regulate the expression of Hes- 1 and NICD to activate
Notch signalling pathway (Figure 2E).
4.2.3 | p53 signalling pathway
The p53 protein, a nuclear transcription factor, modified the expres-
sion of multiple genes which are involved in a variet y of biological
processes, including the cell c ycle, apoptosis, and DNA repair.46
Thorenoor et al demonstrated that decreased ZFAS1 contributed to
cell- cycle arrest and induction of apoptosis by reducing the expres-
sion of p53 and cyclin B1 and promoting PARP cleavage (Figure 2F).
This finding suggests that ZFAS1 activated the p53 signalling path-
way to act oncogenic roles in various cancers.
5 | CONCLUSION AND FUTURE
PERSPECTIVES
With the rapid development of next- generation sequencing tech-
nology, a large amount of evidence has identified dysregulated
lncRNAs as potential oncogenes or tumour suppressor genes that
play crucial regulatory roles in tumourigenesis and tumour progres-
sion.47, 48 ZFAS1, a newly identified lncRNA, was found to be widely
upregulated in various human cancers (expect breast cancer).
Amplified ZFAS1 was dramatically correlated with multiple clinico-
pathological features and prognosis, such as TNM stage, lymph-
node metastasis, and overall survival. In vitro, knockdown of ZFAS1
repressed cell proliferation, invasion, and promoted cell apoptosis
in multiple cancers, suggesting ZFAS1 contributed to tumourigen-
esis and tumour progression. In vivo experiments also confirmed
that silencing ZFAS1 could obviously restrain tumourigenesis of
cancer cells. The underlying molecular mechanisms of ZFAS1 in-
volved in multiple cancers have been explored preliminarily. At the
molecular level, ZFAS1 could function as a ceRNA in the regulation
of specific gene expression by competing for specific microRNAs,
representing its oncogenic function in various cancers. ZFAS1 also
could interact with some proteins to regulate gene expression at
the transcriptional level. Fur thermore, ZFAS1 was demonstrated to
take part in several signalling pathways contributing to c arcinogen-
esis and cancer progression, including EMT, Notch, and p53 signal-
ling pathways.
Although some of the underlying mechanisms of ZFAS1 in-
volved in dif ferent cancers have been identified, there are many
problems that still need to be addressed. First, the molecular
mechanism of ZFAS1 in each cancer type should be clarified. Many
lncRNAs function at the epigenetic, transcriptional, and post-
translational processing level in their contribution to carcinogene-
sis and cancer progression. Second, finding a diagnostic biomarker
or therapeutic target is a promising direction for cancer diagnosis
and treatment. Recently, several lncRNAs have been identified not
only in neoplastic tissues but also in body fluids, such as plasma
and urine, suggesting a potential role as cancer- specific molecular
8 of 9 
|
   HE Et al.
biomarkers in cancer diagnosis, prognosis, and treatment. Other
advantages in cancer diagnosis and treatment, especially in the
early diagnosis and prediction of prognosis, include simple and
noninvasive detection as well as a moderate treatment price.
Therefore, the clinical value of ZFAS1 in the diagnosis and treat-
ment of cancers requires more attention.
In summar y, ZFAS1 has been shown to have oncogenic function
in tumourigenesis and tumour progression and may act as a poten-
tial cancer- specific molecular biomarker in the general diagnosis,
prognosis, and treatment of cancer. Until this point, research on the
mechanism of ZFAS1 has made some progression, but remains in the
early st ages. Future works will need to emphasize exploration of the
precise molecular regulatory mechanisms of ZFAS1 in carcinogen-
esis and cancer progression, to facilitate the clinical applic ation of
ZFAS1 as early as possible.
ACKNOWLEDGEMENTS
This work was supported by National Natural Science Foundation of
China 81672546, 81602253, 81372746, Natural Science Foundation
of Beijing 71772219, 7152146, The Sh enzhen Municipa l Government
of China (ZDSYS201504301722174, JCYJ20150330102720130,
GJHZ20150316154912494).
AUTHORS’ CONTRIBUTIONS
AH, SH wrote the manuscript. XL and LZ provided the financial sup-
port and reviewed the manuscript. All authors read and approved
the final manuscript.
CONFLICT OF INTEREST
All authors declare no conflict of interest.
REFERENCES
1. Miller KD, Siegel RL, Lin CC , et al. Cancer treatment and survivor-
ship statistic s, 2016. CA Cancer J Clin. 2016;66(4):271-289.
2. SiegelRL,MillerKD,JemalA.CancerStatistics,2017.CA Cancer J
Clin. 2017;67(1):7-3 0.
3. Chen W, Zheng R, Baade PD, et al. C ancer st atistics in China, 2015.
CA Cancer J Clin. 2016;66(2):115-132.
4. Nakashima L . Evolution of cancer treatment and evolving chal-
lenges. Healthc Manage Forum. 2018;31(1):26-28.
5. ENCODE Projec t Consortium. An integrated encyclopedia of DNA
elements in the human genome. Nature. 20 12 ;4 89 (7414):57-74.
6. MercerTR,DingerME ,MattickJS.Longnon-codingRNAs:insights
into func tions. Nat Rev Genet. 2009;10(3):155-159.
7. Pandey RR, Kanduri C. Transcriptional and posttr anscriptional
programming by long noncoding RNAs. Prog Mol Subcell Biol.
20 11; 51 :1-27.
8. Ponting CP, Oliver PL, Reik W. Evolution and functions of long non-
coding RNAs. Cell. 20 09;136(4):629-641.
9. Lujambio A , Lowe SW. The microcosmos of cancer. Nature.
2012;482( 7385):347-355.
10. Gut schner T, Diederichs S . The hallmarks of cancer: a long non-
coding RNA point of view. RNA Biol. 2012;9(6):703-719.
