Abstract
Despite improvement in our understanding of long noncoding RNAs
(lncRNAs) role in cancer, efforts to find clinically relevant
cancer-associated lncRNAs are still lacking. Here, using nascent RNA
capture sequencing, we identify 1145 temporally expressed
S-phase-enriched lncRNAs. Among these, 570 lncRNAs show significant
differential expression in at least one tumor type across TCGA data
sets. Systematic clinical investigation of 14 Pan-Cancer data sets
identified 633 independent prognostic markers. Silencing of the top
differentially expressed and clinically relevant S-phase-enriched
lncRNAs in several cancer models affects crucial cancer cell hallmarks.
Mechanistic investigations on SCAT7 in multiple cancer types reveal
that it interacts with hnRNPK/YBX1 complex and affects cancer cell
hallmarks through the regulation of FGF/FGFR and its downstream
PI3K/AKT and MAPK pathways. We also implement a LNA-antisense
oligo-based strategy to treat cancer cell line and patient-derived
tumor (PDX) xenografts. Thus, this study provides a comprehensive list
of lncRNA-based oncogenic drivers with potential prognostic value.
__________________________________________________________________
Although we know lncRNAs play a role in cancer, the identification of
clinically relevant and functional lncRNAs is lacking. Here, the
authors identify 633 prognostic markers, 570 S-phase cancer-associated
lncRNAs, and show SCAT7 regulates FGF/FGFR and PI3K/AKT/MAPK pathways
via interaction with hnRNPK/YBX1 complexes.
Introduction
Recent ultra-high-throughput transcriptome sequencing data suggest that
nearly two-thirds of the human genome is pervasively transcribed,
giving rise to more than double the number of lncRNAs compared to
protein-coding RNAs^[40]1. It is evident from studies that lncRNAs do
not represent transcriptional noise, but they participate in diverse
biological functions with implications in development, differentiation,
and disease^[41]2–[42]4. lncRNAs operate at the transcriptional and
post-transcriptional level to control gene expression in a
spatiotemporal fashion^[43]5. Considering the influence of lncRNAs in a
wide-range of biological processes, and that a significant portion of
disease-associated single-nucleotide polymorphisms (SNPs) map to lncRNA
loci^[44]6, one could expect a greater role for lncRNAs in human
disease. Recent investigations have also implicated numerous lncRNAs in
cancer progression and metastatic dissemination^[45]7–[46]10. Despite
the increased number of studies addressing the role of lncRNAs in
cancer, our understanding of lncRNAs in cancer initiation and
progression as well as their relevance to clinical prognosis is in its
infancy. Hence large-scale lncRNA-based functional screens, coupled
with clinical investigations, are required to understand their roles in
cancer progression and their use for diagnostic and prognostic
purposes.
The ability to sustain chronic proliferation is considered one of the
hallmarks of cancer^[47]11. The cell division cycle plays a central
role in the control of normal and chronic cell proliferation via
responding to cell cycle regulators such as cyclins and
cyclin-dependent kinases (CDKs), growth factors and repressors, tumor
suppressors and oncogenes^[48]11. Cell division comprises two phases:
the DNA synthesis (S) phase and the mitotic (M) phase. The pathways
controlling accurate DNA replication during S-phase are critical for
normal cell proliferation, as replication errors could result in
abnormal cell proliferation^[49]12,[50]13. Thus, characterization of
molecules that control S-phase progression would be of immense
importance in understanding the role of S-phase in normal and disease
conditions.
The role of lncRNAs in S-phase regulation is still poorly understood in
comparison to protein-coding genes. Accumulated knowledge has so far
been unable to explain the precise molecular pathways that control cell
proliferation in normal and cancer conditions^[51]11,[52]14. Thus,
exploring the functional role of lncRNAs in cell cycle progression may
provide insights into hitherto unanswered questions about how cell
division is controlled in normal and cancer cells. Consistent with
this, several investigations have shown that lncRNAs regulate cell
cycle progression through controlling the expression of critical cell
cycle regulators such as cyclins, CDKs, CDK inhibitors, and other
factors^[53]15–[54]17. In addition, a recent investigation implicated a
subset of lncRNAs from 56 well-known cell cycle-linked loci, showing
periodic expression patterns across cell cycle phases, in cell cycle
regulation^[55]18. However, a comprehensive characterization of lncRNAs
showing temporal expression during S-phase and their functional link to
cell cycle regulation, cancer progression, and their use in prognosis
has not been investigated.
In this study, we address two fundamental propositions that can enhance
the current comprehension of the critical roles of lncRNAs in cancer
development and progression. First, we study the contribution of
S-phase-specific lncRNAs to cell cycle progression and other important
cancer-associated hallmarks. Second, we investigate the capability of
S-phase-specific lncRNAs to act as independent prognostic biomarkers in
different cancers. To address these aims, we employ nascent RNA-seq
during S-phase progression and generate a resource of lncRNA-based
oncogenic cancer drivers that can be used as prognostic markers in the
risk assessment as well as potential therapeutic regimens in the
treatment of cancer.
Results
Identification of temporally expressed lncRNA across S-phase
We have previously reported an optimized nascent RNA capture assay for
detecting temporally expressed S-phase RNAs in HeLa cells^[56]19.
Compared to the standard steady-state RNA isolation from various
S-phase stages, this method detects accurate expression timing of the
analyzed transcripts^[57]19. Briefly, HeLa cells were synchronized at
the G1/S boundary using thymidine and hydroxyurea. Subsequently, the
G1/S block was released and the cells were allowed to proceed in the
presence of 5-ethynyl uridine (EtU) in order to specifically label the
newly synthesized nascent RNA populations across different S-phase time
points. To identify S-phase-specific lncRNAs, EtU labeled and
steady-state (unlabeled) RNAs were individually collected at different
time points spanning S-phase (T1, T2, and T3) in addition to RNA from
unsynchronized cells (Fig. [58]1a). Following strand-specific library
preparation, all RNA samples were subjected to deep sequencing
(Supplementary Data [59]1). Principal component analysis of whole RNA,
noncoding RNAs, and lncRNA expression profiles revealed that
EtU-labeled RNAs had a better separation across different time points
of S-phase compared to unlabeled samples (Fig. [60]1b). Based on the
enrichment over unsynchronized samples, we identified 1734 and 1674
lncRNAs in EtU labeled and unlabeled samples, respectively. Further, to
unveil the dynamically expressed S-phase lncRNAs, we performed Short
Time-series Expression Miner (STEM) clustering^[61]20. We obtained 1145
lncRNAs with four significant temporal expression patterns, and 937
lncRNAs with two significant temporal patterns, in EtU labeled
(Fig. [62]1c; Supplementary Data [63]1) and unlabeled samples
(Supplementary Fig. [64]1a and Supplementary Data [65]1), respectively.
Both EtU labeled and unlabeled samples shared 394 temporally expressed
lncRNAs (Fig. [66]1c; Supplementary Fig. [67]1b). We next investigated
the enrichment of E2F1 transcription factor binding sites at the
promoters (±2 kb from transcription start site (TSS)) of temporally
expressed lncRNAs and found that 72 EtU labeled and 67 unlabeled lncRNA
promoters contain E2F1 binding sites (Supplementary Data [68]1). We
randomly selected dynamically expressed S-phase lncRNAs and validated
their temporal expression patterns across different cell cycle phases
(G1, S, and G2/M) in EtU labeled and unlabeled samples (Supplementary
Fig. [69]1c). To further confirm the specificity of this method in
capturing temporally expressed lncRNAs during S-phase, we validated
selected lncRNAs using a drug-free synchronization of HeLa cells using
the serum starvation method (Supplementary Fig. [70]1d, e).
Fig. 1.
[71]Fig. 1
[72]Open in a new tab
Identification of temporally expressed lncRNAs across S-phase using
nascent RNA capture assay. a Cell cycle diagram showing Nascent RNA
capture at three different time points (2, 3.5, and 5 h) of S-phase. b
Principle component analysis (PCA) of expression profiles by
considering complete profile (noncoding and protein-coding), all
noncoding and only lncRNAs. c Time-series analysis of S-phase
associated lncRNAs with twofold enrichment at least in one time point
over the unsynchronized sample. The S-phase lncRNAs show four
significant temporal patterns with STEM clustering. Venn diagram shows
the overlap of lncRNAs enriched in EtU labeled and unlabeled samples.
The p-values are obtained using permutation tests from STEM clustering.
d Molecular pathway analysis (left) and gene ontology (right)
enrichment analysis for nearby (<50 kb proximity) protein-coding genes
to S-phase associated lncRNAs. The KEGG pathways or biological
functions presented in the heatmaps show at least 10 genes with
p-value < 0.05. The hypergeometric p-values are obtained from GeneSCF
for pathway and GO enrichment analysis. e Workflow of the analysis for
the identification of S-phase associated lncRNAs and its significance
in different cancers
Since lncRNAs exert their actions via regulating protein-coding RNAs in
cis and/or trans^[73]21–[74]24, we performed enrichment analysis using
GeneSCF^[75]24 for the neighboring protein-coding genes (within ±50 kb
window of S-phase-specific lncRNAs). Interestingly, the neighboring
protein-coding genes associated with EtU-labeled S-phase-specific
lncRNAs demonstrated significant enrichment of molecular pathways
related to cancer (Fig. [76]1d; Supplementary Data [77]1). Moreover,
biological process analysis revealed an enrichment of transcriptional
regulation, cell division, DNA repair, and cell migration processes. On
the other hand, the protein-coding genes associated with unlabeled
lncRNAs showed less enrichment of cancer-related pathways and cell
division processes. These results indicate that nascent RNA enrichment
via EtU labeling can efficiently distinguish temporally expressed
lncRNAs during S-phase, which may be functionally engaged in vital
cellular processes.
S-phase lncRNAs show differential expression in cancers
We next designed a workflow which integrated epigenomic, functional,
and clinical approaches to address the functional implications of
EtU-labeled S-phase lncRNAs in multiple cancer types (Fig. [78]1e).
Since, cell cycle perturbation is one of the hallmarks of cancer
development^[79]11, we addressed the functional connection between
S-phase lncRNAs and cancer initiation and progression. We utilized the
RNA-sequencing data of 6419 solid tumors and 701 normal tissue samples
spanning 16 cancer types, from The Cancer Genome Atlas (TCGA)^[80]25
(Supplementary Data [81]2). Our analysis revealed that 570 out of 1145
S-phase-specific lncRNAs were differentially expressed in at least one
cancer type with log-fold change ± 2 and false discovery rate
(FDR) < 1E–004 (Methods section, Fig. [82]2a; Supplementary
Data [83]2). Interestingly, nearly 73% of the differentially expressed
lncRNAs show upregulation in corresponding cancer types. We found that
84 S-phase lncRNAs were differentially expressed between normal and
tumor tissues, in more than five cancer types, with four of them
differentially expressed in 10 cancer types. Also, different cancer
types originating from same tissues shared many differentially
expressed S-phase lncRNAs (Fig. [84]2b). For instance, lung
adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) share 98
differentially expressed S-phase lncRNAs. Likewise, kidney chromophobe
(KICH), kidney papillary carcinoma (KIRP), and kidney clear cell
carcinoma (KIRC) harbor 16 common differentially expressed S-phase
lncRNAs. Since the latter cancers originate from distinct cell
types^[85]26,[86]27, it remains to be seen whether these common and
differentially expressed S-phase lncRNAs underpin the etiology of
different kidney and lung cancers in a cell-independent manner. These
observations indicate that temporally expressed lncRNAs show
differential expression across multiple cancers and could serve as
potential candidates for understanding their role in cancer development
and progression.
Fig. 2.
