Skip to main content
Erschienen in: Cancer Cell International 1/2021

Open Access 01.12.2021 | Primary research

AL355338 acts as an oncogenic lncRNA by interacting with protein ENO1 to regulate EGFR/AKT pathway in NSCLC

verfasst von: Qian Hua, Dongliang Wang, Lin Zhao, Zhihui Hong, Kairu Ni, Yizhen Shi, Zengli Liu, Baoming Mi

Erschienen in: Cancer Cell International | Ausgabe 1/2021

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Non-small cell lung cancer (NSCLC) is a malignancy with considerable morbidity and mortality. Abnormal metabolism is a hallmark of cancer; however, the mechanism of glycolysis regulation in NSCLC progression is not completely understood. Recent studies suggest that some dysregulated long non-coding RNAs (lncRNAs) play important roles in tumor metabolic reprogramming.

Methods

To identify glycolysis-associated-lncRNAs in NSCLC, we compared RNA-sequencing results between high 18F-fluorodeoxyglucose (FDG)-uptake NSCLC tissues and paired paratumor tissues. The transcript abundance of AL355338 in 80 pairs of clinical samples was evaluated by quantitative real-time PCR assay and fluorescence in situ hybridization. The biological role of AL355338 on NSCLC cells were evaluated by functional experiments in vitro and in vivo. Moreover, RNA pull-down, mass spectrometry and RNA immunoprecipitation (RIP) assays were used to identify the protein interacted with AL355338. Co-immunoprecipitation, in situ proximity ligation assays and western blotting were applied to define the potential downstream pathways of AL355338.

Results

AL355338 was an upregulated glycolysis-associated lncRNA in NSCLC. Functional assays revealed that AL355338 was critical for promoting aerobic glycolysis and NSCLC progression. Mechanistic investigations showed that AL355338 directly bound with alpha-enolase (ENO1) and enhanced the protein’s stability by modulating its degradation and ubiquitination. A positive correlation was observed between AL355338 and ENO1 in NSCLC, and ENO1 was subsequently confirmed to be responsible for the oncogenic role of AL355338. Furthermore, AL355338 was capable of modulating ENO1/EGFR complex interaction and further activating EGFR-AKT signaling.

Conclusions

This study indicates that AL355338 confers an aggressive phenotype to NSCLC, and targeting it might be an effective therapeutic strategy.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12935-021-02232-z.
Qian Hua, Dongliang Wang and Lin Zhao contributed equally to this study

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
lncRNA
Long noncoding RNA
NSCLC
Non-small cell lung cancer
EMT
Epithelial-mesenchymal transition
FDG
Fluorodeoxyglucose
TNM
Tumor-node-metastasis staging system
IP
Immunoprecipitation
RIP
RNA immunoprecipitation
FISH
Fluorescence in situ hybridization
IHC
Immunohistochemistry
IF
Immunofluorescence
PLA
Proximity Ligation Assay
ENO1
Enolase 1
HK2
Hexokinase II
PKM2
Pyruvate kinase isozymes M2
LDHA
Lactate dehydrogenase A
EGFR
Epidermal growth factor receptor
AKT
Serine/threonine kinase
TCGA
The cancer genome atlas

Background

Among malignancies, lung cancer is one of the most lethal with high global mortality and incidence rates [1]. Non-small cell lung cancer (NSCLC) is the most commonly diagnosed type, accounting for ~ 85% of cases [2]. Early diagnosis of NSCLC is difficult, and most patients are diagnosed at advanced stages and have poor clinical outcomes [2]. In-depth investigation into the molecular alterations and their mechanisms that underlie NSCLC progression is extremely essential to develop and optimize therapeutic strategies.
Cancer cell growth depends on sufficient energy. Previous studies suggested that glucose metabolism reprogramming, also known as the Warburg effect, provided fuel for cancer’s malignant behavior [3]. 18F-fluorodeoxyglucose (FDG) is an analogue of glucose and radioactive tracer that enables tracking of glucose uptake in solid tumors in positron emission tomography/computed tomography (PET/CT) imaging, which is the gold standard for quantifying glycolytic metabolism [4]. Enhanced glucose uptake ability has been observed in patients with various types of cancer, and especially in NSCLC [5]. Cancer biology studies using genomic techniques revealed that the orchestrated performance of oncogenes and downstream pathway activation exacerbate metabolic disorder [6]. However, the underlying mechanistic details underlying glycolysis regulation in NSCLC progression remain to be determined.
Long non-coding RNAs (lncRNAs) are RNA transcripts (> 200 nucleotides) with limited coding potential [7] that are abnormally expressed in all stages of cancer development and play vital regulatory roles in oncogenic processes [8]. A recent study reported that lncRNA dysregulation is closely related to tumor metabolic reprogramming [9]. Further work is needed to identify glycolysis-related-lncRNAs in NSCLC and clarify their potential roles and specific mechanisms.
Alpha-enolase (ENO1) is an enolase isoform originally characterized as a crucial glycolytic enzyme catalyzing the dehydration of 2-phosphoglycerate into phosphoenolpyruvate; it also generates ATP during glycolysis [10]. Besides its well-known enzymatic functions, decades of research have shown that ENO1 is aberrantly expressed in multiple tumor types and participates in diverse cellular processes through non-metabolic mechanisms [11]. Interestingly, some studies reported that lncRNAs interact with ENO1 and affect its expression or activity [12, 13], highlighting a novel approach to elucidate the non-metabolic roles of metabolic genes in cancer.
Recently, we aimed to identify glycolysis-associated-lncRNAs through RNA-sequencing (RNA-seq) of high 18F-FDG-uptake NSCLC tissues and paired paratumor tissues. AL355338 was the top-scoring upregulated lncRNA. In this present study, we observed that high AL355338 expression positively correlated with NSCLC clinical stage and worse patient prognosis. Functional assays demonstrated the influences of AL355338 silencing on NSCLC proliferation, metastasis, and glycolysis in vitro and in vivo. Mechanistically, our evidence indicated that AL355338 could directly interact with ENO1 and stabilize the protein, preventing its ubiquitination and degradation. AL355338 exerted its biological functions partially by upregulating ENO1 expression. Further experiments showed that AL355338 positively modulated the interaction between ENO1 and epidermal growth factor receptor (EGFR), subsequently activating EGFR/AKT signaling. Collectively, these results strongly suggest that the AL355338-ENO1-EGFR/AKT regulatory axis may harbor NSCLC diagnostic biomarkers and therapeutic targets.