11. Isin M, Dalay N. LncRNAs and neoplasia. Clin Chim Acta.
2015;444:280-288.
12. Askarian-Amiri ME, Crawford J, French JD, et al. SNORD-host
RNA Zfas1 is a regulator of mammary development and a potential
marker for breast cancer. RNA. 2011;17(5):878-891.
13. Wu T, Wu D, Wu Q, et al. Knockdown of Long Non- Coding RNA-
ZFAS1 Protect s Cardiomyocytes Against Acute Myocardial
Infarction Via Anti- Apoptosis by Regulating miR- 150/CRP. J Cell
Biochem. 2017;118(10):3281-3289.
14. Zhang Y, Sun L, Xuan L, et al. Reciprocal changes of circulating long
non- coding RNAs ZFAS1 and CDR1AS predict acute myocardial in-
farction. Sci Rep. 2016;6:22384.
15. Ye Y, Gao X, Yang N. LncRNA ZFAS1 promotes cell migration and
invasion of fibroblast- like synoviocy tes by suppression of miR- 27a
in rheumatoid arthritis. Hum Cell. 2018;31(1):14-21.
16. Dong D, Mu Z, Wang W, et al. Prognostic value of long noncod-
ing RNA ZFAS1 in var ious carcinomas: a meta- analysis. Oncotarget.
2017;8(48):84497-84505.
17. Romano G , Veneziano D, Acunzo M, Croce CM. Small non- coding
RNA and cancer. Carcinogenesis. 2017;38(5): 485-491.
18. L an T, Lan X, Li G, Zheng Z, Zhang M, Qin F. Prognostic role of long
noncoding RNA ZFAS1 in cancer patients: a systematic review and
meta- analysis. Oncotarget. 2017;8(59):10 0490-100 498.
19. Song W,TianC,Zhang RJ,Zou SB,WangK .Meta-analysisofthe
prognos tic value of lncRNA ZFAS1 in patients with solid tumors.
Oncotarget. 2017;8(52):90301-90307.
20. Liu F, Gao H, Li S , Ni X, Zhu Z . Long non- coding RNA ZFAS1 cor-
relates with clinic al progression and prognosis in cancer patients.
Oncotarget. 2017; 8( 37):61 561-61569.
21. Fan g C, Zan J, Yue B , Liu C, He C , Yan D.L ong non-cod ing ribo-
nucleic acid zinc finger antisense 1 promotes the progression of
colonic c ancer by modulating ZEB1 expression. J Gastroenterol
Hepatol. 2017;32(6):1204-1211.
22. Wang W, Xing C. Upregulation of long noncoding RNA ZFAS1 pre-
dicts poor prognosis and prompts invasion and metast asis in col-
orect al cancer. Pathol Res Pract. 2016;212(8):690-695.
23. Xie S, Quanxing G , Wang X, Sun X, Kang Y. Long non- coding RNA
ZFAS1 sponges miR- 484 to promote cell proliferation and invasion
in colorectal cancer. Cell Cycle. 2017;17:1-21.
24. Thorenoor N, Faltejskova-Vychytilova P, Hombach S, et al. Long
non- coding RNA ZFAS1 inter acts with CDK1 and is involved in
p53- dep endent cell cycle control and apoptosis in colorectal can-
cer. Oncotarget. 2016;7(1):622-637.
25. Zhou H, Wang F, Chen H, et al. Increased expression of long-
noncoding RNA ZFAS1 is associated with epithelial- mesenchymal
transi tion of gastri c cancer. Aging ( Albany NY). 2016;8(9):2023-2038.
26. Pan L , Liang W, Fu M, et al. Exosomes- mediated transfer of long
noncoding RNA ZFAS1 promotes gastric cancer progression. J
Cancer Res Clin Oncol. 2017;143(6):991-1004.
27. Nie F, Yu X, Huang M, et al. Long noncoding RNA ZFAS1 promotes
gastric cancer cells proliferation by epigenetically repressing KLF2
and NKD2 expression. Oncotarget. 2017;8(24):38227-38238.
28 . Li T, Xie J, Shen C , et al. Amplification of long noncoding RNA
ZFAS1 promotes metastasis in hepatocellular carcinoma. Cancer
Res. 2015;75(15):3181-3191.
29. ShiH,LiuZ,PeiD,JiangY,ZhuH,ChenB.Developmentandvalida-
tion of nomogram based on lncRNA ZFAS1 for predicting survival
in lymph node- negative esophageal squamous cell carcinoma pa-
tients. Oncotarget. 2017;8(35):59048-59057.
30. Gao K, Ji Z, She K, Yang Q, Shao L.Long non-codingRNAZFAS1
is an unfavourable prognostic factor and promotes glioma cell
progression by activation of t he Notch signaling pathway. Biomed
Pharmacother. 2017;87:555-560 .
31. Lv QL, Chen SH , Zhang X, et al. Upregulation of long noncoding
RNA zinc finger antisense 1 enhances epithelial- mesenchymal
    
|
 9 of 9
HE Et al.
transition in vitro and predicts poor prognosis in glioma. Tumour
Biol. 2017;39(3):1010428317695022.
32. Liu G, Wang L, Han H , et al. LncRNA ZFAS1 promotes growth and
metastasis by regulating BMI1 and ZEB2 in osteosarcoma. Am J
Cancer Res. 2017;7( 7) :1450-1462 .
33. LiN,SunZH,FangM,XinJY,WanCY.Longnon-codingRNAZFAS1
sponges miR- 486 to promote osteosarcoma cells progression and
metas tasis in vi tro and vivo . Oncotarget. 2017; 8(61 ):10 416 0-10 4170 .