[87]Fig. 2
[88]Open in a new tab
Characterization of S-phase lncRNAs as oncogenic drivers and
independent prognostic markers using pan-cancer TCGA data sets. a
Heatmap of 570 S-phase lncRNAs showing significant differential
expression at least in one cancer type from TCGA. The significance was
considered based on log-fold change ± 2 and FDR < 1E–004
(Benjamini–Hochberg’s method). The highlighted lncRNAs are the ones
selected for functional validation. The plot on the left side shows the
frequency of individual lncRNAs differentially expressed across
different cancer types. b Venn diagrams illustrating the overlap
between common S-phase lncRNAs which are differentially expressed in
different types of kidney and lung cancers. c Heatmap of S-phase
lncRNAs with anti-correlative promoter methylation status to the
differential expression levels in corresponding cancer. Hypomethylated
promoter associated with higher expression and hypermethylation to less
expression compared to normal. The highlighted lncRNAs are
differentially methylated in more than 100 patients (samples)
supporting the methylation pattern in corresponding cancer. Box plots
of highlighted lncRNAs denoting the anti-correlative relation between
promoter methylation and transcript expression. Box plots middle line
shows the median, the box limits are 25th and 75th percentiles,
whiskers are nearer quartile ± 1.5 times interquartile range and points
beyond whiskers are the outliers. The p-values for the comparisons were
obtained using Mann–Whitney test and corrected p-values using Family
Wise Error Rate (FWER). The significant differentially methylated
regions were filtered on the basis of FWER < 0.05. The above
information for statistical analysis was extracted from COSMIC
repository. d Heatmap showing the potential independent prognostic
values of 520 S-phase lncRNAs based on dichotomization approach. The
red color indicates higher expression of lncRNAs that predicts poor
survival outcome. The blue color indicates lower expression of lncRNAs
associated with poor survival in patients. e Venn diagrams indicating
the numbers of S-phase lncRNAs that independently predict the survival
outcome in different types of kidney and lung cancers
S-phase lncRNAs show epigenetic alterations
We set out to address whether differential CpG methylation underlies
the differential expression of S-phase lncRNAs seen in normal and tumor
samples. We analyzed CpG methylation over their genomic regions in
relation to gene expression. We utilized the processed CpG methylation
data from the catalog of somatic mutations in cancers (COSMIC) for 12
TCGA cancer types (Infinium Human Methylation 450 beadchip platform;
~4000 samples, from international cancer genome consortium (ICGC)
release 18)^[89]28 (Supplementary Data [90]2). Differential methylation
was considered significant only if a particular pattern
(hypomethylation or hypermethylation) was supported by at least 10
patient samples and with null patients opposing the pattern (Methods
section). CpG methylation analysis over 570 differentially expressed
S-phase lncRNA promoters revealed 35 lncRNAs that exhibit differential
methylation (Fig. [91]2c). Among these, 22 lncRNAs were hypomethylated
with higher expression in tumors, whereas 13 lncRNAs were
hypermethylated with lower expression in tumors. Since gene-body
methylation has been shown to be positively correlated with gene
expression^[92]29, we analyzed gene-body methylation over S-phase
lncRNAs and found 20 transcripts with differential methylation over the
gene-body that correlated with gene expression (Supplementary
Fig. [93]2). Out of 20 lncRNAs, 7 were hypermethylated and highly
expressed, whereas 13 lncRNAs were hypomethylated with lower expression
in tumors. Importantly, the promoter region of RP11-152P17.2 was
hypomethylated and highly expressed in HNSC, KIRP, and LIHC tumors
compared to normal tissues (Supplementary Data [94]2).
S-phase lncRNA act as independent prognostic markers
We employed a systematic approach to identify the prognostic value of
S-phase lncRNAs across 14 cancers. For each cancer type, we stratified
the patients into high and low expression groups based on the
expression levels of each lncRNA (Methods section). Then, we assessed
the difference in survival using the Kaplan–Meier (KM) method. We
tested the randomness in survival prediction using receiver operating
characteristic^[95]7 curves and filtered candidates with an area under
the curve (AUC) less than 0.5. To calculate the hazard ratios
associated with the expression of each S-phase lncRNAs in each
respective cancer type, we generated a multivariate Cox-regression
model (Supplementary Data [96]2). The accuracy of the predictive
multivariate prognostic models was investigated using Brier score-based
prediction error curve over the reference model. Our systematic
analysis based on dichotomization approach identified 520 S-phase
lncRNAs that predict survival and also act as potential independent
biomarkers at least in one cancer type (Fig. [97]2d, e; Supplementary
Data [98]2). In addition, we identified 375 S-phase lncRNAs as
continuous biomarkers in at least one cancer type (Supplementary
Data [99]2) with a significant overlap of 262 S-phase lncRNAs with the
dichotomization-based approach (Supplementary Fig. [100]2b and
Supplementary Data [101]2). Notably, the multivariate models
demonstrated that KIRC harbors the maximum number of S-phase
lncRNA-based prognostic biomarkers. Of note, RP11-59D5__B.2 is the top
candidate in our clinical investigations and it acts as an independent
prognostic indicator in five cancers: BLCA, KIRC, KIRP, STAD, and HNSC
(Supplementary Fig. [102]2c-f and Supplementary Data [103]2). Taken
together, a large proportion of S-phase lncRNAs show an independent
prognostic capacity, in relation to clinical covariates, in different
cancer types.
SCATs modulate cell cycle progression and cell proliferation
For further functional validation, we selected eight S-phase lncRNAs
among the top candidates having a higher frequency of differential
expression across TCGA data sets (Fig. [104]2a) which also
independently predict the patients’ survival in multiple cancers
(Fig. [105]2d, Fig. [106]3a; Supplementary Fig. [107]3). Hereafter, we
address the selected candidates as S-phase cancer-associated
transcripts (SCATs). We depleted the SCATs using small-interfering RNA
oligonucleotides (siRNAs) or locked nucleic acid (LNA) antisense
oligonucleotides using HeLa cells as a model system (Supplementary
Fig. [108]3i). We assessed the effect of SCATs downregulation on cell
proliferation using the colorimetric MTT assay (Fig. [109]3b). SCATs
depletion induced a significant effect on cell proliferation.
Similarly, the impact of SCATs loss-of-function on cell cycle
progression was assessed using flow cytometry (Fig. [110]3c). Loss of
function of SCATs induced cell cycle perturbations, accompanied with an
accumulation of cells at the G1 phase and a decrease in DNA synthesis.
Furthermore, downregulation of SCATs induced cellular apoptosis, as
indicated by a significant increase in caspase 3/7 activity
(Fig. [111]3d).
Fig. 3.
[112]Fig. 3
[113]Open in a new tab
The top clinically relevant S-phase lncRNAs regulate crucial cancer
cell hallmarks. a Kaplan–Meier plots of SCAT1 –SCAT8 indicating overall
survival of patients in KIRC. The higher expression of all SCATs is
correlated with poor overall survival. The expression cut-off and the
significance value for each SCAT are indicated in the plots. UQ
represents upper quartile of the patients’ expression levels. The
Forest plots represent the multivariate models derived for each SCAT in
combination with the significant clinical parameters. The hazard ratio
(HR) using Cox proportional hazard analysis and the associated p-values
were calculated using Wald statistics. b Proliferation capacity of HeLa
cells as measured by MTT colorimetric assay 48 h post-silencing of
SCATs using two different LNAs (SCAT1, SCAT2, SCAT4, SCAT5, and SCAT6)
or siRNAs (SCAT3 and SCAT7). Data are represented as percentage
compared to cells transfected with respect to the negative control. No
significant difference was observed between LNA-negative control and
siRNA-negative control. c Cell cycle profiles of HeLa cells depleted
with two different LNAs or siRNAs targeting the seven SCATs. d
Estimation of the caspase 3/7 activities 48 h post-silencing of SCATs
in HeLa cells. Data are expressed as fold change with respect to the
corresponding negative controls. e MTT proliferation assay of Caki-2
(KIRC) cell line depleted with two independent LNAs (SCAT4, SCAT8) or
siRNAs (SCAT7). f Cell cycle profiles of Caki-2 cells depleted with two
different LNAs or siRNAs. g Estimation of the caspase 3/7 activities
48 h post-silencing of the corresponding SCAT in Caki-2 cells. Data in
b–g are shown as mean ± SEM of three independent experiments.
Significance levels were derived using unpaired two-tailed Students’
t-test. (*p ≤ 0.05; **p = 0.01 – 0.001; ***p < 0.001)
Since some of the SCATs showed differential expression and predict
survival in KIRC tumors, we used a KIRC cell line Caki-2, to further
validate their functional role in cell homeostasis. First, we
investigated the temporal expression patterns of SCAT4, SCAT5, SCAT7,
and SCAT8 during cell cycle progression in serum-starved Caki-2 cells
and found their elevated expression during S-phase (Supplementary
Fig. [114]3j). Then, we downregulated SCAT4, SCAT7, and SCAT8
(Supplementary Fig. [115]3k) and measured cell proliferation, cell
cycle profile, and caspase activity. Consistent with the
loss-of-function experiments in HeLa cells, depletion of the selected
SCATs in Caki-2 cells altered cell proliferation, inhibited cell cycle
progression, and induced apoptosis (Fig. [116]3e-g).
SCAT1 and SCAT5 act as as independent prognostic factors
Our clinical investigations revealed CTD-2357A8.3 (SCAT1) and LUCAT1
(SCAT5) as common independent prognostic biomarkers for lung and
kidney-derived cancers, respectively (Fig. [117]2e). SCAT1 is
differentially expressed in 10 cancers (Fig. [118]4a), including LUAD
and LUSC. Higher expression of SCAT1 in both LUAD and LUSC correlates
with the poor clinical outcome (Fig. [119]4b; Supplementary
Fig. [120]4a-f). Additionally, SCAT1 shows an elevated expression in
the S-phase of synchronized A549 cells (Supplementary Fig. [121]4g).
Consistent with the clinical data, downregulation of SCAT1 in A549
cells exhibited a drastic inhibition of cell proliferation
(Fig. [122]4c; Supplementary Fig. [123]4h), cell cycle arrest at G1
phase (Fig. [124]4d), and promoted cellular apoptosis (Fig. [125]4e),
indicating its role in lung carcinogenesis. Similarly, SCAT5, which is
differentially expressed in five cancers (Fig. [126]4f), acts as an
independent prognosticator in all three types of kidney cancers (KIRP,
KIRC, and KICH) (Figs. [127]3a and [128]4g; Supplementary
Fig. [129]4i-n and Supplementary Data [130]2). Its higher expression
predicts poor clinical outcome in all three cancers and similar to
other SCATs, SCAT5 expression is induced during S-phase in Caki-2 cells
(Supplementary Fig. [131]3j). The depletion of SCAT5 in Caki-2 cells
led to decrease in cell proliferation, cell cycle arrest at G1 phase,
and induces apoptosis (Fig. [132]4h-j; Supplementary Fig. [133]4o).
Fig. 4.