Materials and methods

Patient and clinical information

Eighty paired NSCLC tissues and paratumor tissues were collected from the surgery specimen archives of the Second Affiliated Hospital of Soochow University. Patient follow-up continued to December 2020. No recruited patients received preoperative chemotherapy or radiotherapy. This research was approved by the institutional clinical research ethics committee of the Second Affiliated Hospital of Soochow University. Patient stratification was based on the new TNM staging criteria in lung cancer (8th edition) [14].

Cell culture and treatment

The human normal lung epithelial cell line HBE and four human NSCLC cell lines (A549, PC9, H1650, H1299) were obtained from ATCC (Manassas, VA, UA) and cultured in Dulbecco's modified Eagle’s medium (Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (Gibco) at 37 °C, in a humid 5% CO2 incubator. Two small interfering RNA (siRNA) constructs (GenePharma, Shanghai, China) were designed to knock down AL355338 expression, and non-specific siRNA served as the negative control; all siRNAs were transfected with Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). For ectopic AL355338, ENO1, and EGFR expression, the appropriate cDNA was amplified with reverse transcription polymerase chain reaction (RT-PCR) and subcloned into a pcDNA-4/TO vector. Neofect transfection reagent (Neofect biotech, Beijing, China) was used for plasmid transfection. The sh-AL355338, sh-ENO1 and sh-NC lentiviruses were purchased from GenePharma. The siRNA and sh-RNA sequences are listed in Additional file 1: Table S1.

Total RNA isolation and quantitative RT-PCR (qRT-PCR) assays

Total RNA was extracted from cells or tissues with TRIzol reagent (Omega, Norcross, GA, USA) according to the manufacturer’s instructions. RNA quality and quantity were assessed with the NanoDrop2000 platform (Thermo Fisher Scientific, Waltham, MA, USA). cDNA Synthesis kits (TaKaRa, Otsu, Japan) were used for mRNA reverse transcription. Real-time PCR analysis was performed with SYBR Green PCR Kits (TaKaRa) on the 7500 Sequence Detection System (Applied Biosystems, Foster City, CA, USA). The amplified transcript levels of genes were normalized to β-actin with the optimized comparative 2−ΔΔCt value method. The primer sequences are in Additional file 1: Table S1.

Nuclear-cytoplasmic fractionation

The nuclear and cytoplasmic fractions were generated with a PARIS™ Kit (Ambion, Austin, TX, USA) according to the manufacturer protocol. Nuclear and cytoplasmic RNA levels were normalized to U6 and β-actin, respectively using real-time PCR.

Fluorescence in situ hybridization (FISH)

FISH analyses were performed with RNA FISH kits (RiboBio, Guangzhou, China) using a previously described method [15]. The AL35538 probe sequence was 5′-DIG-ACTTCGAGACCAGCATGGCCAACATGGTGAAGCCC-DIG-3′. Nuclei were stained with DAPI, and high-resolution images were taken with a confocal microscope (ZEISS, Oberkochen, Germany). Percentages of positive stained cells were quantified as: 1 = 0–25%, 2 = 26–50%, 3 = 51–75%, and 4 = 76–100%. Staining intensity scores were: 0 = none, 1 = weak, 2 = moderate, and 3 = strong. The final scores were calculated by multiplying the percentage and intensity scores of positive cells. Samples were divided into low (score 0–6) and high (score 7–12) expression groups.

Flow cytometry assay

Transfected cells were harvested and fixed in pre-cooled 70% ethanol for 1 h on ice. Then cells were washed with cold phosphate-buffered saline (PBS) three times and incubated with RNase for 30 min at 37 °C. After staining the cells with propidium Iodide for 15 min at room temperature, they were analyzed with a flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA). The percentages of cells in the G0/G1, S, and G2/M phases were calculated using ModFit software (Verity Software, Topsham, ME, USA).

Cell proliferation assay

Cell proliferation rates were measured with Cell Counting Kit-8 (bimake, Shanghai, China) according to the manufacturer’s protocol. Briefly, 5 × 103 cells per well were seeded into 96-well plates. At 0, 24, 48, and 72 h after cell transfection, 10 μL CCK-8 solution was added to each well. After incubating he plates for 2 h at 37 °C, the optical density at 450 nm was measured for each well. For colony formation assays, transfected or control A549 and PC9 cells were harvested 24 h after transfection, and 500 cells per well were seeded into 6-well plates. After 2 weeks of incubation, the colonies were fixed in 4% paraformaldehyde and stained with crystal violet solution. Cell proliferation was also assessed using ethynyldeoxyuridine (EdU) analyses (RiboBio) according to the manufacturer’s instructions as previously reported [16].