34. Xia B, Hou Y, Chen H, et al. Long non- coding RNA ZFAS1 interacts
with miR- 150- 5p to regulate Sp1 expression and ovarian cancer cell
malignancy. Oncotarget. 2017;8(12):19534-1954 6.
35. Liu R, Zeng Y, Zhou CF, et al. Long noncoding RNA expression sig-
nature to predict platinum- based chemotherapeutic sensitivity of
ovarian cancer patients. Sci Rep. 2017;7(1) :18.
36. Tian FM, Meng FQ, Wang XB. Overexpression of long- noncoding
RNA ZFAS1 decreases sur vival in human NSCLC patients. Eur Rev
Med Pharmacol Sci. 2016;20(24):5126-5131.
37. Hansji H, Leung E Y, Baguley BC , et al. ZFAS1: a long noncoding
RNA associated with ribosomes in breast cancer cells. Biol Direct.
2016;11(1):62.
38. TayY,Rinn J, Pandolfi PP.The multilayeredcomplexity of ceRNA
crosstalk and competition. Nature. 2014;505(7483):344-352.
39. Karreth FA, Pandolf i PP. ceRNA cross- talk in c ancer: when ce- bling
rivalries go awry. Cancer Discov. 2013;3(10):1113-1121.
40. Pang A, Carbini M, Moreira AL, Maki RG. Carcinosarcomas and re-
lated cancers: tumors caught in the act of epithelial- mesenchy mal
transition. J Clin Oncol. 2018;36(2):210-216.
41. Shibue T, Weinberg R A. EMT, CSCs, and drug resistance: the
mechanistic link and clinical implications. Nat Rev Clin Oncol.
2017;14(10):611-629.
42. Collu GM, Hidalgo-Sastre A, Brennan K. Wnt- Notch signal-
ling crosstalk in development and disease. Cell Mol Life Sci.
2014;71(18):3553-3567.
43. Schwanbeck R. The role of epigenetic mechanisms in Notch signal-
ing during development. J Cell Physiol. 2015;230 (5):969-981.
4 4. Brzozowa-Zasada M, Piecuch A , Michalski M, et al. Notch
and its oncogenic activit y in human malignancies. Eur Surg.
2017;49(5):199-209.
45. Liu ZH, Dai XM, Du B. Hes1: a key role in stemness, metastasis and
multidrug resistance. Cancer Biol Ther. 2015;16(3):353-359.
46. HarrisSL,LevineAJ.Thep53pathway:positiveandnegativefeed-
back loops. Oncogene. 2005;24(17):2899-2908.
47. Marchese FP, Raimondi I, Huarte M. The multidimensional mecha-
nisms of lon g noncoding R NA functio n. Genome Biol. 2017;18(1):206.
4 8. Tang Y, Cheung BB, Atmadibrata B, et al. The regulator y role of long
noncoding RNAs in cancer. Cancer Lett. 2017;391:12-19.
How to cite this article: He A, He S, Li X, Zhou L . ZFAS1: A
novel vital oncogenic lncRNA in multiple human cancers. Cell
Prolif. 2019;52:e12513 . https://doi.o rg /10.1111/cpr.1 2513
... In another study, ZFAS1 directly controlled miR-150-5p, which in turn increased ovarian cancer growth via modulating Sp1, and findings revealed that ZFAS1 was necessary for EOC cell chemoresistance, but miR-150-5p rendered EOC cells more vulnerable to Cisplatin and Paclitaxel [26]. ZFAS1 may also play a role in other chemoresistance in EOC cells [74]. Moreover, in lung adenocarcinoma, ZFAS1 induces proliferation, migration, and chemoresistance [74]. ...
... ZFAS1 may also play a role in other chemoresistance in EOC cells [74]. Moreover, in lung adenocarcinoma, ZFAS1 induces proliferation, migration, and chemoresistance [74]. So, ZFAS1 can act as a diagnostic or therapeutic marker for cisplatin or other drug resistance in these cancers. ...
Article
Full-text available
Zinc finger antisense 1 (ZFAS1), a newly discovered long noncoding RNA, is expressed in various tissues and organs and has been introduced an oncogenic gene in human malignancies. In various cancers, ZFAS1 regulates apoptosis, cell proliferation, the cell cycle, migration, translation, rRNA processing, and spliceosomal snRNP assembly; targets signaling cascades; and interacts with transcription factors via binding to key proteins and miRNAs, with conflicting findings on its effect on these processes. ZFAS1 is elevated in different types of cancer, like colorectal, colon, osteosarcoma, and gastric cancer. Considering the ZFAS1 expression pattern, it also has the potential to be a diagnostic or prognostic marker in various cancers. The current review discusses the mode of action of ZFAS1 in various human cancers and its regulation function related to chemoresistance comprehensively, as well as the potential role of ZFAS1 as an effective and noninvasive cancer-specific biomarker in tumor diagnosis, prognosis, and treatment. We expected that the current review could fill the current scientific gaps in the ZFAS1-related cancer causative mechanisms and improve available biomarkers.
... The versatile roles of ZFAS1 in cancer biology could be attributed to its ability to interact with different microRNAs and modulate multiple downstream target mRNAs. Therefore, ZFAS1 is considered as a pivotal oncogenic factor in cancer development [18]. ...
... LncRNA ZFAS1 was found to be increased in HK-2 cell under HG condition, suggesting that the upregulation of ZFAS1 may be an implication in DN development. In the field of cancer research, it was reported that the elevated ZFAS1 expression level was positively linked with advanced clinical stage and dismal prognosis in different malignancies [18]. However, currently there are no reports about its role in DN. ...