[134]Fig. 4
[135]Open in a new tab
SCAT1 and SCAT5 act as oncogenic drivers and prognostic markers for
lung-derived and kidney-derived cancers, respectively. a Bar graph
showing the significant differential expression levels of SCAT1
expressed as log[2] fold change across 10 different cancer types
obtained from TCGA data sets. b Kaplan–Meier plots of SCAT1 indicating
overall survival of patients in LUAD (upper left panel) and LUSC (lower
left panel) cancer types. The higher expression of the SCAT1 is
correlated with poor overall survival. The upper and lower right panels
represent the multivariate models of LUAD and LUSC cancers,
respectively, derived from Cox proportional hazard analysis and
associated p-values were calculated using Wald statistics. c MTT
proliferation assay of A549 (LUAD) cell line depleted with two
different LNA oligos targeting SCAT1. d Cell cycle profiles of control
and SCAT1 KD A549 cells. e Estimation of caspase 3/7 activity in
control and SCAT1 KD A549 cells. f Bar graph showing the significant
differential expression levels of SCAT5 expressed as log[2] fold change
across five different cancer types obtained from TCGA data sets. g
Kaplan–Meier plots of SCAT5 indicating overall survival of patients in
KICH (upper left panel) and KIRP (lower left panel) kidney cancer. The
higher expression of the SCAT5 is correlated with poor overall
survival. The upper and lower right panels represent the multivariate
models of KICH and KIRP cancers, respectively. h MTT proliferation
assay of Caki-2 cell line 48 h post-silencing of SCAT5 using two
different LNAs. i Cell cycle profiles of control and SCAT5 KD Caki-2
cells. j Estimation of caspase 3/7 activity in Caki-2 cells 48 h
post-silencing of SCAT5. The significance in figures a and f was
derived using Benjamini–Hochberg’s method. Note that the data presented
in c–e and h–j represents the mean values of three independent
experiments and statistical significance was derived using a two-tailed
unpaired Student’s t-test. Data are plotted as mean ± SD (*p ≤ 0.05;
**p = 0.01 – 0.001; ***p < 0.001)
SCAT7 modulates hallmarks of cancer across cell lines
We chose RP11-465N4.4 (SCAT7) to gain mechanistic insights into S-phase
lncRNA induced cancer initiation and progression. Current annotations
refer to SCAT7 as a polyadenylated antisense lncRNA with two variants
of 788 and 500 nucleotides. The 788 nucleotide variant spans two
protein-coding genes (RNPEP and ELF3). Northern blot analysis also
confirmed the presence of these two variants (Supplementary
Fig. [136]5a). The noncoding capacity of SCAT7 was confirmed using both
CPAT probability and CPC score (Supplementary Fig. [137]5a). The
sequence conservation analysis of SCAT7, as determined by phastCons
score and phyloP, indicates no sequence homology among vertebrates
(Supplementary Fig. [138]5b). SCAT7 RNA copy number analysis of HeLa
and A549 cells by droplet digital PCR (ddPCR) revealed 224 and 496
copies/ng RNA, respectively (A549 cells known to have >2500 copies
MALAT1 and NEAT1^[139]30).
SCAT7 was upregulated in multiple cancers (BLCA, BRCA, KIRP, LIHC,
LUAD, LUSC, PRAD, and UCEC) (Fig. [140]5a). Moreover, our detailed
clinical investigations demonstrated its potential to independently
predict clinical outcome in KIRC and COAD patients (Fig. [141]3a;
Supplementary Fig. [142]3g and Supplementary Data [143]2). Furthermore,
SCAT7 promoter showed significant differential methylation in UCEC
patients (Fig. [144]2c; Supplementary Data [145]2). The temporal
expression analysis of SCAT7 during cell cycle progression indicated
elevated levels in the S-phase of HeLa and Caki-2 cell lines
(Supplementary Figs. [146]1c, d and [147]3j). Furthermore, qPCR
expression analysis indicated that SCAT7 exhibits an elevated
expression in LUAD cell lines (A549 and H2228) compared to normal lung
cells (Supplementary Fig. [148]5c). Collectively, these results
indicate that SCAT7 could be an oncogenic lncRNA with a potential
prognostic capability. Hence, we sought to investigate the functional
role of SCAT7 in the maintenance of cancer cell hallmarks in different
cell lines representing multiple cancer types. To this end, both
variants of SCAT7 were downregulated in cell lines representing KIRC
(Caki-2 and 786-O), LUAD (A549 and H2228), LIHC (HEPG2), and BRCA
(MCF7) using siRNA or short hairpin RNA (shRNA). Downregulation of
SCAT7 variants in the knockdown cells in Northern blot analysis
indicates the specificity of our RNAi knockdown experiments
(Supplementary Fig. [149]5a). Downregulation of SCAT7 in Caki-2, 786-O,
A549, H2228, HepG2, and MCF7 cells affected cell proliferation
(Fig. [150]5b; Supplementary Fig. [151]5d, e) and cell cycle
progression, with preferential accumulation of cells at G1 phase
(Fig. [152]5c; Supplementary Fig. [153]5f). Additionally, the knockdown
of SCAT7 led to decreased proliferation even in the non-cancerous
HEK293 cells (Supplementary Fig. [154]5g). Furthermore, SCAT7 depletion
altered the S-phase progression in multiple cell lines as indicated by
the nascent 5-ethynyl-2’-deoxyuridine analog (EdU) incorporation assay
(Supplementary Fig. [155]5h). Notably, compared to other cell lines,
the transient knockdown of SCAT7 in H2228 cells induced a significant
accumulation of cells at G2/M phase (Supplementary Fig. [156]5f).
Fig. 5.
[157]Fig. 5
[158]Open in a new tab
SCAT7 acts as an oncogenic driver in renal, lung, and liver cancers. a
SCAT7 expression as log[2] fold change across cancer types from TCGA
data sets. The significance was obtained using Benjamini–Hochberg’s
method. b MTT of Caki-2, A549, and HepG2 cells upon SCAT7 KD with two
shRNAs or siRNAs. c Cell cycle profiles of Caki-2, A549, and HepG2
cells after shRNA or siRNA-based SCAT7 KD. d Percentage of apoptotic
cells in SCAT7 KD Caki-2 and A549 48 h post-seeding. e Migration areas
for stable SCAT7 KD Caki-2 and A549 cells were calculated with respect
to a starting (t = 0) migration control area for each cell line. f
Matrigel transwell assay in Caki-2 and A549 SCAT7 stable KD cells. The
number of invasive cells was counted 24 h post-seeding. g Soft agar
colony-forming assay using Caki-2 and A549 KD cells. h MTT of HeLa,
Caki-2, and A549 cells overexpressing SCAT7. i Colorimetric
β-galactosidase staining of BJ-BRAF and TIG3-BRAF human fibroblasts
72 h post-silencing SCAT7 using three siRNAs. Senescent cells are in
dark blue color. j Quantification of senescent cells upon SCAT7 KD in
BJ-BRAF and TIG3-BRAF cells shown as percentage of the whole cells
populations. k SCAT7 qPCR in serial passages of BJ-BRAF cells. NS, not
significant. l SCAT7 expression in BJ-BRAF cells at day 0 and day 3
upon tamoxifen-induced senescence (200 nM) at one passage interval. m
MTT assay of BJ-BRAF cells overexpressing SCAT7 compared to empty
vector. n Percentage of positively stained senescent cells 3 days
post-tamoxifen treatment in control and SCAT7-overexpressing BJ-BRAF
cells. o, p Expression of SCAT7, p16, p15 (o) and IL8 (p) in control
BJ-BRAF and SCAT7-overexpressing cells at day 0 and day 3
post-treatment with tamoxifen. q Quantification of senescent cells upon
SCAT7 KD in A549 cells. The values are expressed as percentage of the
whole cell population. Note that the data presented in b–h and j–q
represents mean values of three independent experiments and the
statistical significance was derived using a two-tailed unpaired
Student’s t-test. Data are plotted as mean ± SD (*p ≤ 0.05; **p = 0.01
– 0.001; ***p < 0.001)
We next focused on Caki-2, 786-O, and A549 stable KD cell lines to
elucidate the functional role of SCAT7 in apoptosis, cell viability,
cell migration and invasion. SCAT7 knockdown in Caki-2 and A549 cell
lines increased caspase 3/7 activity (Fig. [159]5d) and affected their
viability, migration, and invasion properties (Fig. [160]5e,f;
Supplementary Fig. [161]5i-m). The invasion capacity was severely
suppressed in Caki-2 and A549 knockdown cells, while the effect was
very limited in the case of 786-O knockdown cells. Additionally, we
investigated the anchorage-independent cell growth using soft agar
colony formation assay for Caki-2, 786-O, and A549 knockdown cell lines
10 days after incubation (Fig. [162]5g; Supplementary Fig. [163]5n).
The ability of knockdown cells to form anchorage-independent colonies
was drastically reduced, and accordingly, the total number of colonies,
as well as the area of each individual colony was decreased. In
accordance with its role in restricting the cellular proliferation upon
knockdown, overexpression of SCAT7 increased cell proliferation by
2.25, 2.08, and 1.9 fold in HeLa, Caki-2, and A549 cells, respectively
(Fig. [164]5h). However, we were unable to overexpress SCAT7 in the
786-O cell line due to its inherited resistance to plasmid
transfection. Collectively, our data demonstrate the critical role of
SCAT7 in regulating some of the most important cancer hallmarks in
multiple cancer cell lines.
Downregulation of SCAT7 induces cell senescence
Given that SCAT7 knockdown induces cell cycle perturbations and
deformed cell morphology (Supplementary Fig. [165]5o), we investigated
the contribution of SCAT7 in cell senescence. To this end, we used two
fibroblast cell lines; BJ and TIG3, immortalized with B-RAF
transformation. Transient downregulation of SCAT7 using three siRNAs
for 72 h induced senescence in both the cell lines as indicated by
β-galactosidase staining and upregulation of senescence markers like
p16, IL8, and p21 (Fig. [166]5i, j; Supplementary Fig. [167]5p). We
also observed a spontaneous decrease in the expression levels of SCAT7
during serial passaging of fibroblast cells mimicking the normal
physiological aging (Fig. [168]5k). The immortalized BJ-BRAF
fibroblasts express the activated form of mouse B-RAF (V600E) under the
control of estrogen receptor (ER:B-RAF)^[169]31. Thus, the addition of
4-hydroxytamoxifen (4-OHT) activates B-RAF and consequently
senescence-associated signaling pathways^[170]32,[171]33. We sought
then to assess the transcriptional activity of SCAT7 upon induction of
premature senescence. Towards this, we treated BJ-BRAF cells with
200 nM of 4-HT for 72 h and checked the transcriptional activation of
some senescence-associated markers (Supplementary Fig. [172]5q).
Interestingly, the onset of premature senescence resulted in
significant reduction of SCAT7 expression (Fig. [173]5l). On the other
hand, overexpression of SCAT7 in BJ-BRAF fibroblasts increased their
proliferation (Fig. [174]5m; Supplementary Fig. [175]5r) as well as
transcriptional activity of key senescence markers (Supplementary
Fig. [176]5s). Cells overexpressing SCAT7 were able to bypass the
tamoxifen-induced senescence, as indicated by less β-galactosidase
activity and transcriptional repression of the key senescence markers
p16, p15, and IL8 (Fig. [177]5n-p). We next tested the hypothesis that
SCAT7 silencing in cancer cell lines may promote senescence. Toward
this, we performed β-galactosidase staining on HeLa, A549 and Caki-2
cell lines upon SCAT7 downregulation. Downregulation of SCAT7 induced
the senescence phenotype in both HeLa and A549 cells while Caki-2 cells
remained unaffected (Fig. [178]5q). Therefore, our data demonstrate the
crucial role of SCAT7 in regulating cellular senescence, highlights the
crosstalk between cell proliferation, cell cycle progression, and
senescence.
SCAT7 regulates FGF/FGFR, PI3K/AKT, and Ras/MAPK pathways
To gain an insight into the SCAT7-mediated molecular pathways and
cancer-related processes, we performed RNA sequencing of HeLa, Caki-2,
and A549 cells upon SCAT7 downregulation (Supplementary Fig. [179]6a).
Subsequent functional enrichment analysis of RNA-seq data revealed that
the depletion of SCAT7 affected major signaling pathways and vital
biological processes (Fig. [180]6a-c; Supplementary Data [181]3). For
instance, FGF/FGFR and the downstream PI3K/AKT and Ras/MAPK pathways
were largely affected while cell proliferation, cell adhesion, cellular
senescence, cell migration, and apoptotic processes were also
perturbed. We validated some of the dysregulated genes at the
transcriptional and translational levels (Fig. [182]6d and
Supplementary Fig. [183]6b) in the SCAT7 downregulated cells. We
detected a reduction in FGFR, p-AKT, and p-ERK1/2 levels in the SCAT7
downregulated HeLa, Caki-2, and A549 cells. We also investigated the
status of some of the well-known cell cycle master regulators; such as
CCND1, CDK4, RB, and Phospho-Ser795 RB (p-RB), in SCAT7 depleted cells
(Fig. [184]6e). CCND1 was downregulated at the transcriptional and
translational level upon SCAT7 knockdown while p-RB, but not RB, was
downregulated at the protein level. Conversely, the CDK4 expression was
neither affected at the transcriptional nor the translational levels.