Cell migration and invasion assay

Wound healing assays were carried out as previously described [17]. Transwell assays were performed using Boyden chambers containing transwell membrane filters (Corning, Corning, NY, USA). The detailed protocol was published elsewhere [17]. Images were collected with an inverted microscope (Olympus, Tokyo, Japan) and analyzed with ImageJ software (National Institutes of Health, Bethesda, MD, USA). At least five random fields of view were analyzed in each chamber.

Measurement of glucose uptake, lactate production, and ATP

As a glucose analogue, 18F-FDG uptake reflects the intracellular glucose uptake level. In vitro experiments were performed as previously reported [16]. Lactate production was measured in cells that were seeded into 12-well plates in triplicate for 24 h and then refreshed with 1 mM glucose culture medium overnight. Culture supernatants were collected to measure lactate concentrations (Nanjing Jiancheng Bioengineering Institute, Nanjing, China), and lysed cell pellets were used to measure ATP levels (Nanjing Jiancheng) using the manufacturer’s instructions. 18F-FDG uptake and lactate production were normalized to number of cells in each sample; ATP levels were normalized to cell protein mass.

Western blot

Western blotting was performed following a standard protocol [18]. The antibodies used are listed in Additional file 1: Table S2. Immunoreactive bands were detected using ECL western blot kits (Amersham Biosciences, Little Chalfont, UK).

Immunofluorescence (IF)

IF was performed as described elsewhere [18]. Protein expression and localization were observed under a confocal microscope (ZEISS). Antibodies used for IF are listed in Additional file 1: Table S2.

RNA pull-down assay

Biotin-RNA pull-down assays were performed as described elsewhere [15]. Briefly, full-length sense, antisense, and serial deletion sequences of AL355338 were amplified with a T7-containing primer and reverse transcribed using MAXIscript™ T7 Transcription Kits (Thermo Fisher Scientific). Pierce™ RNA 3′ End Desthiobiotinylation Kits (Thermo Fisher Scientific) were used to label targeted RNA. The biotin-labeled AL355338 probe was incubated with total cell lysates of A549 cells using Pierce™ Magnetic RNA–Protein Pull-Down Kits (Thermo Fisher Scientific). Eluted proteins were purified by silver staining and subjected to mass spectrometry (MS) or western blotting.

RNA immunoprecipitation (RIP) assay

Magna RNA-binding protein immunoprecipitation kits (Millipore, Billerica, MA, USA) were used to determine specific protein binding with targeted RNA according to the manufacturer’s protocol and as described previously [15]. Briefly, cells were lysed with RIP lysis buffer, and lysates were incubated overnight at 4 °C with magnetic beads conjugated with human anti-ENO1 antibody or anti-mouse immunoglobulin G (IgG). The next day, co-immunoprecipitated RNA was extracted for qRT-PCR analyses. Total RNA (input controls) and IgG controls were simultaneously assayed to confirm that the RNA specifically bound to ENO1.

Co-immunoprecipitation (Co-IP)

Reciprocal exogenous and endogenous co-IP assays were performed as described elsewhere reported [18]. IP samples and input controls were analyzed by western blot. The co-IP antibodies are listed in Additional file 1: Table S2.

Proximity ligation assay (PLA)

PLA (Duolink®, Sigma-Aldrich, St. Louis, MO, USA) was performed to evaluate interactions between ENO1 and EGFR as previously reported [19]. Briefly, A549 and PC9 cells were grown on glass coverslips in 24-well plates at 37 °C overnight. The next day, cells were fixed in 4% paraformaldehyde for 20 min and permeabilized with 0.1% Triton X-100 for 5 min. After blocking for 30 min, the cells were incubated with anti-ENO1 and anti-EGFR antibodies diluted in blocking buffer at 4 °C overnight. The next day, cells were incubated with species-specific PLA probes at 37 °C for 1 h, then 1× ligase for 30 min, and finally 1× polymerase diluted with amplification stock solution at 37 °C for 100 min to amplify the fluorescence signal. Finally, coverslips were mounted on slides with anti-fluorescence quenching sealing liquid containing DAPI. Images were acquired under confocal microscopy (ZEISS).

In vivo xenograft assays

Four-week-old male BALB/c nude mice were purchased from the Animal Center of Soochow University. Animal experiments were carried out according to the protocol approved by the Animal Care and Ethics Committee of the Second Affiliated Hospital of Soochow University. A total of 20 mice were subcutaneously injected with 5 × 106 A549 cells in the right flank with sh-AL355339, and sh-NC was given in the left flank to establish an NSCLC xenograft model. Tumor volume was measured every 5th day and calculated according to the equation V = length × width2 × 1/2. Micro-PET/CT (Super Nova® PET/CT, Pingseng, China) was performed 3 weeks after cell injection to measure 18F-FDG uptake by xenograft tumors. All tumor-bearing mice were fasted for 8 h prior to PET/CT. 18F-FDG (0.1 mL, 7.4 MBq) was injected via tail vein. After 30 min, they were anesthetized with 2% isoflurane in oxygen and immobilized for 15-min scans. 18F-FDG uptake was measured as maximum standard uptake (SUVmax) in the regions of interest covering the tumors. After 3 weeks, mice were humanely sacrificed by euthanasia. The mice were inhaled with CO2 for 5 min. The CO2 was gradually filled into an airtight chamber at 30% of the chamber volume/min rate. The xenograft tumors were harvested and measured.