Article
Full-text available
Diabetic nephropathy (DN) is a common clinical syndrome in diabetic patients. Functional characterization of non-coding (ncRNAs) involved in the progression of DN can provide insights into the diagnosis and therapeutic management of DN. Human kidney proximal tubular epithelial cells (HK-2) were challenged by high glucose (HG, 50 mM) as a cell model of DN. The expression level of long non-coding RNA (lncRNA) ZFAS1 was quantified by qRT-PCR. The proteins and cytokines related to fibrosis and scortosis in DN (NLRP3, GSDMD-N, IL-1β and Caspase 1, fibronectin, collagen I, collagen III, IL-1β, and IL-18) were examined by western blot or ELISA. RNA precipitation and luciferase reporter activity experiments were conducted to assess the molecular associations. ZFAS1 and SGK1 were highly induced in HK-2 cells challenged with HG, while miR-525-5p downregulated upon HG treatment. ZFAS1 knockdown attenuated HG-induced fibrosis and scortosis in HK-2 cells by reducing the levels of NLRP3, GSDMD-N, Caspase 1, fibronectin, collagen I/III, IL-1β, and IL-18. Mechanically, ZFAS1 knockdown protected HK-2 cells from HG-induced injury by upregulating miR-525-5p and repressing SGK1 expression. Overall, our results suggest that knocking down ZFAS1 may be formulated as a protective strategy in ameliorating DN progression through regulating miR-525-5p/SGK1 pathway. Targeting ZFAS1 could be further explored as a potential approach for the management of DN.
... ZFAS1 is a gene that is found in several cancers including hepatocellular carcinoma, colorectal cancer, gastric cancer, ovarian cancer, glioma, osteosarcoma, esophageal squamous cell carcinoma, acute myeloid leukemia, non-small cell lung cancer, breast cancer etc. There are abnormally expressed lncRNAs (Zhou et al. 2016) and up-regulation of ZFAS1 expression is positively connected with lymph node metastasis of colorectal cancer, gastric cancer, ovarian cancer and non-small cell lung cancer (He et al. 2019); ZFAS1 can regulate miR-588 and regulate the body's inflammatory response through the miR-588/ROCK1 axis (Geng et al. 2022) and miR-588 has been reported to be involved in the pathogenesis of lung cancer, human breast cancer, colorectal cancer and other tumors (Yu et al. 2017;Fattahi et al. 2019;Dahiri et al. 2022); therefore, ZFAS1 can regulate inflammatory responses and is abnormally expressed in many tumors, further exploring its mechanism of action will help discover the role of lncRNA in tumor occurrence and development. ...
Article
Full-text available
The intricate relationship between inflammation and cancer is a well-established paradigm in oncology. Inflammation is recognized as one of the key hallmarks of cancer, profoundly influences tumor progression, malignant transformation, and the efficacy of chemotherapy. The tumor microenvironment, comprising various factors and signaling mechanisms, plays a pivotal role in shaping the biological characteristics of tumors. This review delves into the components of the tumor microenvironment, including inflammasomes, cytokines, and non-coding RNA, elucidating the impact of inflammation on tumor microenvironment dynamics throughout tumorigenesis. Particularly, inflammation exerts its influence by inducing immune suppression, with acute inflammation paradoxically fostering anti-tumor immune responses. The interplay of inflammatory factors significantly regulate immune responses within the tumor microenvironment, involving inhibiting immune cell activity, allowing tumors to evade surveillance, and promoting their proliferation and metastasis. Moreover, inflammation contributes to oxidative stress, generating reactive oxygen species and reactive nitrogen species. These oxidative agents can inflict damage on proteins, lipids, and nucleic acids, inducing genetic mutations and neoplastic transformations. Non-coding RNA molecules, crucial players in the tumor microenvironment, significantly contribute to the regulation of tumors and inflammation through diverse pathways. A comprehensive understanding of the interplay between inflammation and the tumor microenvironment holds immense potential for advancing cancer research and developing novel treatment strategies. This review provides a theoretical foundation for combating tumors by exploring the nuanced connections among inflammation, tumor microenvironment, and cancer development.
... ZNFX1 has been shown to activate intracellular immune pathways [58]. ZNFX1 antisense RNA 1, a novel long non-coding RNA transcribed in the antisense orientation of ZNFX1, is upregulated in multiple cancers, including gastric cancer and hepatocellular carcinoma, contributing to cancer development and progression [59]. However, the role of ZNFX1 in tumorigenesis remains to be elucidated. ...
Article
Full-text available
Background Targeted protein degradation of neosubstrates plays a crucial role in hematological cancer treatment involving immunomodulatory imide drugs (IMiDs) therapy. Nevertheless, the persistence of inevitable drug resistance and hematological toxicities represents a significant obstacle to their clinical effectiveness. Methods Phenotypic profiling of a small molecule compounds library in multiple hematological cancer cell lines was conducted to screen for hit degraders. Molecular dynamic-based rational design and cell-based functional assays were conducted to develop more potent degraders. Multiple myeloma (MM) tumor xenograft models were employed to investigate the antitumor efficacy of the degraders as single or combined agents with standard of care agents. Unbiased proteomics was employed to identify multiple therapeutically relevant neosubstrates targeted by the degraders. MM patient-derived cell lines (PDCs) and a panel of solid cancer cell lines were utilized to investigate the effects of candidate degrader on different stage of MM cells and solid malignancies. Unbiased proteomics of IMiDs-resistant MM cells, cell-based functional assays and RT-PCR analysis of clinical MM specimens were utilized to explore the role of BRD9 associated with IMiDs resistance and MM progression. Results We identified a novel cereblon (CRBN)-dependent lead degrader with phthalazinone scaffold, MGD-4, which induced the degradation of Ikaros proteins. We further developed a novel potent candidate, MGD-28, significantly inhibited the growth of hematological cancer cells and induced the degradation of IKZF1/2/3 and CK1α with nanomolar potency via a Cullin-CRBN dependent pathway. Oral administration of MGD-4 and MGD-28 effectively inhibited MM tumor growth and exhibited significant synergistic effects with standard of care agents. MGD-28 exhibited preferentially profound cytotoxicity towards MM PDCs at different disease stages and broad antiproliferative activity in multiple solid malignancies. BRD9 modulated IMiDs resistance, and the expression of BRD9 was significant positively correlated with IKZF1/2/3 and CK1α in MM specimens at different stages. We also observed pronounced synergetic efficacy between the BRD9 inhibitor and MGD-28 for MM treatment. Conclusions Our findings present a strategy for the multi-targeted degradation of Ikaros proteins and CK1α against hematological cancers, which may be expanded to additional targets and indications. This strategy may enhance efficacy treatment against multiple hematological cancers and solid tumors.