Since SCAT7 overlaps two neighboring protein-coding genes ELF3 and
RNPEP, we investigated the effect of its downregulation on the
neighboring genes. SCAT7 depletion, using different siRNAs, shRNAs and
LNAs targeting the unique non-overlapping exon of SCAT7, did not affect
the expression of neighboring genes in HeLa and Caki-2 cell lines.
However, we found reduced ELF3 expression only in A549 cells
(Supplementary Data [185]3).
Fig. 6.
[186]Fig. 6
[187]Open in a new tab
SCAT7 regulates cell cycle progression and cell proliferation via FGF
signaling. a–c Heatmaps showing upregulated and downregulated genes
with corresponding molecular pathways and biological processes upon
silencing of SCAT7 in HeLa (a), Caki-2 (b), and A549 (c) cell lines. d
Western blot showing the proteins levels of FGFR2, FGFR3, AKT, Ser 473
Phospho-AKT (p-AKT S473), ERK1/2, and Phoshpo-ERK1/2 (p-ERK 1/2) upon
silencing of SCAT7 in HeLa, Caki-2, and A549 cell lines. FGFR4 protein
levels were only investigated in A549 cells. e Real-time qPCR
validation (left panel) and Western blot (right panel) showing a
significant reduction in the expression levels of CCND1 but not CDK4. f
Real-time qPCR validation of the expression levels of SCAT7 and its
targets FGFR2, FGFR3, and FGFR4 in HeLa, Caki-2, and A549 cells upon
SCAT7 KD with two independent siRNAs or shRNAs. g Expression of SCAT7
and its target FGFR2 in HeLa and Caki-2 cells overexpressing SCAT7.
Data are shown as relative fold change normalized to GAPDH. h MTT assay
in HeLa and Caki-2 cells, after transfection with siRNAs targeting
FGFR2, and A549 cells, transfected with esiRNA to silence FGFR3. i Cell
cycle profiles of HeLa and Caki-2 cells transfected with siRNAs
targeting FGFR2 (left and middle). The right panel shows the cell cycle
profile of A549 cells transfected with FGFR3 esiRNA. Note that the data
presented in e–i represent mean values of three independent experiments
and the statistical significance was derived using a two-tailed
unpaired Student’s t-test. Data are plotted as mean ± SD (*p ≤ 0.05;
**p = 0.01 – 0.001; ***p < 0.001)
Based on RNA-seq analysis and subsequent validations across different
cell lines, we hypothesized that SCAT7 modulates some of the cancer
hallmarks through the regulation of different FGF/FGFR members. For
instance, SCAT7 knockdown affected the mRNA levels of FGFR2, FGFR3,
FGF7, and FGF21 in HeLa cells while only FGFR2 was downregulated in the
Caki-2 and 786-O cells (Fig. [188]6f; Supplementary Fig. [189]6c).
Similarly, FGFR3 and FGFR4 were downregulated upon SCAT7 depletion in
A549 cells (Fig. [190]6f). Conversely, overexpression of SCAT7 restored
the expression of FGFR2 in HeLa and Caki-2 cells (Fig. [191]6g).
Moreover, the expression pattern of different FGFR members matched
SCAT7 expression during cell cycle progression in the investigated cell
lines (Supplementary Fig. [192]6d) and in tumor tissues derived from
LUAD, KIRC, and UCEC (Supplementary Data [193]3). For instance, in HeLa
cells, SCAT7, FGFR2, and FGFR3 are expressed early in the S-phase. In
the case of A549 cells, SCAT7, FGFR3, and FGFR4 demonstrate a higher
expression at G2 phase. To test our hypothesis that SCAT7 executes its
actions via FGF/FGFR signaling, FGFR2 was depleted in HeLa and Caki-2
cells using two siRNAs, and FGFR3 and FGFR4, separately, using esiRNAs
in A549 cells (Supplementary Fig. [194]6e). Interestingly, the effects
of the FGFR2 depletion on cell cycle, cell proliferation, and vitality
phenocopied the effects of the SCAT7 depletion in the HeLa and Caki-2
cells (Fig. [195]6h, i; Supplementary Fig. [196]6f). Furthermore,
SCAT7-induced cell proliferation was attenuated when FGFR2 was
silenced; indicating that SCAT7-induced cell proliferation is, in part,
carried out by the FGF/FGFR2 signaling (Supplementary Fig. [197]6g).
Though downregulation of both FGFR3 and FGFR4 in A459 cells led to a
decrease in cell proliferation, only FGFR3 downregulation affected cell
cycle progression in A549 cells (Fig. [198]6h, i; Supplementary
Fig. [199]6h, i). Collectively, our data strongly suggest the
involvement of SCAT7 in the modulation of cellular homeostasis and cell
cycle progression through the regulation of the FGF/FGFR and the
downstream PI3K/AKT and Ras/MAPK signaling pathways.
SCAT7/hnRNPK/YBX1 complex regulates cancer cell hallmarks
We next studied the mechanisms by which SCAT7 regulates cancer
progression via FGF/FGFR signaling in HeLa cells. We first determined
the subcellular distribution of SCAT7 and found it to be enriched in
the nucleoplasmic and chromatin compartments (Supplementary
Fig. [200]7a). Then, we performed chromatin oligo-affinity
precipitation (ChOP) in UV crosslinked cells using biotinylated
oligonucleotides to pull-down SCAT7 and its interacting proteins. Using
two independent biological replicates, we identified 96 proteins that
were specifically enriched in both replicates of SCAT7 ChOP
(Fig. [201]7a; Supplementary Data [202]3). We selected two proteins,
hnRNPK and YBX1, well-known for functions related to transcriptional
regulation and cell cycle progression, for detailed mechanistic
investigations^[203]34,[204]35. We validated the interaction of hnRNPK
and YBX1 proteins with SCAT7 using RNA immunoprecipitation (RIP) assay
(Fig. [205]7b) and ChOP pull-down followed by Western blot
(Fig. [206]7c).
Fig. 7.
[207]Fig. 7
[208]Open in a new tab
SCAT7 interacts with hnRNPK and YBX1 to regulate cell proliferation and
cell cycle progression. a Venn diagram showing SCAT7 interacting
proteins in HeLa cells identified using ChOP-MS in two independent
biological replicates. b RIP using hnRNPK or YBX1 antibody followed by
qPCR for SCAT7. c Validation of SCAT7 interaction with hnRNPK and YBX1
by ChOP followed by immunoblotting in HeLa cells. LacZ and SCAT7
reverse biotinylated probes were used as negative controls. d MTT in
HeLa cells after transfection with two siRNAs targeting hnRNPK or YBX1.
e Cell cycle analysis upon hnRNPK and YBX1 KD in HeLa cells. f FGFR2
expression by real-time qPCR in hnRNPK and YBX1 KD HeLa cells. g
Western blot of FGFR2, FGFR3, AKT, Ser 473 Phospho-AKT (p-AKT S473),
ERK1/2, and Phoshpo-ERK1/2 (p-ERK 1/2) in hnRNPK and YBX1 KD HeLa
cells. h, i ChOP followed by qPCR for SCAT7 enrichment at FGFR2 (h) and
FGFR3 (i) promoters in HeLa cells. Four primer pairs were used to
assess the occupancy at every 250 bp upstream of FGFR2 and FGFR3 TSS.
j, k ChIP using hnRNPK or YBX1 antibody followed by qPCR for hnRNPK and
YBX1 occupancy at FGFR2 (j) and FGFR3 (k) promoters in control and
SCAT7 KD HeLa cells. l Interaction of SCAT7 with hnRNPK and YBX1 by
SCAT7 ChOP followed by immunoblotting in A549 cells. m ChOP followed by
qPCR for SCAT7 enrichment at FGFR3 promoter in A549 cells. n hnRNPK or
YBX1 ChIP followed by qPCR depicting the occupancy of hnRNPK and YBX1
at the FGFR3 promoter in control and SCAT7 KD A549 cells. o SCAT7 ChOP
followed by immunoblotting with hnRNPK or YBX1 antibody in Caki-2
cells. p ChOP followed by qPCR quantification of SCAT7 enrichment at
FGFR2 promoter in Caki-2 cells. q hnRNPK ChIP followed by qPCR
depicting the occupancy of hnRNPK at FGFR2 promoter in control and
SCAT7 KD Caki-2 cells. Note that the data presented in b, d–f, h–k, m,
n, p, and q represent mean values of three independent experiments and
the statistical significance was derived using a two-tailed unpaired
Student’s t-test. Data are plotted as mean ± SD (*p ≤ 0.05; **p = 0.01
– 0.001; ***p < 0.001)
Next, we investigated the role of hnRNPK and YBX1 in the regulation of
cell proliferation, cell cycle progression, FGFR2, and FGFR3
expression, and pathways downstream to FGFRs in HeLa cells. Transient
knockdown of hnRNPK or YBX1 by siRNAs (Supplementary Fig. [209]7b) led
to a significant decrease in cell proliferation (Fig. [210]7d) and cell
cycle arrest at the G1 phase (Fig. [211]7e). Also, FGFR2 and FGFR3
expression was substantially downregulated at the mRNA (Fig. [212]7f)
and protein levels (Fig. [213]7g) in hnRNPK or YBX1 knockdown cells.
The activities of the MAPK and AKT pathways downstream to FGFRs were
also reduced upon hnRNPK or YBX1 knockdown (Fig. [214]7g). To
understand the mechanism of FGFR2 and FGFR3 regulation by
SCAT7/hnRNPK/YBX1 ribonucleoprotein (RNP) complex, we first checked the
interaction of SCAT7/hnRNPK/YBX1 complex with mRNA and the proximal
promoters of FGFR2 and FGFR3 genes. In our RIP assays with hnRNPK and
YBX1, we failed to detect FGFR2 or FGFR3 mRNAs, indicating that SCAT7
does not modulate FGFR2 and FGFR3 mRNA levels post-transcriptionally
(Supplementary Fig. [215]7c). We then assessed the occupancy of SCAT7
over the FGFR2 and FGFR3 promoter regions using ChOP-qPCR
(Fig. [216]7h, i). Our analysis revealed a significant enrichment of
SCAT7 at the FGFR2 (−250 to −750 bp relative to TSS (Fig. [217]7h) and
FGFR3 (−750 bp to −1 kb relative to TSS) (Fig. [218]7i) promoters. This
observation led us to check whether SCAT7 mediates the transcriptional
activation of FGFR2 and FGFR3 via recruiting hnRNPK and YBX1 to the
FGFR2 and FGFR3 promoters. We detected specific binding of hnRNPK and
YBX1, as assessed by ChIP assay, at the FGFR2 promoter (−250 to −500 bp
relative to TSS) and this binding was substantially reduced upon SCAT7
downregulation (Fig. [219]7j). Similarly, hnRNPK and YBX1 occupied the
−500 bp to −1 kb promoter region of FGFR3 and their binding was
affected specifically at the −750 bp to −1 kb promoter region following
SCAT7 downregulation (Fig. [220]7k).
Further, we investigated the role of SCAT7/hnRNPK/YBX1 complex in A549
and Caki-2 cells for transcriptional regulation of FGFR3 and FGFR2,
respectively. We performed SCAT7 ChOP pull-downs in A549 (Fig. [221]7l)
and Caki-2 (Fig. [222]7o) cells followed by Western blotting.
ChOP-Western indicated a specific interaction of SCAT7 with both hnRNPK
and YBX1 proteins in A549 cells (Fig. [223]7l), but only with hnRNPK in
Caki-2 cells (Fig. [224]7o). ChOP-qPCR assays in A549 and Caki-2 cells
revealed SCAT7 occupancy at the promoter regions of FGFR3 (−750 bp to
−1 kb) and FGFR2 (−250 bp to −500 bp), respectively (Fig. [225]7m, p).