In vivo metastasis assays

Stably transfected A549 cells (1 × 106/0.2 ml PBS) were injected into the tail vein of nude mice. Micro-CT scans were performed 5 weeks later to observe lung metastasis. After 5 weeks, mice were humanely sacrificed by euthanasia. Following sacrifice, the lungs were surgically dissected and embedded in paraffin for hematoxylin and eosin (HE) staining. The numbers of pulmonary metastatic nodules were compared between groups.

Immunohistochemistry (IHC)

IHC was performed and quantified as previously described [20].

Statistical analysis

All data are expressed as mean ± standard deviation. Statistical analysis was performed with SPSS (version 22.0, IBM Corp., Armonk, NY, USA) and graphs were generated with GraphPad Prism software (version 8.0, GraphPad Inc., San Diego, CA, USA). Student’s t-tests or one-way analyses of variance were used to assess significant differences between groups. Categorical data were compared with Chi-square tests. Survival analyses were performed using Kaplan–Meier analyses.

Results

AL355338 was upregulated in aggressive NSCLC and associated with poor patient prognosis

To identify glycolysis-related lncRNAs in NSCLC, we performed RNA-seq assay with 18F-FDG high-uptake NSCLC tissues and pair-matched normal tissues from our previous study [16]. We detected 222 upregulated genes (fold change > 1.5, false discovery rate < 0.005). The top 20 significantly upregulated lncRNAs (Additional file 1: Figure S1A) were submitted for Gene Ontology analysis, and the results showed that they were most enriched in metabolic pathways (Additional file 1: Figure S1B). To screen more functional lncRNAs, our next investigation used more stringent filtering criteria (> 400 bp, not fully overlapped with other coding transcripts, efficient PCR amplification). In the current study, we focused on the uncharacterized AL355338, which was the top-scoring upregulated lncRNA in the RNA-seq data, and explored its function in NSCLC development. AL355338 is a 913-bp transcript on human chromosome 13 with 2 exons (Additional file 1: Figure S1C). The Coding-Potential Assessment Tool (CPAT), PhyloCSF tool, and ORFfinder all predicted that AL355338 had no coding potential (Additional file 1: Figure S1D, E). To explore the clinical significance of AL355338, we analyzed RNA-seq data from the publicly available TCGA dataset (StarBase V3.0 platform). AL355338 expression was significantly upregulated in lung adenocarcinoma and lung squamous carcinoma tissues (Additional file 1: Figure S2A, B). Kaplan–Meier survival analysis revealed that patients with high AL355338 expression has shorter overall survival than patients with low AL355338 expression (Additional file 1: Figure S2C, D).
QRT-PCR analysis confirmed that AL355338 was increased in tumor tissues compared with the corresponding paratumor tissues in a panel of 80 specimens from NSCLC patients (Fig. 1A). Furthermore, AL355338 expression was remarkably higher in metastatic samples compared with non-metastatic samples, and elevated AL355338 expression correlated with advanced TNM stage (Fig. 1B, C). Kaplan–Meier survival analysis of overall survival showed that high AL355338 expression was associated with shorter survival (Fig. 1D; P = 0.032). We next detected AL355338 with FISH and evaluated the relationship between its expression and clinicopathological characteristics. Consistent with the qRT-PCR results, more positive AL355338 expression signals were observed in tumor tissues compared to adjacent normal tissues (Fig. 1E). Expression score analyses demonstrated that expression percentages of AL355338 were higher in stage III + IV than in stage I + II NSCLC samples (Fig. 1F). High AL355338 expression was positively associated with T stage, N stage, distant metastasis, and TNM stage (Table 1). However, it was not related to patient age, sex, or tumor location. Uni- and multivariate regression analyses revealed that AL355338 expression was an independent predictor of NSCLC aggressiveness and could predict clinical outcome (Fig. 1G, H). Collectively, these results suggested that AL355338 was upregulated in NSCLC and might be a useful prognostic biomarker.
Table 1
Correlation of the expression of AL355338 in NSCLC with clinicopathologic features
Characteristics
 
AL355338
P value
Low (n)
High (n)
Age (years)
 < 60
34 (42.5%)
16
18
0.54
 ≥ 60
46 (57.5%)
21
25
 