... As a newly recognized lncRNA, ZFAS1 (ZNFX1 antisense RNA 1; located on chromosome 20q13.13) has been associated with varied modulatory roles in an array of diseases [127]. For instance, ZFAS1 underexpression abolishes pharmacological lipid peroxidation during pneumonic conditions [128]. ...
Article
Full-text available
Ferroptosis, a recently identified type of non-apoptotic cell death, triggers the elimination of cells in the presence of lipid peroxidation and in an iron-dependent manner. Indeed, ferroptosis-stimulating factors have the ability of suppressing antioxidant capacity, leading to the accumulation of reactive oxygen species (ROS) and the subsequent oxidative death of the cells. Ferroptosis is involved in the pathophysiological basis of different maladies, such as multiple cancers, among which female-oriented malignancies have attracted much attention in recent years. In this context, it has also been unveiled that non-coding RNA transcripts, including microRNAs, long non-coding RNAs, and circular RNAs have regulatory interconnections with the ferroptotic flux, which controls the pathogenic development of diseases. Furthermore, the potential of employing these RNA transcripts as therapeutic targets during the onset of female-specific neoplasms to modulate ferroptosis has become a research hotspot; however, the molecular mechanisms and functional alterations of ferroptosis still require further investigation. The current review comprehensively highlights ferroptosis and its association with non-coding RNAs with a focus on how this crosstalk affects the pathogenesis of female-oriented malignancies, from breast cancer to ovarian, cervical, and endometrial neoplasms, suggesting novel therapeutic targets to decelerate and even block the expansion and development of these tumors.
... Dysregulated lncRNA expression which modulates Wnt/βcatenin modulators could have great potential for therapeutic intervention. We first selected 2 novels, Wnt/beta-catenin-related lncRNAs in digestive cancer by literature search and lncRNA disease database (Yuan et al. 2021;Zhuo et al. 2022;Hu et al. 2018;He et al. 2019;Xu et al. 2022). We analyzed the expression of the selected 2 lncNAs using available GEO datasets from pancreatic cancer tissues to define whether the expression of selected lncRNAs alters in pancreatic cancer (Fig. 1). ...
Article
Full-text available
Background The oncogenic Wnt/β-catenin signaling plays a critical role in carcinogenesis, prognosis, and resistance to therapy. Pancreatic cancer (PC) has high mortality because of its poor prognosis. Several studies have suggested that lncRNAs are directly involved in the development and progression of PC as well as in Wnt/β-catenin signaling. In this study, we investigated and compared the expression of Wnt/β-catenin signaling-related ZFAS1 and HCG11 lncRNAs, and their targets, CTNNB1 and IGF2BP1 genes in the blood of patients with PC and healthy individuals. A total of 47 PC patients and 50 healthy individuals participated in this study. RNA was extracted from the peripheral blood samples of participants, and cDNA was synthesized. The expression level of the selected genes was quantified by real-time PCR. The expression of HCG11 lncRNA and CTNNB1 genes in patients with PC was significantly upregulated compared to healthy individuals, and the expression of the ZFAS1 lncRNA was significantly downregulated. According to the analysis of the ROC curve, the diagnostic powers of ZFAS1 and CTNNB1 in PC were 0.67 and 0.69, respectively. Altogether, the present study suggests a role for ZFAS1 and HCG11 lncRNAs and CTNNB1 and IGF2BP1 in the pathogenesis of pancreatic cancer. Moreover, the peripheral expression of these lncRNAs may be useful as potential biomarkers for PC.
... HCC is a major cause of mortality in Asia and America, mainly due to the poor performance of conventional treatment and the heterogeneity (39,40). LncRNA has been extensively studied as oncogenes or tumor suppressor genes that play a crucial role in the physiological and pathological processes of various cancers, including gastric cancer, HCC, and glioma (41). Recent studies have shown that pyroptosis, a type of programmed cell death, has an important role in anti-cancer defense, activation of the immune system, and enhancing the efficacy of cancer treatment combined with chemotherapy (42,43). ...
Article
Full-text available
Background Hepatocellular carcinoma (HCC) is an invasive malignant tumor, and pyroptosis makes an important contribution to the pathology and progression of liver cancer. Many prognostic models have been proposed for HCC based on the quantitative expression level of candidate genes, which are unsuitable for clinical application due to their vulnerability against experimental batch effects. The aim of this study was to develop a novel pyroptosis-related long non-coding RNA (lncRNA)-based prognostic index (PLPI) for HCC based on relative expression orderings (REOs). Methods Firstly, the pyroptosis-related lncRNAs were identified through the Wilcoxon rank-sum test and gene co-expression analyses. Then, the novel prognostic model PLPI was constructed by pyroptosis-related lncRNA pairs, which were identified by multiple machine learning algorithms. Gene set enrichment, somatic mutation, and drug sensitivity analyses were conducted to measure the differences between high- and low-risk patients. Multiple immune analyses were used to explore the association between PLPI and the immunological microenvironment. Results In this study, a novel prognostic model PLPI based on 10 pyroptosis-related lncRNA pairs was constructed, which was proven to be an independent prognostic risk factor. The receiver operating characteristic (ROC) curves showed that the model had a good prognostic ability in the training, testing, and external set, respectively [5-year area under the curve (AUC) =0.73, 5-year AUC =0.81, 4-year AUC =0.79]. The results of survival, somatic mutation, and immune analyses showed that the patients in the low-risk group had a better prognosis, lower rates of somatic mutation, and better immune cell infiltration. Personalized chemotherapeutic drugs were also identified for the patients with HCC. Conclusions The novel PLPI not only greatly predicted the prognosis of patients with HCC but could also offer novel ideas and approaches for the therapeutic management of HCC.