Similar to our observations in HeLa cells, we detected the occupancy of
hnRNPK and YBX1 at the −750 bp to −1 kb FGFR3 promoter region in A549
cells and their occupancy was reduced upon SCAT7 downregulation
(Fig. [226]7n). In Caki-2 cell line, hnRNPK was enriched at the −250 to
−500 bp FGFR2 promoter region and its occupancy was affected upon SCAT7
downregulation (Fig. [227]7q). In addition, we observed a decrease in
the RNA Polymerase II occupancy over the coding region of FGFRs in
SCAT7 depleted cells (Supplementary Fig. [228]7d). Taken together,
these observations indicate that SCAT7/hnRNPK/YBX1 RNP plays a crucial
role in the transcriptional activation of FGFR2 and/or FGFR3 in
different cancer models.
SCAT7 as potential therapeutic target in cancer treatment
Our in vitro data, as well as the mechanistic studies, clearly
demonstrate the oncogenic nature of SCAT7 and its crucial role in
promoting cancer-associated signaling pathways. Hence, we wanted to
elucidate the role of SCAT7 in malignant tumorigenesis in vivo. To this
end, we generated two different xenograft models engrafted with either
786-O or A549 SCAT7 stable knockdown cells. Eight weeks
post-engraftment, both SCAT7 depleted 786-O and A549 xenografts showed
a significant decrease in growth parameters compared to control
xenografts. Ki67 immunostaining of the dissected tumors confirmed the
restricted proliferation capacity of SCAT7 knockdown cells in vivo
(Fig. [229]8a, b; Supplementary Fig. [230]8a, b).
Fig. 8.
[231]Fig. 8
[232]Open in a new tab
SCAT7 is a potential therapeutic target for different tumor types. a
Tumor growth in Balb/c nude mice 8 weeks after subcutaneous inoculation
of 1 × 10^6 Csh or SCAT7-sh2 786-O cells (n = 8 each group). b Tumors
from Balb/c nude mice after subcutaneous injection of 1 × 10^6 Csh or
SCAT7-sh1 or SCAT7-sh2 A549 cells for 8 weeks (n = 6 for each group).
In a and b tumor volumes (cm^3) are expressed as mean ± SD, compared
with Csh. c Tumor growth inhibition (TGI) for A549 subcutaneous Balb/c
nude xenografts treated with 60 pmol of SCAT7 antisense
oligonucleotides, LNA-1 and LNA-2, respectively, for a total of four
injections (n = 5 for each group). d Average tumor growth of the
xenografts treated with control or SCAT7 LNAs was calculated at each
injection as follows: Vtx-Vt0. e Real-time qPCR of SCAT7 in A549 tumors
collected after treatment with Ctrl-LNA, SCAT7-LNA1, and SCAT7-LNA2.
The red bar represents a tumor treated with LNA2, but no downregulation
was observed. Values are normalized to endogenous GAPDH. f Scatter plot
showing the correlation between SCAT7 expression in vivo and the tumor
volumes. The red dot represents the tumor that had no significant
downregulation of SCAT7 upon LNA treatment. g Immunohistochemistry
images of Ki67 staining and TUNEL assay for A549 xenografts treated
with Ctrl-LNA, SCAT7-LNA-1, and SCAT7-LNA-2 (blue: DAPI, green:
TUNEL-GFP). h Patient-derived xenograft (PDX) of NSG mice models
treated with 100 pmol of Ctrl-LNA or SCAT7-LNA1 for a total of five
injections (n = 6 for each group). i Average growth of tumor volumes of
the PDX models treated with control or SCAT7 LNAs. j, k Model depicting
the functional involvement of SCAT7 in regulating cancer hallmarks (j)
and mechanism of FGF/FGFR signaling regulation by SCAT7 (k). The
statistical significance shown in b–e and i was derived using a
two-tailed unpaired Student’s t-test. Data are plotted as mean ± SD
(*p ≤ 0.05; **p = 0.01 – 0.001; ***p < 0.001)
We next tested the hypothesis that SCAT7 can serve as a target for
therapeutic intervention. Towards this, we engrafted A549 cells
subcutaneously into nude mice. Six weeks post-engraftment, we applied a
treatment regimen based on the subcutaneous injection of two
independent LNA antisense oligonucleotides targeting SCAT7, twice a
week (Methods). We measured the tumor volumes after a course of four
injections in two independent experiments and observed 40–50% tumor
growth inhibition (TGI) in SCAT7 LNA groups compared to the scrambled
LNA control group (Fig. [233]8c, d; Supplementary Fig. [234]8c). We
also monitored the weight of the mice during the tumor development and
post-injections in both experiments to score for any weight loss
induced by the LNA treatment. Additionally, we measured the weights and
sizes of liver, spleen, and kidneys to assess the cytotoxicity of the
treatment and found no difference between the two groups (Supplementary
Data [235]3). Interestingly, the expression levels of SCAT7 in
xenografts correlated with tumor volumes (Fig. [236]8e, f) and tumor
growth reduction was observed only in the tumors that had an efficient
SCAT7 downregulation. Then, we checked the expression levels of FGFR3,
FGFR4, and p21 (Supplementary Fig. [237]8d). In line with the in vitro
data, FGFR3 and FGFR4 downregulation and p21 upregulation was detected
in tumors treated with SCAT7 LNAs. Ki67 immunostaining, and TUNEL
staining of the dissected tumors revealed the potent effect of SCAT7
targeting on cancer malignancy in vivo (Fig. [238]8g). Furthermore, we
implemented SCAT7 LNA-dependent therapeutic intervention over a period
of 15 days on a lung metastatic patient-derived xenograft (PDX) mouse
model bearing an oncogenic mutated form of KRAS (Methods). PDX mice
models injected with SCAT7 LNA exhibited a significant reduction in the
growth rate as well as the volumes (~46%) of the engrafted tumors
(Fig. [239]8h, i; Supplementary Fig. [240]8e). These results
collectively suggest that SCAT7 is an oncogenic lncRNA and it can be
used as a possible therapeutic target in the treatment of different
cancers.
Discussion
The main aim of the current investigation is based on the hypothesis
that temporally expressed transcripts across S-phase may harbor crucial
functions in the control of cell cycle progression and that their
deregulation may underlie tumor development and progression. In the
recent past, compared to protein-coding mRNAs, there is a growing
appreciation for investigating the role of lncRNAs in cancer
development and progression given their surprising roles in
spatiotemporal gene expression. Through characterizing the dynamic
transcriptional changes across S-phase in real time, we identified 1145
lncRNAs showing S-phase-specific temporal expression patterns.
Enrichment of cancer-associated pathways in the GO analyses of
neighboring protein-coding genes of S-phase lncRNAs, and their
differential expression across Pan-Cancer data sets, indicate that
S-phase lncRNAs may be strongly associated with cancer development. One
of the interesting aspects of S-phase lncRNAs’ differential expression
analysis across TCGA data sets is that nearly 60% of them show
differential expression at least in two cancer types. To confirm our
hypothesis, we have selected top eight S-phase lncRNAs that show
differential expression across multiple cancers and investigated their
role in cancer progression via analyzing the effect of their
downregulation on cancer cell hallmarks in various cancer models.
Confirming our hypothesis, downregulation or overexpression of SCATs
affected cancer cell hallmarks such as cell cycle progression, cell
proliferation, apoptosis, cellular senescence, and cell
invasion/migration, indicating that our cell cycle-based functional
screen identified potential oncogenic drivers.
DNA methylation analyses of S-phase lncRNAs revealed that DNA
methylation may underlie the differential expression of S-phase lncRNAs
between tumor and normal tissue pairs. For instance, SCAT3 is
upregulated in six types of cancers (Supplementary Data [241]2) and
this is consistent with its promoter’s hypomethylation in several
cancers (HNSC, KIRP, and LIHC). Conversely, our analysis identified a
number of S-phase lncRNAs showing a hypermethylation pattern that
correlates with their diminished expression in respective cancers.
Therefore, our DNA methylation investigations of S-phase lncRNAs
reflect the role of epigenetic alterations in modulating their
transcriptional activities during tumorigenesis.
Another important aspect of the current study is the comprehensive
investigation of the clinical relevance of the S-phase lncRNAs across
TCGA data sets. Our analysis identified 633 S-phase lncRNAs that act as
independent biomarkers with high prediction accuracy (Supplementary
Fig. [242]2b). Strikingly, our clinical investigation indicated that
KIRC, COAD, LIHC, and HNSC harbor more than 50 S-phase lncRNAs as
potential independent prognostic markers. For instance, SCAT8 appeared
to be the top prognostic indicator in our studies with the higher
hazard ratio in our multivariate models and it also interferes with
cancer cell hallmarks, indicating that it may be an oncogenic driver in
multiple cancers. Further, our clinical investigations identified SCAT5
as a common independent prognostic biomarker in kidney cancers KIRP,
KIRC, and KICH. Although SCAT5 was first identified in lung cancer cell
patients in response to cigarette smoking^[243]36, its role in the
etiology of different kidney cancers has not been investigated.
Notably, its strong prognostic significance coupled with effect of its
downregulation on the cancer cell hallmarks in KIRC cell lines,
indicate that SCAT5 could be a potential therapeutic target in the
treatment of kidney cancers. Similarly, SCAT1, which is upregulated in
10 different cancers, shows a functional involvement and independent
prognostic capacity in four cancers including LUAD and LUSC. Thus, our
functional and clinical investigations on SCAT8, SCAT5, and SCAT1,
suggests that our cell cycle-based functional screen indeed has
identified potential lncRNA-based cancer drivers. Importantly, our
analysis indicates that clinically relevant S-phase lncRNAs can be
considered as independent prognostic biomarkers with a strong potential
to be used in the clinical setting for better risk stratification and
prediction of clinical outcome.
For a further understanding of the mode of action of S-phase-enriched
lncRNAs in tumorigenesis, we performed a detailed mechanistic
investigation using SCAT7 as a model. The consistency of the obtained
phenotypic effects upon SCAT7 knockdown in different model systems
indicates a conserved functional interaction between SCAT7 and its
downstream target genes to ensure cellular homeostasis. The crosstalk
between cell cycle progression, apoptosis, and cellular senescence has
been firmly established in the maintenance of cellular
homeostasis^[244]37 and our data clearly demonstrated that the
silencing of SCAT7 strongly interferes with these interconnected
biological processes, thereby affecting cellular homeostasis. Upon
SCAT7 knockdown different types of proliferative cell lines exhibit the
characteristic features of cellular senescence including
β-galactosidase secretion, cell cycle arrest, and induction of
different tumor suppressor genes^[245]38 (p21, p16, and p15). This is
consistent with the enrichment of biological processes such as cell
cycle, apoptosis, cell proliferation, and cell senescence. Moreover,
SCAT7 depletion in multiple cell lines show accumulation of cells in G1
phase and S-phase progression defects. The drastic reduction in the
EdU+ve cells upon SCAT7 depletion is also a clear indication of the
diminished S-phase within DNA-replicating cells. Reduced G1 to S phase
progression in SCAT7 depleted cells may be due to CCND1 downregulation
and RB hypophosphorylation. CCND1 downregulation could be the result of
inhibitory effects of FOXO1/3 factors due to the reduced AKT
activity^[246]39,[247]40. Though these observations support the role of
SCAT7 in G1 to S phase progression, more work is needed to ascertain
the functions of SCAT7 in S-phase progression. Thus, our functional
investigations unequivocally implicate SCAT7 in the cellular
homeostasis through regulating the cancer cell hallmarks
(Fig. [248]8j).
The dissection of the regulatory pathways mediated by the action of
SCAT7 indicated its crucial involvement in regulating pivotal signaling
pathways across multiple cancer models. lncRNAs have been reported to
regulate signaling pathways like Wnt^[249]41,[250]42, Notch^[251]43,
PI3K/AKT^[252]44, and RAS/MAPK^[253]45. However, the regulation of
FGF/FGFR signaling by lncRNAs has not been explored mechanistically in
great detail. Given that several FGF/FGFR members were deregulated upon
SCAT7 knockdown in multiple cancer models, we hypothesized a genuine
connection between SCAT7 and FGF signaling in the context of cancer.