Gender
 Male
41 (51.2%)
17
24
0.26
 Female
39 (48.8%)
20
19
 
TNM stage
 I
23 (28.7%)
19
4
< 0.01**
 II
19 (23.8%)
14
5
 
 III
22 (27.5%)
4
18
 
 IV
16 (20.0%)
0
16
 
T stage
 T1
21 (26.3%)
12
9
0.04*
 T2
37 (46.3%)
19
18
 
 T3
16 (20.0%)
5
11
 
 T4
6 (7.5%)
1
5
 
N stage
 N0
32 (40.0%)
23
9
0.03*
 N1
17 (21.3%)
7
10
 
 N2
23 (28.7%)
6
17
 
 N3
8 (10.0%)
1
7
 
M stage
 M0
65 (81.3%)
37
28
< 0.01**
 M1
15 (18.8%)
0
15
 
Mortality
 Survive
30 (37.5%)
17
13
0.04*
 Die
50 (62.5%)
20
30
 
Histological grade
 I
5 (6.2%)
2
3
0.38
 II
42 (52.5%)
24
18
 
 III
33 (41.3%)
11
22
 
Tumor location
 Left
35 (43.8%)
15
20
0.55
 Right
45 (56.2%)
22
23
 
*P < 0.05; **P < 0.01

AL355338 promoted NSCLC cell proliferation in vitro

To explore the potential function of AL355338 on cellular behavior, we first detected the expression levels of AL355338 in NSCLC cell lines. Its expression in NSCLC cell lines was significantly higher than that in the normal lung epithelial cell line HBE (Additional file 1: Figure S3A). We then examined its subcellular localization in NSCLC cells. Cellular FISH experiments and analyses of RNA isolated from the cytoplasmic and nuclear compartments of NSCLC cells showed that AL355338 was mainly present in the cytoplasm (Fig. 2A, B). Then, we downregulated AL355338 by transfecting two different siRNAs in A549 and PC9 cells. qRT-PCR was performed to verify the knockdown efficiency of AL355338; both si-AL355338#1 and si-AL355338#2 successfully downregulated AL355338 expression to 20–30% in A549 and PC9 cells (Fig. 2C).
Flow cytometry analyses of cell cycle distribution indicated that AL355338 downregulation significantly decreased the number of cells in S phase, with an increase of cells in G0/G1 phase for both A549 and PC9 cells (Fig. 2D–F). CCK-8 and cell colony formation assays demonstrated that the proliferation and colony formation ability of A549 and PC9 cells were impaired after AL355338 knockdown (Fig. 2E, G, H). Subsequent EdU assays indicated that DNA synthesis by A549 and PC9 cells was markedly reduced after silencing AL355338 (Fig. 2I). These results indicated that AL355338 was effective in facilitating NSCLC cell proliferation.

AL355338 facilitated aggressive phenotypes and glycolytic metabolism of NSCLC cells

To further assess the migratory and invasive ability conferred by AL355338, we performed transwell and wound healing assays. The transwell results in Fig. 3A–C show that downregulation of AL355338 suppressed the migratory and invasive activities of A549 and PC9 cells compared to the negative control. Wound healing assays showed the same pattern (Fig. 3D–F). The initiation of epithelial-mesenchymal transition is a driving force of tumor invasion and metastasis. Analyses of the protein expression of epithelial and mesenchymal markers revealed that AL355338 silencing reduced N-cadherin and vimentin expression but enhanced E-cadherin expression in A549 and PC9 cells (Fig. 3G). Consistently, IF staining also showed that the fluorescence intensity of Vimentin was attenuated in AL355338 downregulated A549 cells (Additional file 1: Figure S3B).
Considering AL355338 expression was upregulated in association with high 18F-FDG uptake, we investigated the effect of AL355338 in glycolysis regulation. Glucose enzyme expression levels were examined, and the results showed that alteration of AL355338 expression could positively affect hexokinase II, pyruvate kinase isozymes M2, and lactate dehydrogenase A protein levels (Fig. 3H). To confirm the effect of AL355338 on glycolysis, we measured 18F-FDG uptake, lactate release, and ATP production to reflect the flux distribution of glucose. AL355338 knockdown led to a decreases in all three parameters (Fig. 3I–K). These results demonstrated that AL355338 could regulate glycolytic metabolism in NSCLC.

Silencing AL355338 inhibited tumorigenesis, aerobic glycolysis, and metastasis in vivo

To determine whether AL355338 expression could affect NSCLC cell growth in vivo, A549 cells were stably transfected with sh-AL355338 or negative control sh-NC lentivirus. In vivo xenograft experiments showed that tumor volumes in the sh-AL355338 group were obviously smaller than in the sh-NC group (Fig. 4A). Notably, tumor weights were markedly reduced in the sh-AL355338 group compared with controls (Fig. 4B). In addition, xenograft glycolysis uptake ability was observed by 18F-FDG micro-PET/CT imaging. Tumor glycolysis was significantly blocked in sh-AL355338 xenografts, with lower SUVmax (mean SUVmax: 1.15) compared with sh-NC xenografts (mean SUVmax: 2.08; Fig. 4C, D). IHC revealed that the tumor tissue grown from sh-AL355338 cells displayed lower Ki-67, ENO1, and EGFR staining than those formed from sh-NC cells. FISH analyses confirmed the inhibitory effect of knocking down AL355338 expression in A549 cells (Fig. 4E).
We then investigated the function of AL355338 on tumor metastasis in vivo. Micro-CT images showed more metastatic lesions in the lungs of mice in the sh-NC group, while fewer and smaller metastatic lung nodules were seen in the AL355338 knockdown group (Fig. 4F). Summarized data on HE staining analysis demonstrated that more metastatic lung foci were formed in the sh-NC group compared to the AL355338 knockdown group (Fig. 4G). These results indicated that inhibiting AL355338 expression in NSCLC cells could efficiently suppress tumor metastasis in vivo.