... 机制尚不清楚 [74] 。目前有研究 [75] 认为,COX-2 在前 [78] ,可以导致肿瘤中的 G:C 到 A:T 颠换 [ [110][111] ,并且 ZFAS1 表达上调与结直肠癌、胃癌、卵巢癌和非小 细胞肺癌的淋巴结转移呈正相关 [112] ;ZFAS1 可以调 节 miR-588,通过 miR-588/ROCK1 轴来调控机体的 炎症反应 [113] ,而 miR-588 已被报道参与肺癌 [114] 、人 乳腺癌 [115] 、结直肠癌 [116] [120] 或 miRNA 转录变化 [121] 的结果。尽管生物合成缺陷会导致 miRNA 水平下降, 但 许 多 致 癌 miRNA 在 肿 瘤 中 显 著 增 加 [122] 。 致 癌 miRNA 在肿瘤中表达增加的机制是多样且复杂的。 研究 [123] 发现,外泌体中包含的 miRNA 可以调节肿瘤 免疫和微环境,可能促进肿瘤侵袭、转移和血管 生成。 一 些 miRNAs 可 以 调 节 炎 症 和 肿 瘤 的 联 系 。 MiR-146a 是 miRNAs 的一员,其在炎症反应中高表 达;通过调控多种靶基因的表达,有助于调节炎症 细胞因子的分泌 [124] 。MiR-146a 的功能主要通过抑制 炎症信号通路的激活来发挥作用。MiR-146a 的异常 表达与各种肿瘤的发生和进展有关,MiR-146a 的表 达在乳腺癌、激素难治性前列腺癌和胰腺癌中下 调 [125][126][127] 。此外,在肿瘤中炎症反应的激活可以促使 肿瘤细胞的侵袭能力增强,从而导致肿瘤的转移和 扩散。Zhang 等 [128] [129] 。在关于 EMT 介导的 TAM 活化的类似研究 [130] 中,EMT 转录因子 Snai1 在肿瘤中起重要作用,它能 够激活 miR-21,从而导致肿瘤细胞释放含有 miR-21 [J]. Cancers, 2021, ...
Article
ZNFX1 Antisense RNA 1 (ZFAS1) act as an oncogenic long noncoding RNA in multiple types of cancer. Ferroptosis is an iron-dependent cell death characterized by excessive iron accumulation and lipid peroxidation. However, to date, the functional role and mechanism of ZFAS1 in ferroptosis in hepatocellular carcinoma (HCC) remains largely unknown. The present study revealed that ZFAS1 was upregulated in HCC and upregulation of ZFAS1 indicated poor clinical outcome of HCC patients. Loss- and gain-of-function experiments demonstrated that knockdown of ZFAS1 inhibited HCC cell proliferation and induced ferroptosis, while overexpression of ZFAS1 exerted opposite effects. ZFAS1 enhanced cell proliferation via suppression of ferroptotic death. Mechanistically, ZFAS1 interacted with miR-150 and decreased its expression. AIFM2, the critical ferroptosis protector, was a direct target of ZFAS1/miR-150. ZFAS1 accelerated HCC proliferation and inhibited ferroptosis by the regulation of the miR-150/AIFM2 axis. These discoveries intimate an essential part of ZFAS1/miR-150/AIFM2 in governing HCC ferroptosis, which may provide a promising therapeutic strategy for HCC patients.
Article
Allergic rhinitis (AR) is a disease that is difficult to cure and accompanies the patient's life. Proinflammatory cytokines (GM-CSF and eotaxin) and MUC5AC are key mediators promoting AR progression. Herein, the function of lncRNA ZFAS1 in AR was investigated. Nasal epithelial cells (NECs) were subjected to 50 ng/mL IL-13 for 24 h to construct an AR cell model. The mRNA and protein expressions were assessed using qRT-PCR and western blot. The levels of GM-CSF, eotaxin, IL-1β, IL-6, TNF-α and MUC5AC in cell supernatant were examined by ELISA. The binding relationships between HDAC3, ZFAS1, miR-7-5p and SIRT1 were analysed using dual luciferase reporter or ChIP assays. Herein, our results displayed that ZFAS1 and SIRT1 were lowly expressed in AR, while miR-7-5p and HDAC3 were highly expressed. Functional experiments displayed that ZFAS1 overexpression suppressed IL-13-induced proinflammatory cytokines and mucin production in NECs. The highly expressed HDAC3 in AR inhibited ZFAS1 expression by binding with ZFAS1 promoter. In addition, our experiments revealed that ZFAS1 targeted miR-7-5p, and miR-7-5p targeted SIRT1. As expected, miR-7-5p overexpression or SIRT1 silencing abrogated ZFAS1 upregulation's repression on IL-13-induced proinflammatory cytokines and MUC5AC secretory levels in NECs. ZFAS1 suppressed proinflammatory cytokines, inflammatory cytokines, and MUC5AC secretory levels in AR by regulating the miR-7-5p/SIRT1 axis. Thus, our work suggested that ZFAS1 might serve as a novel target for AR treatment and prevention.