The functional conservation of SCAT7-hnRNPK-YBX1 RNP complex in
different cancer models presents a mechanistic model of FGF/FGFR
regulation. The recruitment of the SCAT7-hnRNPK-YBX1 RNP complex at the
promoter regions of FGFR2 and FGFR3 promotes transcriptional activation
of the FGF/FGFR pathway, leading to sustained cell proliferation via
PI3K/AKT and Ras/MAPK pathways (Fig. [254]8k). It is pertinent to note
that the identification of hnRNPK and YBX1 as the interacting proteins
of SCAT7 fits well in the current understanding of the functional roles
of hnRNPK and YBX1 in the regulation of cell cycle progression, gene
expression, and cell signaling^[255]34,[256]35,[257]46. However, it
will be interesting to scrutinize whether cell identity, as shown in
the KIRC model, affects SCAT7/RNPs interactions in a lineage-specific
manner. Nevertheless, lineage-specific interactions between lncRNAs and
RNPs have been shown to be involved in developmentally regulated
biological functions^[258]23,[259]47–[260]49.
The functional significance of FGF/FGFR members in normal development,
maintenance of stem cell properties, and senescence is fairly well
understood^[261]50. Regulation of FGF/FGFR members by SCAT7 in HeLa,
KIRC, and LUAD cancer models highlight the functional role of FGF
signaling in SCAT7-mediated tumor initiation and progression.
Consistent with our data, several investigations have implicated FGF
signaling in renal and lung cancers. For instance, a SNP in FGFR2 was
shown to affect the progression-free survival alone in the metastatic
KIRC patients undergoing anti-VEGF-targeted therapy^[262]51. Also, a
missense mutation in FGFR2 was found to drive a durable response to
nucleolin-targeted therapy in metastatic KIRC^[263]52. Despite the fact
that various FGFR members harbor activating mutations in lung cancer
patients and confer acquired resistance to tyrosine kinase inhibitors
(TKIs)^[264]53,[265]54, only a handful of studies have addressed the
role of these mutations in LUAD tumorogenesis. Tchaicha et al.
generated the first genetically engineered lung cancer mouse model
harboring an FGFR mutation in p53 null background^[266]55. The
engineered mouse model showed more than 50% tumor regression when
treated with a pan-FGFR inhibitor. More recently, Manchado et al.
reported a combinatorial approach including FGFR1 inhibitor to overcome
the adaptive resistance to MEK inhibitor in KRAS-mutant LUAD^[267]56.
Considering the functional nexus between SCAT7 and FGF signaling,
targeting SCAT7 alone is sufficient to inhibit tumor progression via
repressing different members of the FGF/FGFR pathway. Supporting this,
SCAT7 LNA-based treatment used in this study established a possible
therapeutic intervention regimen for multiple cancers in vivo. Based on
these observations, we suggest that a combinatorial treatment strategy
involving SCAT7 repression alongside treatment with potent FGFRs
inhibitors or TKIs will hold promise for lncRNA-based therapeutics.
Collectively, we provide a comprehensive list of lncRNA-based oncogenic
drivers with potential prognostic value. More importantly, this
systematically analyzed functional and clinically relevant lncRNAs can
serve as a resource for delineating the functional link between lncRNA
and tumor development and progression.
Methods
Cell lines and cell synchronization
The adherent Caki-2, 786-O, and HepG2 cell lines were purchased from
CLS-GmbH (Germany). The LUAD cell lines A549 and H2228 were kindly
provided by Bengt Hallberg’s lab at the department of Medical
Biochemistry and Cell Biology, University of Gothenburg. HeLa and MCF-7
cell lines were routinely maintained in our lab. The immortalized human
fibroblasts cell lines, BJ-BRAF and TiG3-BRAF, were kindly provided by
Andres Lund’s lab (Biotech Research and Innovation Center, University
of Copenhagen). We cultured Caki-2, 786-O, and H2228 cell lines in
RPMI-1640 medium (Gibco, Life Technologies, USA). A549, BJ-BRAF,
TiG3-BRAF, and MCF-7 cell lines were maintained in DMEM medium (Gibco,
Life Technologies; USA). HeLa and HepG2 cell lines were maintained in
MEM medium (Gibco, Life Technologies; USA). All media were supplemented
with 2 mM l-glutamine, 1× penicillin-streptomycin antibiotic, and 10%
fetal bovine serum. All cell lines were tested negative for Mycoplasma
contamination. For RT-qPCR validation, HeLa cells were synchronized
following the addition of 2 mM of thymidine into a fresh medium for
10 h then aspirating the medium and adding hydroxyurea to a final
concentration of 1 mM overnight. The synchronized cells were collected
at 2, 3.5, 5, and 9 h representing T1, T2, T3, and T4, respectively.
For serum starvation, HeLa, A549, and Caki-2 cells were cultured in
serum-free media for 36–44 h. The synchronized HeLa cells were
collected at the indicated time points while Caki-2 cells were
harvested at 4, 8, and 20 h representing T1, T2 and T3, respectively.
In A549 cells, T1, T2, and T3 synchronized cells were harvested at 4,
6, and 18 h, respectively.
Nascent RNA capture assay
To capture nascent RNA and total RNA from different stages of S-Phase,
on day 1 HeLa cells were plated at 500,000 cells per F75 plate, to
ensure cell confluency of around 35% during synchronization. On day 2
cells were incubated for 10 h in medium supplemented with 2 mM
thymidine (Sigma). After 10 h, cells were washed with PBS and incubated
with medium supplemented with 1 mM hydroxyurea (Sigma) for 14 h. The
synchronized cells are now just at the beginning of S-phase with a
confluency of 70%, which allowing them to resume dividing upon release
from cell cycle block without suffering from contact inhibition. After
a brief PBS wash, medium is then changed back to normal, and the cells
are allowed to progress through S-phase in a synchronized manner. The
time at which the block is released is thereafter termed “T0”. The
labeling of nascent RNA was carried out for 2-h periods at different
stages of S-phase. At the beginning of the labeling period, media was
supplemented with EtU (Invitrogen) to a final concentration of 1 mM,
and cells were harvested 2 h later. The labeling periods were defined
as follows: T0h–T2h (beginning of S-phase), T1.5h–T3.5 h (middle of
S-phase), and T3h–T5h (end of S-phase). This labeling protocol ensured
that, for all three S-phase samples, the labeled nascent transcripts
would represent only 2 h of transcription, in order to provide a
detailed and accurate picture of the timing of transcriptional events
occurring throughout S-phase. For unlabeled total RNA samples, we
followed the same procedure but with medium without EtU supplement.
Cells from EtU labeled and unlabeled samples were harvested at the end
of their respective 2 h labeling period, and RNA were extracted with
Tri reagent (Ambion). We performed DNA digestion with RQ1 DNase I
(Promega) for 1 h at 37 °C, and re-extracted RNA with Tri reagent. An
aliquot of 10 µg RNA was re-suspended in low volume and rRNA depletion
was carried out with Ribominus kit (Invitrogen). From each S-phase
sample, 2, 3 µg of rRNA-depleted RNA was biotinylated with Click-It
Nascent RNA Capture kit (Invitrogen) following manufacturer’s protocol.
The biotinylated RNA was captured using streptavidin magnetic beads.
The captured biotinylated RNA was eluted by incubating the beads in
200 µl buffer containing 2 mM biotin, 1 M NaCl, 50 mM MOPS, 5 mM EDTA,
2 M 2β-Mercaptoethanol, pH 7.4 for 3 min at 95 °C. Supernatant
containing eluted RNA was recovered immediately after heating and the
RNA was precipitated in 30 µl 3 M sodium acetate (pH 5.2), 1 µl
glycoblue (Invitrogen), 750 µl 100% ethanol. RNA was then resuspended
in nuclease-free water, dosed on a Qubit fluorometer (Invitrogen), and
RNA size profile was assessed on Bioanalyzer (Agilent) with RNA pico
6000 kit and sent for library construction and sequencing on a Solid
platform at Uppsala Genome Center. For unlabeled RNA samples, total RNA
was extracted and subjected to rRNA depletion, and used for
RNA-sequencing.
Knockdown of target genes and cloning
We performed transient transfection using siRNA, esiRNA, and
antisense LNA^™GapmeR molecules. Scramble siRNA, custom-made siRNAs,
and pre-designed esiRNAs were designed and synthesized by
Sigma-Aldrich. We obtained both in vivo and in vitro grade negative
control and custom-made LNA^™GapmeR molecules from Exiqon. Transfection
was carried out in standard 24-well plates using Lipofectamin® RNAiMAX
transfection reagent (Invitrogen, California) according to the
manufacturer’s instructions with a final concentration of 35 pmol and
20 pmol/well of siRNA and LNA, respectively. The transfections were
performed in three biological replicates and the efficiency of KD was
confirmed by RT-qPCR. We generated stable cell lines using Lentifect™
Purified shRNA lentivirus particles designed and synthesized by
GeneCopoeia™ to target SCAT7 or scramble negative control. The
transduction efficiency was visualized by the integrated GFP reporter
gene. The stable 786-O, Caki-2, and A549 KD cells were selected using
2 µg/ml, 3 µg/ml, and 2.5 µg/ml of puromycin, respectively. All
custom-made siRNAs, LNAs, and shRNAs particles were designed to target
the unique non-overlapping exons of the respective transcript. For
SCAT7 overexpression, the SCAT7 fragment was cloned into pGEM-T Easy
vector and the correct orientation was verified by sequencing. The
confirmed clone was digested using NotI and HindIII enzymes and
sub-cloned into the mammalian overexpression vector pcDNA3.1. HeLa,
Caki-2, and A549 cells were transfected with either 1 µg of
pcDNA3.1-SCAT7 vector or pcDNA3.1 empty vector using Lipofectamin® 2000
transfection reagent. The RNA levels were measured with RT-qPCR. The
sequences of siRNAs, shRNAs, LNAs, primers, and antibodies are listed
in Supplementary Data [268]4.
Flow cytometry, cell cycle, and Click-iT EdU assay
We assessed the cell cycle profiles of transient KD cells and control
cells 48 h post-transfection. The media were aspirated, and the cells
were washed with PBS, trypsinized, pelleted by centrifugation, washed
twice with cold PBS, fixed with ice-cold 70% ethanol, and stored at
−20 °C for at least 2 h. The fixed cells were re-collected by
centrifugation, re-suspended in PBS, and kept for 30 min at 37 °C.
Then, cells were collected and stained with PI solution containing 1%
RNase A in PBS and kept at 4 °C for at least 4 h. The PI-stained cells
were assayed using Eclipse single-cell flow cytometry system ec800 and
data were analyzed with the manufacturer’s software. The cell cycle
profiling was validated on another system using NucleoCounter NC-3000
platform (Chemometec, Denmark). The fixed cells were stained with DAPI
solution provided by the manufacturer and analyzed according to
manufacturer’s instructions. All KD experiments were assayed three
times independently and statistical significance was derived using
two-sided unpaired Student’s t-test. The EdU incorporation assay was
performed according to the manufacturer’s protocol using the Click-iT™
EdU Alexa Fluor™ 488 Imaging Kit and the green fluorescence wad
detected using the EVOS FL Auto Cell Imaging System.
Northern blotting analysis
Total RNA was extracted from wild-type A549 and SCAT7-KD cells and an
equal amount of 10 µg of each sample was loaded into 1% formaldehyde
agarose gel alongside 1.0 µg of 0.5–10 kb RNA ladder (Invitrogen). We
used 1 × MOPS-formaldehyde as a running buffer in RNase-free
conditions. Following the gel electrophoresis, the wet transfer was
performed overnight using 20 × SSC as a transfer buffer at room
temperature. After the transfer, the membrane was fixed under UV then
it was cut and immediately incubated 3 h with rapid-hyb hybridation
buffer (GE Healthcare Life Sciences) for pre-hybridization at 42 °C.
The radioactive probes were prepared freshly for SCAT7 and RNA ladder.