AL355338 specifically interacted with ENO1 and enhanced ENO1 protein stability

To investigate the underlying mechanism of AL355338-mediated glycolysis and carcinogenesis in NSCLC, we set out to identify intracellular AL355338-binding factors in an unbiased manner. Biotinylated AL355338 and negative control were incubated with total protein extracts from A549 cells and pulled down with streptavidin (Additional file 1: Figure S4A). MS was conducted following RNA pull-down to screen endogenous AL355338 binding proteins (Additional file 1: Table S3). From the MS data files, we selected ENO1 for further binding validation as it is a key enzyme responsible for the glycolytic pathway. Subsequent western blotting of proteins retrieved from AL355338 pull-down assays confirmed that the ENO1 protein directly bound to the sense AL355338 sequence but did not bind the antisense control (Fig. 5A, upper). In addition, RIP assays with an ENO1 antibody and IgG antibody control were performed using A549 and PC9 cellular extracts. Correspondingly, we observed significantly higher AL355338 enrichment with the ENO1 antibody compared to IgG (Fig. 5A, lower). Both assays demonstrated that AL355338 directly interacted with ENO1. To better identify which region of ENO1 interacted with AL355338, we performed RIP assays using FLAG-tagged truncated forms of ENO1. The results demonstrated that the ENO1-C domain (237–434 aa) was essential for AL355338 binding (Fig. 5B). To map the ENO1-binding regions of AL355338, we constructed four AL355338 deletion fragments. Subsequent RNA pull-down assays followed by western blotting revealed that the ENO1 specific binding sequence was located between 528 and 913 nucleotides (Fig. 5C). RNA FISH of AL355338 combined with IF of ENO1 confirmed cytoplasmic co-localization of AL355338 and ENO1 in A549 cells (Fig. 5D).
A recent study demonstrated that lncRNAs interact with proteins through several modes of action, such as regulating protein expression and function, modulating protein–protein interactions, or guiding protein subcellular localization [21]. To investigate the molecular effect of AL355338 on ENO1, western blotting was performed to measure ENO1 expression after A549 and PC9 cells were subjected to different treatments with AL355338. The results showed that AL355338 positively regulated ENO1 protein expression (Fig. 5E). Similar trends were observed in IF assays (Additional file 1: Figure S4B). Interestingly, loss or gain of function of AL355338 had no influence on ENO1 mRNA expression (Additional file 1: Figure S4C, D), suggesting that AL355338 may regulate ENO1 protein stability. Next, cycloheximide (CHX) chase assays were performed to observe the effect of AL355338 on ENO1 protein degradation. Western blotting showed that ENO1 protein was more stable in AL355338-transfected cells, while it had a shorter half-life in AL355338-silenced cells (Fig. 5F, G). Moreover, the proteasome inhibitor MG132 rescued the reduction of ENO1 caused by AL355338 repression (Fig. 5H), suggesting that AL355338 elevated ENO1 levels by reducing its degradation. As reported, cytoplasmic protein degradation involves the ubiquitin–proteasome pathway [22]. Thus, whether AL355338 was involved in the ubiquitin-mediated ENO1 degradation was tested with in vitro ubiquitination assays. The results illustrated that upregulation of AL355338 decreased ENO1 ubiquitination, whereas silencing of AL355338 enhanced ENO1 ubiquitination (Fig. 5I). Collectively, our data indicated that AL355338 directly bound the ENO1 protein and enhanced its stability in NSCLC cells.

ENO1 participated in the oncogenic function of AL355338

To clarify whether the RNA–protein interaction between AL355338 and ENO1 mediated the key oncogenic function of AL355338 in NSCLC, complementary rescue experiments were performed. Stable ENO1-knockdown A549 and PC9 cells were constructed and then transfected with AL355338 overexpression plasmid. Transfection efficiency was confirmed by qRT-PCR and western blot (Fig. 6A, B). As was expected, CCK-8 and cell colony formation assays showed that ectopic AL355338 expression increased the proliferation activity and colony formation ability of A549 and PC9 cells, while ENO1-knockdown partially abrogated the effects induced by AL355338 upregulation (Fig. 6C, D). EdU assays revealed that increased DNA synthesis capability of A549 and PC9 cells induced with AL355338 was reversed by ENO1 silencing (Fig. 6E). Cell migration and invasion abilities were also assessed, and results demonstrated that AL355338 exerted minimal effects on cell migration and invasion in the absence of ENO1 (Fig. 6F). Importantly, the stimulatory effect of AL355338 on epithelial and mesenchymal marker expression was recovered by repressing ENO1 (Fig. 6G). In glycolysis metabolism assays, ENO1 downregulation dramatically blocked the AL355338-induced increase in 18F-FDG uptake, as well as lactate production by A549 and PC9 cells (Fig. 6H, I). The above results suggested that AL355338 regulation of glycolytic metabolism and cell proliferation were dependent on ENO1 stability in NSCLC cells.