Article
Full-text available
This article will provide an overview of the evolving nature of cancer treatment, the benefits and challenges of these new treatments, and the leadership strategies required to manage the evolution from a system perspective. The number and complexity of novel cancer therapies, while offering improved patient outcomes, has become a challenge for the healthcare system due to the high cost of these new therapies. In Canada, the implementation of the pan-Canadian Oncology Drug Review and the pan-Canadian Pharmaceutical Alliance helps to streamline the review and negotiating process and to ensure consistency across provinces. Strategies to support these processes include ensuring safe patient treatment, patient counselling, clinician education, and practice innovation.
Article
Full-text available
A major shift in our understanding of genome regulation has emerged recently. It is now apparent that the majority of cellular transcripts do not code for proteins, and many of them are long noncoding RNAs (lncRNAs). Increasingly, studies suggest that lncRNAs regulate gene expression through diverse mechanisms. We review emerging mechanistic views of lncRNAs in gene regulation in the cell nucleus. We discuss the functional interactions that lncRNAs establish with other molecules as well as the relationship between lncRNA transcription and function. While some of these mechanisms are specific to lncRNAs, others might be shared with other types of genes.
Article
Full-text available
Long noncoding RNAs (lncRNAs) have been wildly demonstrated to participate in the osteosarcoma tumorigenesis. ZFAS1 is a novel identified lncRNA, however, its role in osteosarcoma is still unclear. In present study, we utilize lncRNA microarray assay to screen the lncRNA expression profile in osteosarcoma tissue, and investigate the regulatory function of ZFAS1 in osteosarcoma. LncRNA microarray assay revealed that lncRNA ZFAS1 was significantly up-regulated in 3 pairs of osteosarcoma and adjacent non-tumor tissue, which was confirmed by RT-PCR. Furthermore, in 53 pairs of osteosarcoma patient samples, the up-regulated expression of ZFAS1 was closely related to poor prognosis. In vitro, loss-of-function experiments showed that ZFAS1 knockdown significantly suppressed the proliferation, induced cycle arrest at G0/G1 phase and enhance apoptosis. In vivo, ZFAS1 knockdown inhibited the tumor growth. Bioinformatics online programs predicted that ZFAS1 sponge miR-486 at 3’-UTR with complementary binding sites, which was validated using luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Rescue experiments confirmed that miR-486 could reverse the functions of ZFAS1 on osteosarcoma genesis. In conclusion, our results demonstrate that ZFAS1 act as competing endogenous RNA (ceRNA) for miR-486, and act as oncogene in osteosarcoma tumorigenesis, and discover the functional regulatory pathway of ZFAS1 sponging miR-486.
Article
Full-text available
A number of studies have revealed that zinc finger antisense 1 (ZFAS1), a long noncoding RNA (lncRNA), is aberrantly regulated in various cancers, and high ZFAS1 expression is associated with poor prognosis and increased risk of lymph node metastasis (LNM). This meta-analysis was conducted to identify the potential value of ZFAS1 as a biomarker for cancer prognosis. We searched electronic database PubMed, Web of Science, and China Wanfang Data (up to June 1, 2017) to collect all relevant studies and explore the association of ZFAS1 expression with overall survival (OS) and LNM. The results showed that cancer patients with high ZFAS1 expression had a worse OS than those with low ZFAS1 expression (HR: 1.94, 95% confidence interval [CI]: 1.41–2.47, P < 0.001), and high ZFAS1 expression was significantly associated with LNM (OR: 2.60, 95% CI: 1.54–4.42, P < 0.001). Subgroup analysis revealed that high ZFAS1 expression was significantly related to high incidence of LNM in subgroups of sample size more than 88 (OR: 3.16, 95% CI: 2.06–4.86, P < 0.001), non-digestive system malignancies (OR: 4.05, 95% CI: 2.49–6.60, P < 0.001), and studies reported in 2017 (OR: 4.86, 95% CI: 2.67–8.84, P < 0.001) without significant heterogeneity. Further meta-regression by the covariates showed that tumor type, sample size, quality score, cut off value and publication year did not result in the inter-study heterogeneity. In conclusion, the present meta-analysis demonstrates that high ZFAS1 expression may potentially serve as a reliable biomarker for poor clinical outcome in various cancers.
Article
Full-text available
Background Increasing evidence has demonstrated that Notch signaling is deregulated in human hematological malignancies and solid tumors. This signaling has a protumorigenic effect but may also act as a tumor suppressor. How induction of a single pathway gives rise to the opposite effects in different cell types is still unknown. Methods This review article includes available data from peer-reviewed publications associated with the role of Notch signaling during cancer pathogenesis. Results Numerous reports have indicated that alterations in Notch signaling and its oncogenic activity were originally associated with the pathogenesis of T‑cell acute lymphoblastic leukemia/lymphoma (T-ALL), an aggressive hematologic tumor affecting children and adolescents. The possibility that Notch could play a significant role in human breast cancer development comes from studies on mouse mammary tumor virus-induced cancer. Numerous findings over the past several years have indicated that alterations in Notch signaling are also responsible for ovarian cancer development. Mention should also be made of the connection between expression of Notch 3 and increased resistance to chemotherapy, which remains a major obstacle to successful treatment. Notch as an oncogenic factor is also involved in the development of colon cancer, lung carcinoma and Kaposi’s sarcoma. Conclusion Notch is a binary cell fate determinant and its overexpression has been described as oncogenic in a wide array of human malignancies. This finding led to interest in therapeutically targeting this pathway, especially by the use of gamma-secretase inhibitors (GSIs) blocking the cleavage of Notch receptors at the cell membrane by the inhibition of Notch intracellular domain (NICD) releasing. Preclinical cancer models have revealed that GSIs suppress the growth of cancers such as pancreatic, breast and lung cancer.