For SCAT7, we utilized the T4 PNK enzyme (NEB) to label the SCAT7 short
probes (the same probes used in ChOP experiment) using γ^32-P ATP
(PerkinElmer). For the RNA ladder probes, we used the RNA ladder as a
template to synthesis the complementary probes using labeled α-dCTP in
a reverse transcription reaction. The probes were purified using G25
columns (GE Healthcare Life Sciences) then they were hybridized
separately with the corresponding membrane at 42 °C overnight. The
membrane was washed twice at room temperature with low stringent buffer
(2 × SSC and 0.1% SDS) for 10 min each followed by two washes of warm
high stringency buffer (0.1 × SSC and 0.1% SDS) at 55 °C for 15 min
each. The membranes were left for exposure overnight and scanned with
Fuji FLA7000 phosphoimager.
Overview of annotations and tools used in this study
The hg19 (GRCh37) genome version for alignment and transcript
annotation from GENCODE version 19 (equivalent Ensembl GRCh37)^[269]57
was used for processing RNA-sequencing samples. Color space reads from
SOLID sequencing platform were aligned using LifeScope, and
HISAT^[270]58 aligner was used for reads from Illumina sequencing
platform. The reads for gencode transcripts were quantified using
HTSeq-count^[271]59 with ‘intersection-strict’ mode.
Sequencing and alignment
For each S-phase time point, RNA from two independent experiments were
pooled into one sample and deep sequenced on SOLID sequencing platform
at Uppsala Genome Center. The alignment resulted in 18.5, 39.3, 21.3,
and 29 million mappable 75 bp reads for unsynchronized, S-phase T1,
S-phase T2, and S-phase T3 EtU-labeled samples, respectively. For
unlabeled samples, we obtained 19.6, 30, 29, and 27 million mappable
75 bp reads for unsynchronized, S-phase T1, S-phase T2, and S-phase T3
samples, respectively.
Transcript annotation and read quantification
The uniquely mapped reads were assigned to long noncoding transcripts
from Gencode v19 annotation and obtained 4039 and 3966 lncRNAs from EtU
labeled and unlabeled samples, respectively, which are expressed at
least at one time point of S-phase. The obtained reads were normalized
to their library sizes and transcript length (reads per kilobase of
transcript per million—RPKM normalization). The log-fold change values
were derived from obtained RPKM values by comparing individual
time-point against unsynchronized samples.
Time-series analysis to identify lncRNAs across S-phase
The lncRNAs having log-fold change greater than one in at least one
S-phase time-points over unsynchronized sample were taken for further
analysis. We obtained 1734 and 1674 lncRNAs in EtU labeled and
unlabeled samples respectively. The Short Time-series Expression Miner
(STEM) clustering which is specifically designed to handle short
time-series gene expression data was used to find the significant
temporal patterns in S-phase-derived lncRNAs. The significant temporal
expression patterns were obtained from expression profiles of S-phase
lncRNAs in three different time-points (2, 3.5, and 5 h) by assuming
unsynchronized sample as a base time point. There were 1145 (out of
1734) lncRNAs in EtU labeled and 937 (out of 1674) lncRNAs in unlabeled
samples, showed significant temporal expression patterns across S-phase
as in Fig. [272]1c and Supplementary Fig. [273]1a.
E2F1-bound EtU-labeled S-phase lncRNAs
E2F1 transcription factor ChIP-seq peaks were obtained for HeLa cell
line from ENCODE (ENCSR000EVJ) hg19 processed files. Using Homer
‘annotatePeaks.pl’, the E2F1 peaks were associated with promoter of the
transcripts from Gencode v19 annotation. Individual peaks are assigned
to the promoter if it is within ±2 kb to the TSS of a transcript.
Functional significance of nearby protein-coding genes
The nearby protein-coding genes to EtU labeled, unlabeled and common
S-phase lncRNAs were tested for their functional significance. We
observed most of the first hit to the S-phase lncRNAs are within 50 kb
distance. Protein-coding gene first hit within 50 kb window relative to
S-phase lncRNAs from EtU labeled and unlabeled samples were extracted
using BedTools ‘closest’ with parameters ‘-D ref’ and further used for
functional enrichment analysis. GeneSCF v1.1^[274]24 was used to
perform gene enrichment analysis. It is important for an enrichment
tool to be updated frequently to get more reliable results and avoid
misinterpretation of the data^[275]60. The major advantage of GeneSCF
is that it uses the updated or recent terms and updated genes from the
databases. GeneSCF is more reliable compared to most enrichment
analysis tools because it has real-time based enrichment analysis
feature. The code for GeneSCF is also regularly updated to accommodate
the changes from the source database like KEGG, Gene Ontology, and
Reactome. In this study, the terms are considered significant only if
it is enriched with p < 0.05 and with minimum of 10 genes involved in a
particular function. These genes were tested against two different
functional annotations, Gene Ontology biological process (GO-BP), and
KEGG pathways by assuming all protein-coding gene numbers from Gencode
v19 as background (20,345) or reference set.
Processing S-phase lncRNAs in multiple cancers from TCGA
We have aligned RNA-sequencing reads using HISAT from 16 cancer types
from TCGA. The read abundance was quantified for Gencode v19
transcripts using HTSeq-count in “intersection-strict” mode (options
--no-mixed --no-discordant --no-unal –known-splicesite-infile) for
obtaining the read counts per transcript. The obtained reads (counts)
were normalized to their library sizes and transcript length (RPKM
normalization). Using these normalized counts, the significance of
differential expression between normal samples and corresponding solid
tumor was obtained using Wilcoxon signed-rank non-parametric test and
corrected for multiple testing with Benjamini–Hochberg’s method (FDR).
The cell cycle-associated lncRNA to be considered as significantly
differentially expressed cancer-associated transcript, we used log-fold
change ± 2 and FDR < 1E–004 in at least one cancer type as criteria.
These cancer-associated lncRNAs were further subjected to clinical
analysis. The information on each cancer type and the number of tumor
and normal tissue samples processed for individual cancer type is
provided in Supplementary Data [276]2.
Processing S-phase lncRNAs for TCGA CpG methylation data
Processed differential methylation data was downloaded from
COSMIC^[277]28 repository for GRCh37 genome and version 74 transcript
annotation equivalent to Gencode v19 annotation. COSMIC analysis
predicted differentially methylated regions (DMRs) by comparing the
beta-values derived from TCGA data level 3 for tumor and matched normal
samples. The cancer types containing more than 19 normal samples were
considered for statistical analysis. The p-values for the comparisons
were obtained using Mann–Whitney test and corrected p-values using
family wise error rate (FWER). The significant DMRs were filtered on
the basis of FWER < 0.05. The above information for statistical
analysis was extracted from COSMIC repository.
The significant DMRs from COSMIC were assigned to promoter (−2000 bp
and +500 bp from TSS) and gene body (TSS + 550 to length of transcript)
regions of the cancer-associated lncRNAs from our study. We considered
any lncRNA as differentially methylated only if more than 10 patient
samples support the methylation status and also absence of any patients
which supports opposite methylation pattern for the same region. As an
example, the promoter region has to be considered as hypermethylated if
there were at least 10 patients supporting hypermethylation status but
no patients (null) show hypomethylation at the same promoter region or
other regions within the transcript. All presented hypo and
hypermethylation lncRNAs has FWER < 0.05 obtained with matched normal
and tumor comparison in respective cancer types. The number of tumor
samples used for the methylation analysis for each cancer type is
included in Supplementary Data [278]2 and the detailed description on
COSMIC differential methylation analysis is available at
“[279]http://cancer.sanger.ac.uk/cosmic/analyses” under “Methylation”
sub-heading.
Clinical investigation of S-phase lncRNAs in TCGA tumors
For clinical investigations, we have chosen 1145 lncRNAs that were
showing significant temporal expression in EtU-labeled samples. The
selected lncRNAs were dichotomized with respect to the expression
cut-off based on either mean, median, or quartiles and considered
whichever gave the best discrimination. Thereafter, overall survival
rate in patients above and below the selected cut-off levels, was
calculated according to the KM method, and the log-rank test was used
to assess differences in survival. We used survival ROC R package, in
which the time-dependent ROC is calculated using the KM estimator for
the expression cut-offs^[280]61. Points above the diagonal represent
good classification results (better than random), points below the line
poor results (worse than random). AUC < 0.5 was filtered to prevent
randomness in the discrimination of poor and good survival groups. To
further investigate the relation to survival, dichotomization and
continuous levels of S-phase lncRNAs were assessed in a Cox regression
model and hazard ratios calculated. Based on expression distribution,
Student’s t-test or Wilcoxon rank sum test was used to evaluate mean
expression of SCATs in relation to clinical parameters such as node (N0
vs. N1_N3), stage (Tumor stage 1 & 2 vs. Tumor stage 3 & 4), size
(Tumor size 1 & 2 vs. Tumor size 3 & 4), metastasis (M0 vs. M1), grade
(Tumor grade 1 & 2 vs. Tumor grade 3 & 4), and age ( < median age
vs. > median age). We constructed a multivariate Cox regression model
including clinical parameters and S-phase lncRNAs expression that were
significant in univariate analysis to determine the prognostic effect.
Brier score was used to assess the prediction error in the model. Pec
from the R package was used for Brier score prediction. All the
statistical analyses were performed using R package and p < 0.05 was
considered as significant. We considered the lncRNA as a potential
prognostic marker, if they showed significant survival difference in
log-rank test and had a statistically significant hazard value in coxph
regression analysis either in dichotomization or in continuous scale.
Survival analysis and coxph regression analyses were performed using
Survival package in R.
Sequencing and differential expression analysis
The reads were cleaned for adapter sequence using Trimmomatic (v 0.32)
and the alignment of cleaned reads was done with hg19 reference genome
using HISAT aligner (mode: --sensitive --qc-filter). We used Gencode
v19 annotation to quantify the read abundance using HTSeq-count. The
differential gene expression between knockdown (KD) and control cells
was analyzed using bioconductor package edgeR^[281]62. Genes were
tested for differential expression only if expression (CPM) is greater
than 1 in at least two samples of comparison groups. The significant
candidates were filtered with log-fold change > ± 1 and FDR ≤ 0.05.
Pathway enrichment analysis of deregulated genes
The significantly deregulated genes from siRNA knockdown samples were
tested for pathway enrichment using tool GeneSCF v1.1. The parameters
for GeneSCF were set to database Reactome with background genes 20,345
(all protein-coding genes from Gencode v19 annotation used in this
study). The enriched functions were filtered based on p < 0.05 and
minimum of 10 genes involved in corresponding process.
Proliferation and vitality assays
We assayed the proliferation capacity of transiently transfected cells
48 h post-transfection using CellTiter 96® Non-Radioactive
Proliferation Assay kit (Promega, USA) with some modifications to the
manufacturer's protocol. The media were aspirated and cells were washed
once with PBS, and 425 µl of fresh medium plus 75 µl of MTT dye were
added and incubated at 37 °C in dark for 4 h. Then, each reaction was
terminated using 500 µl of stop solution and the cells were kept
overnight in dark at 4 °C to solubilize the MTT dye. The dye intensity
was measured using microplate reader at 570 nm. Standard deviation (SD)
and statistical significance were derived from three independent
experiments. For assaying the proliferation capacity of stable KD cell
lines, we seeded the same number of control and KD cells and applied
the same protocol used in the transient transfection experiments. We
performed the vitality assay for transient KD cells and stable KD cell
lines using NucleoCounter NC-3000 platform. The cells were harvested
and stained with a mixture of VB-48™, PI, and acridine orange dyes
according to the manufacturer’s instructions. The results were viewed
and analyzed by the manufacturer’s software.