AL355338 activated the EGFR/AKT pathway by targeting the ENO1/EGFR interaction

The EGFR/AKT signaling pathway influences tumor cell survival in many cancers. As previous reports have demonstrated that ENO1 could regulate the AMPK-mediated or FAK-mediated AKT pathway [23, 24], we sought to determine whether ENO1 could interact with EGFR and subsequently activate its downstream AKT kinase signaling pathway. To verify the relationship between ENO1 and EGFR in NSCLC cells, we first examined their TCGA database expression levels. We extracted ENO1 and EGFR data from 34 tumors and found that both were notably upregulated in diverse cancer types, especially lung adenocarcinoma and lung squamous carcinoma tissues (Additional file 1: Figure S5A, B). As shown in Additional file 1: Figure S5C, the correlation coefficient of ENO1 and EGFR reached 0.32 in the NSCLC cohort (P < 0.001, Spearman correlation analysis). Besides, ENO1 shared a similar mutation status with EGFR in NSCLC samples (P < 0.001, Additional file 1: Figure S5D).
Next, we aimed to investigate whether ENO1 directly interacted with EGFR. Reciprocal co-IP assays were performed using both exogenous labeled antibodies and endogenous antibodies to precipitate ENO1 and EGFR, respectively. The co-IP results confirmed physical binding between ENO1 and EGFR proteins in NSCLC cells (Fig. 7A, B). IF staining confirmed ENO1 and EGFR co-localization in A549 and PC9 cells (Fig. 7C). Furthermore, in situ PLA was used to visualize native protein complexes. High densities of PLA-positive protein complexes that included ENO1 and EGFR were observed in A549 and PC9 cells, which was in accordance with the co-IP results (Fig. 7D). Intriguingly, AL355338 knockdown noticeably impaired the ENO1 and EGFR interaction, while AL355338 overexpression had the opposite effect (Fig. 7E, F).
To understand the potential relationship between AL355338 and the EGFR/AKT pathway, we investigated EGFR and AKT expression. Western blot analyses showed that AL355338 suppression decreased the levels of phosphorylated EGFR and AKT, but total protein levels were not affected. Overexpression of AL355338 enhanced EGFR and AKT phosphorylation, indicating that the EGFR/AKT pathway was a downstream effector of AL355338. We then assessed the ability of ENO1 to overcome AL355338-mediated activation of the EGFR/AKT pathway in NSCLC cells. Notably, ENO1 knockdown abrogated AL355338-mediated increases of EGFR and AKT phosphorylation, whereas ENO1 overexpression reversed sh-AL355338-mediated inactivation of the EGFR/AKT pathway (Fig. 7G, H). Based on these results, we inferred that AL355338 could regulate EGFR/AKT pathway activity by targeting ENO1/EGFR interaction.

ENO1 was upregulated in NSCLC and positively correlated with AL355338 expression

To further establish the relationship between AL355338 and ENO1 in NSCLC tissues, we first evaluated ENO1 protein expression with IHC in 80 NSCLC and pair-matched normal tissue samples (Fig. 8A). The results revealed that 76.3% (61/80) NSCLC samples showed increased ENO1 expression compared with their pair-matched normal tissues (Fig. 8B). ENO1 protein levels positively correlated with AL355338 levels (r = 0.382, P < 0.001, Fig. 8C), suggesting potential regulation of ENO1 by AL355338 in clinical NSCLC tissues. Meanwhile, TCGA datasets revealed that ENO1 expression was significantly upregulated in lung adenocarcinoma and lung squamous carcinoma tissues compared to normal tissues (Fig. 8D, E). Kaplan–Meier analysis of the 80-patient NSCLC cohort showed that high ENO1 expression was associated with poor prognosis (P = 0.009, Fig. 8F). This was validated in the TCGA cohort; survival analyses showed that patients with low ENO1 expression had longer overall survival (P = 0.025) and disease-free survival (P = 0.044) compared to those with high ENO1 expression (Fig. 8G, H).