Article
Full-text available
Background There is increasing evidence of a relationship between long non-coding RNA (lncRNA) and cancer. This study aimed to examine the prognostic value of the lncRNA ZFAS1 in esophageal squamous cell carcinoma (ESCC). Results The results showed that ZFAS1 expression was significantly higher in ESCC tissues compared with the corresponding adjacent normal tissues (P < 0.001). ESCC patients with high ZFAS1 expression had a poor overall survival (OS). Histological grade, T stage and ZFAS1 expression were integrated to develop the nomogram. The nomogram showed a significantly better prediction of OS for patients with lymph node-negative ESCC. The ROC curve also showed higher specificity and sensitivity for predicting 3- and 5-year ESCC patient survival compared with the AJCC staging system. The decision curve analysis also indicated a greater potential for the nomogram in clinical application compared with the AJCC staging system. Importantly, our findings were supported by a validation cohort. Materials and Methods We retrospectively investigated 398 lymph node-negative ESCC patients. Data from the primary cohort (n = 246) were used to develop a multivariate nomogram. The nomogram was internally validated for discrimination and calibration with bootstrap samples and was externally validated with an independent patient cohort (n = 152). Conclusions Our proposed nomogram, which integrates clinicopathological factors and ZFAS1 expression, can accurately predict the prognosis of lymph node-negative ESCC patients without preoperative chemoradiotherapy.
Article
Full-text available
Expression of ZFAS1, a newly identified long noncoding RNA (lncRNA), is dysregulated in several types of cancer. Here we assessed the prognostic value of ZFAS1 in solid tumors. A comprehensive literature search was performed by screening the PubMed, EMBASE, MEDLINE, Cochrane Library, CNKI, and Wanfang databases. A total of 874 patients from 10 studies were included. The pooled analysis demonstrated that patients with high ZFAS1 expression had a significantly shorter overall survival (OS) (HR, 1.58; 95% CI, 1.28-1.97; P < 0.001) and recurrence-free survival (RFS) (HR, 1.90; 95% CI, 1.29-2.79; P = 0.001). Moreover, elevated ZFAS1 expression correlated with tumor size, tumor-node-metastasis (TNM) stage, and lymph node metastasis (LNM). These results demonstrate that increased ZFAS1 expression correlates with a poor prognosis in cancer patients, which suggests ZFAS1 might be useful as a potential prognostic biomarker in patients with solid tumors.
Article
In this review, we outline the biology and management of patients with carcinosarcomas and related malignancies, which are often included under the broader concept of sarcomatoid carcinomas. Carcinosarcomas are unusual tumors that are commonly gynecologic in origin, where they are referred to as malignant mixed Müllerian tumors, but may appear in any anatomic site. Although a variety of hypotheses have been presented as to the biphasic nature of these tumors, carcinosarcomas seem to represent the best example in human cancers of the concept of epithelial-mesenchymal transition (EMT), in which the two parts of the tumor are genomically related to one another, as opposed to the mesenchymal component that represents a second neoplasm or (benign) reactive process. In general, patients with carcinosarcomas fare worse than patients with carcinomas of the same anatomic site. Treatment paradigms for carcinosarcomas generally follow those of carcinomas of the same organ site, except where clinical trials provide more specific options. Agents that block or reverse EMT are worth examination in patients with carcinosarcoma and arguably may be even more effective in carcinomas, given evidence of dependence on EMT to generate successful metastases. Information about EMT may also inform other phase transitions in cancer, such as those between prostate or lung carcinoma and more aggressive tumors with neuroendocrine differentiation.
Article
The incidence and mortality rate of colorectal cancer (CRC) have been significantly increasing. However, mechanisms involved in CRC progression are still unclear. LncRNA ZFAS1 has been verified as oncogenic molecular in a series of tumors, including CRC. However, the underlying mechanism of ZFAS1 in CRC carcinogenesis remains unclear. In the present study, our data showed that ZFAS1 expression was significantly upregulated in CRC tissues and cell lines. Correlation analysis showed that high ZFAS1 expression was significantly associated with Helicobacter pylori infection, lymph nodes metastasis, advanced TNM stage and poor overall survival of CRC patients. Loss-of-function experiments revealed that ZFAS1 inhibition could markedly suppress CRC cells proliferation and invasion both in vitro and in vivo. Bioinformatics analysis and luciferase reporter assay revealed that ZFAS1 directly interacted with miR-484. Rescue experiments showed that miR-484 inhibitor reversed the tumor suppressing roles of ZFAS1 knockdown on CRC cells. Therefore, our study suggested that ZFAS1 could act as an oncogene in CRC tumorigenesis, and discovered the functional regulatory pathway of ZFAS1 sponging miR-484.
Article
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. LncRNAs have recently gained widespread attention and have been shown to have crucial roles in various biological regulatory processes. ZFAS1, a newly identified lncRNA, was shown to be dysregulated in several cancers. However, little is known about the alteration and functional significance of ZFAS1 in OS. In the present study, for the first time, we revealed a functional role of ZFAS1 on OS growth and metastasis. The expression of ZFAS1 was significantly overexpressed in OS samples and cell lines, and upregulation of ZFAS1 is significantly associated with unfavorable prognosis of OS patients. Functional assays also demonstrated that ZFAS1 enhanced the growth and metastatic ability of OS cells in vitro and in vivo. Mechanistically, we found that ZFAS1 positively regulated malignant phenotypes by competitively binding the miR-200b and miR-200c and upregulating BMI1. ZFAS1 also interacted with ZEB2 and regulated ZEB2 protein stability. Furthermore, we demonstrated that SP1 functions as an upstream activated factor of ZFAS1. ZFAS1 may be a potential therapeutic target for OS tumorigenesis and progression.