Apoptosis and senescence assays
We performed apoptosis assay for transiently transfected cells 48 h
post-silencing using Caspase-Glo® 3/7 Assay (Promega, USA) and measured
the luminescence according to the manufacturer’s instructions. For
stable KD cells, in addition to the Caspase-Glo® 3/7 Assay, we
performed fluorescent-based Guava Caspase 3/7 FAM Assay (Merk
Millipore, Germany) following the manufacturer’s instructions. We
analyzed the caspase 3/7 FAM activity using NucleoCounter NC-3000
platform. For detection of senescence in fibroblasts and HeLa cells, we
carried out transient transfections for 72 h. In case of A549 stable KD
cells, we seeded the cells at less confluency for 72 h. We then
detected the senescent cells using Senescence β-Galactosidase Staining
Kit (Cell Signaling Technology, USA) following the manufacturer’s
instructions.
Soft agar colony-forming assay
We used a standard procedure for soft agar colony-forming assays in a
24-well plate. In each well, we plated 500 µl of 1:1 mix of 1%
molecular biology grade agar and 2 × medium (RMPI 1640 or DMEM)
supplemented with 20% FBS, then agar layer was left to harden for
30 min. The soft agar layers were prepared by mixing either stable KD
cells or control cells with a 500 µl mix of 1:1 0.6% agar and
2 × medium supplemented with 20% FBS. The soft agar layers containing
2500 cells/well were left for 15 min to harden and then we added 500 µl
of 1 × RPMI 1640 or DMEM supplemented with 10% FBS on the top of the
agar layers to prevent any possible dehydration. For each cell line, we
plated 10 wells. After 10 days of incubation at 37 °C, we captured
pictures of each well using an automated Z-stack function of the EVOS™
FL Auto Cell Imaging System (ThermoFisher Scientific). We counted the
number of total colonies in each condition and measured the surface
area of at least 20 representative colonies of each cell line.
Statistical significance was derived using two-sided unpaired Student’s
t-test.
Cell migration and invasion assays
We assayed the migration properties of stable KD cells using OrisTM
Universal Cell Migration Assembly Kit (Platypus Technologies). Briefly,
stable KD or control Caki-2, 786-O, and A549 cells were seeded into a
96-well plate with Oris Cell Seeding Stoppers at 1.3 × 10^4, 1 × 10^4,
and 1.7 × 10^4 cells per well, respectively. To create the detection
area, the stoppers were removed after 16 h; stoppers were left in place
for the reference wells (t = 0 pre-migration control) until the results
are read. We used EVOS™ FL Auto Imaging System (Life Technologies) to
detect cell migration at 8 and 24 h post seeding in case of Caki-2 and
786-O cell lines, and 24 and 48 h in case of A549 cells. The area of
pre-migration (t = 0) and post-migration (t = 8 and 24 h) were
calculated for each condition.
Transwell invasion assay was performed using the 24-well plates BD
BioCoat Matrigel Invasion Chamber (BD Biosciences) with 8 µm inserts.
For transiently transfected cells, we first performed siRNA and
scramble sequence transfection, as described previously. Eight hours
post-transfection 3.5 × 10^4 Caki-2 transfected cells were re-suspended
in 1% FBS RPMI-1640 media and seeded into matrigel-coated inserts. For
Caki-2 and A549 stable KD cell lines, 3 × 10^4 cells were re-suspended
in the appropriate medium supplemented with 1% FBS and seeded into the
inserts. Lower chambers were filled with 500 µl of complete medium with
10% FBS as a chemoattractant agent. Invasion chambers were incubated at
humidified 5% CO[2] incubator at 37 °C for 22 h. Non-migrated cells
were scraped from the interior of the inserts by using a cotton-tipped
swab. Cells on the lower surface of the membrane were fixed and stained
using the Snabb-Diff kit (Labex), according to the manufacturer´s
instructions. After staining, the inserts were washed twice in
distilled water, and then the membranes were removed from the inserts
and kept in slides. Invading cells were photographed at ×20
magnification, and the total number of migrated cells was counted using
the EVOS™ FL Auto Imaging System (Life Technologies).
Chromatin oligo-affinity precipitation
The ChOP assay was performed as described before by (Akhade et
al.)^[282]63 with some modifications. For identification of SCAT7
interacting proteins, HeLa cells (20 × 10^6) were UV crosslinked. The
crosslinked pellet was obtained by centrifugation at 1000 × g at 4 °C
for 10 min. Cells were resuspended in 2 ml of buffer A (3 mM MgCl[2],
10miM Tris-HCl, pH 7.4, 10 mM NaCl, 0.5%v/v NP-40, 0.5 mM PMSF and 100
units/ml RNasin) and incubated on ice for 20 min. Nuclei were harvested
by centrifugation and resuspended in 1.2 ml of buffer B (50 mM
Tris-HCl, pH 7.4, 10 mM EDTA, 0.5% Triton X-100, 0.1%SDS, 0.5 mM PMSF,
and 100 units/ml RNasin) and incubated on ice for 40 min. An equal
volume of buffer C (15 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1%
Triton X-100, 0.5 mM PMSF, and 100 units/ml RNasin) was then added and
incubated on ice for 15 min. Samples were briefly sonicated using a
Bioruptor sonicator (Diagenode) for 20 cycles (30 s on, 30 s off at
High Pulse). Eight different oligos complimentary to SCAT7 were pooled
with a final concentration of 10 µM and then used for the RNA pull
down. As a control, a pool of eight oligos in reverse orientation to
the SCAT7 targeting oligos was used. The oligos were added to the
chromatin solution along with yeast tRNA (100 µg/ml), salmon sperm DNA
(100 µg/ml), and incubated overnight at 4 °C. Samples were then
incubated with streptavidin agarose beads for 3 h followed by one wash
of each Low salt buffer (20 mM Tris-HCl, pH7.9, 150 mM NaCl, 2 mM EDTA,
0.1% SDS, 1% TritonX-100, 0.5 mM PMSF, and 50 units/ml RNasin), and
high salt buffer (20 mM Tris-HCl, pH7.9, 500 mM NaCl, 2 mM EDTA, 0.1%
SDS, 1% Triton X-100, 0.5 mM PMSF, and 50 units/ml RNasin) and three
washes of 1 × PBS. The protein complex was eluted from the beads by
incubation with PBS + 0.1%SDS with intermittent mixing at 80 °C for
10 min. All biotinylated oligonucleotides used in the ChOP pull-down
are listed in Supplementary Data [283]4. Few changes were done to the
above protocol while assessing the occupancy of SCAT7 at FGFR2
promoter. These include (1) formaldehyde crosslinking of cells (instead
of UV crosslinking), (2) sonication for 40 cycles (instead of 20
cycles) (3) inclusion of BSA (400 µg/ml) in the oligo binding buffer
(4) DNA isolation using phenol–chloroform method after elution from the
beads. The % input calculations were made considering the percentage of
input chromatin used and the Ct values obtained for the target promoter
region from input DNA and ChOP DNA as follows:
[MATH: %Input =%ofstartinginputfraction×2∧Ctinput-CtChOP :MATH]
Chromatin and RNA immunoprecipitation
ChIP was performed according to the protocol described in ref. ^[284]64
with some modifications. HeLa cells (20 × 10^6) were harvested and
crosslinked using 1% formaldehdye for 10 min at room temperature
followed by quenching using 0.125 M of glycine for 5 min. The
crosslinked pellet was obtained by centrifugation at 1000 × g at 4 °C
for 10 min. The cell pellet was resuspended in 1 ml of SDS lysis buffer
(0.1% SDS, 0.5% Triton X-100, 20 mM Tris-HCl, pH 8, and 150 mM NaCl,
1 mM PMSF, and 100 units/ml RNasin) and incubated on ice for 30 min and
sonicated using a Bioruptor for a total of 40 cycles (30 s on, 30 s off
at High Pulse). Insoluble material was removed by centrifugation at
13000 × g at 4 °C for 10 min. Sonicated DNA was enriched in the range
of 100–500 bp. The lysate was incubated with the respective antibodies
for immune-precipitation overnight at 4 °C. An aliquot of 4 μg of each
antibody was used per 1 mg of lysate for immune-precipitation. The
immune-complexes were allowed to bind to Protein G/A Dynabeads for 3 h
at 4 °C. The immune-complexes bound to beads were obtained by magnetic
precipitation followed by one wash of each low salt buffer (0.1% SDS,
1% Triton-X 100, 2 mM EDTA, 20 mM Tris-HCl, 150 mM NaCl, 0.5 mM PMSF,
and 50 units/ml RNasin) and high salt buffer (0.1% SDS, 1% Triton-X
100, 2 mM EDTA, 20 mM Tris-HCl, 500 mM NaCl, 0.5 mM PMSF, and 50
units/ml RNasin). The immune-precipitated material was eluted from the
beads by adding 400 μl of elution buffer (100 mM NaHCO[3], 1% SDS,
0.5 mM PMSF and 50 units/ml RNasin) and the samples were incubated at
55 °C for 30 min. The eluates were then processed for DNA isolation by
phenol–chloroform method. RIP was performed as described
earlier^[285]23. The eluates from the RIP were processed for total RNA
isolation by Trizol method. A aliquot of 250 ng of RNA was used for
cDNA synthesis. The % input calculations were made considering the
percentage of input chromatin used and the Ct values obtained for the
target promoter region from input DNA and ChIP DNA as follows:
[MATH: %Input = %ofstartinginputfraction×2∧Ctinput-CtChIP :MATH]
Droplet digital PCR
Droplet digital PCR was performed using the One-Step RT-ddPCR Advanced
Kit (BioRAD #1864021) as per the manufacturer’s instructions. The PCR
was performed in CFX96 thermal cycler and quantification was done with
QX200 droplet reader.
Mouse xenografts and PDX models
KIRC and LUAD xenograft models were generated using stable KD cells.
786-O and A549 stable KD cells or control cells were re-suspended in
cold PBS with 20% matrigel. We engrafted 1 × 10^6 cells subcutaneously
in the right flank of 6-week-old female Balb/C nude mice (Janvier
labs). Eight weeks post-engrafting, we dissected and measured tumors
volumes using the following formula: (long side) × (short side)^2/2.
For the treatment of LUAD tumors with LNA, we engrafted 1 × 10^6
wild-type A549 cells subcutaneously. We began treatment with in vivo
grade LNA GapmeR molecules at a final concentration of 0.1 mg/Kg when
the outside volume of the majority of the tumors reached around
0.5 cm^3. We used four mice per group. After terminating the
experiment, we dissected the tumors and collected blood samples,
livers, spleens, and kidneys from all mice. We obtained the
immunocompromised NSG mice models engrafted with patient-derived tumors
from the PDX Live™ library at The Jackson Laboratory (Model ID:
TM00302). We validated the expression of SCAT7 in the obtained
xenografts by analyzing the RNA-seq data of the original early passaged
tumors provided confidentially by The Jackson Library. The mice were
kept for one week to acclimatize prior to the LNA injection. For each
group (n = 6), we injected either 100 pmol of scrambled LNA or SCAT7
LNA1 every three days for a total period of 15 days. The tumor volumes
and growth rates were calculated using the same previously mentioned
formula. The animal study protocol was reviewed and approved by the
Animal Ethical Review Board, University of Gothenburg, Sweden (Ethical
permit no. 45–2015).
Antibodies and raw Western blots
Uncropped scans of all western blots are shown in Supplementary
Figs. [286]9 and [287]10. The list of all antibodies used along with
their catalog number and dilutions used is provided as Supplementary
Data [288]5.
Data availability
The data associated with this publication have been deposited in GEO:
[289]GSE92250. All other data are available from the authors upon
reasonable request.
Electronic supplementary material
[290]Supplementary Information^ (3.4MB, pdf)
[291]Peer Review File^ (766.7KB, pdf)
[292]41467_2018_3265_MOESM3_ESM.pdf^ (172.7KB, pdf)
Description of Additional Supplementary Files
[293]Supplementary Data 1^ (1.3MB, xlsx)
[294]Supplementary Data 2^ (825.1KB, xlsx)
[295]Supplementary Data 3^ (1MB, xlsx)
[296]Supplementary Data 4^ (14.7KB, xlsx)
[297]Supplementary Data 5^ (12.5KB, xlsx)
Acknowledgements