Discussion

Despite developments in therapeutic approaches, the prognosis for NSCLC patients diagnosed in an advanced stage remains poor [25]. The enhanced Warburg effect is a leading cause of malignant features in NSCLC. Multiple glycolysis-associated genes have been reported as contributors to cancer progression [26]. Earlier studies indicated that numerous lncRNAs were associated with NSCLC initiation and development of NSCLC [7, 27]. Since 18F-FDG uptake can reflect the Warburg effect, we previously subjected high 18F-FDG-uptake NSCLC tissues to RNA-seq analysis to identify glycolysis-related lncRNAs in our previous study [16]. AL355338 was a newly identified oncogenic lncRNA among the top-scoring upregulated genes. Clinical analysis showed that AL355338 expression was notably higher in tumor tissues than in corresponding adjacent tissues. Importantly, high AL355338 expression in patients with NSCLC was related to advanced TNM stage and poor clinical outcome. Functionally, the positive regulation of AL355338 in promoting NSCLC proliferation and metastasis was identified in a series of in vitro and in vivo experiments. The observation of high AL355338 expression in glycolytically active tissues led us to investigate whether AL355338 could regulate cancer metabolism. In support of our hypothesis, downregulation of AL355338 dramatically weakened the Warburg effect in NSCLC cells. The results indicate an oncogenic role of AL355338 in regulating malignant phenotype, since abnormal cancer cell metabolism is likely to induce cancer aggressiveness.
In addition to its biological importance, we dissected the mechanisms by which AL355338 mediated glycolysis pathway activation. A cross-linking RNA pull-down/MS strategy was used to detect proteins that directly interacted with AL355338. Among the MS data, the glycolytic enzyme ENO1 piqued our interest. As a well-documented glycolytic enzyme responsible for ATP generation, ENO1 is related to mediate energy metabolism and cancer development [11]. Recent studies reported that ENO1 was correlated with cancer differentiation and progression in several tumor types by interacting with lncRNAs. Hong et al. [12] concluded that lncRNA ENO1-IT1, which was activated by Fusobacterium nucleatum abundance, could promote glycolysis and oncogenesis by targeting ENO1 and guiding its histone modification pattern in colorectal cancer. Another group confirmed that lncRNA SNHG18 promoted glioma cell motility by disrupting the nucleocytoplasmic shuttling of ENO1 [28]. Consistent with the previous report, our results suggested that ENO1 was responsible for the oncogenic role of AL355338 in mediating NSCLC progression. ENO1 silencing weakened the exogenous AL355338-induced metabolic shift in NSCLC cells. AL355338 also maintained ENO1 protein stabilization through decreasing ubiquitin-mediated degradation. Notably, we observed simultaneous upregulation and correlation of AL355338 and ENO1 in NSCLC tissues. ENO1 and AL355338 overexpression were strongly associated with poor prognosis in NSCLC patients, suggesting that AL355338 and ENO1 levels may predict patient outcome.
In addition to its canonical roles in glycolysis, accumulating evidence revealed ENO1 as a multifunctional protein involved in regulating several signaling pathways [29]. A recent study reported that ENO1 promoted the self-renewal and malignant phenotype of lung cancer stem cells by affecting the AMPK/mTOR pathway [23]. Fu et al. [24] showed that ENO1 was involved in NSCLC glycolysis, proliferation, and metastasis through the FAK-mediated PI3K/AKT pathway. It is rather remarkable that EGFR is the best-characterized receptor tyrosine kinase and EGFR signaling is among the most dysregulated molecular pathways in NSCLC [30]. Upon ligand binding to EGFR, the AKT signaling pathway will be activated, leading to enhanced glycolysis and oncogenic phenotypes in tumor cells [31]. Given the positive correlation of ENO1 and EGFR expression in NSCLC and their similar functions in AKT pathway regulation, these reports inspired us to explore whether ENO1 might be an interaction partner of EGFR. Notably, a direct interaction between ENO1 and EGFR was observed by reverse co-IP, and native ENO1/EGFR protein complexes were visualized with PLA assays. Moreover, our data clearly showed that ENO1 positively regulated EGFR phosphorylation and thus activated downstream AKT kinase signaling.
Previous studies revealed that lncRNAs might act as a tumorigenic regulator by modulating signaling networks [32]. Apart from directly modulating ENO1 expression, we hypothesized that AL355338 functions through the ENO1-mediated EGFR/AKT pathway in NSCLC cells. As expected, ectopic AL355338 expression could enhance ENO1/EGFR interaction, which subsequently led to higher levels of phosphorylated EGFR and AKT. Opposite results were observed in AL355338-depleted cells. However, ENO1 silencing attenuated the increases in phosphorylation modulated by AL355338 induction. ENO1 overexpression restored the reduction in phosphorylated EGFR and AKT levels induced by AL355338 knockdown. Collectively, these results indicated that AL355338 might regulate the ENO1-mediated EGFR/AKT pathway.

Conclusions

Our study mainly focused on the glycolysis-associated lncRNA AL355338 in NSCLC. Remarkably, AL355338 was increased in NSCLC tissues, and its levels correlated with clinical outcomes, suggesting it could be an independent biomarker in patients with NSCLC. Further functional assays suggested that AL355338 functions as an oncogene by mediating glycolysis activation and promoting NSCLC progression through selective targeting of ENO1 protein. Additionally, we demonstrated that AL355338 stabilized ENO1 protein to prevent ubiquitin-mediated degradation. AL355338 was also capable of modulating interaction between ENO1 and EGFR, further activating EGFR-AKT signaling. In conclusion, our results suggested that upregulated lncRNA AL355338 might serve as a potential biomarker and therapeutic target in the clinical treatment of NSCLC.

Acknowledgements

Not applicable.

Declarations

This study was reviewed and approved by the Ethics Committee of the Second Affiliated Hospital of Soochow University. Written informed consent was obtained from each patient. And this study was performed in accordance with the Declaration of Helsinki.
Not applicable.

Competing interests

The authors declare that they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33.CrossRef Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33.CrossRef
2.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.CrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.CrossRef
3.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.CrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.CrossRef
4.
Zurück zum Zitat Ntziachristos V, Pleitez MA, Aime S, Brindle KM. Emerging technologies to image tissue metabolism. Cell Metab. 2019;29:518–38.CrossRef Ntziachristos V, Pleitez MA, Aime S, Brindle KM. Emerging technologies to image tissue metabolism. Cell Metab. 2019;29:518–38.CrossRef
5.
Zurück zum Zitat Torresano L, Nuevo-Tapioles C, Santacatterina F, Cuezva JM. Metabolic reprogramming and disease progression in cancer patients. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165721.CrossRef Torresano L, Nuevo-Tapioles C, Santacatterina F, Cuezva JM. Metabolic reprogramming and disease progression in cancer patients. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165721.CrossRef
6.
Zurück zum Zitat Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F, Lisanti MP. Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. 2017;14:113.CrossRef Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F, Lisanti MP. Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. 2017;14:113.CrossRef
7.
Zurück zum Zitat Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018;18:5–18.CrossRef Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018;18:5–18.CrossRef
8.
Zurück zum Zitat Tsagakis I, Douka K, Birds I, Aspden JL. Long non-coding RNAs in development and disease: conservation to mechanisms. J Pathol. 2020;250:480–95.CrossRef Tsagakis I, Douka K, Birds I, Aspden JL. Long non-coding RNAs in development and disease: conservation to mechanisms. J Pathol. 2020;250:480–95.CrossRef
9.
Zurück zum Zitat Xu Y, Qiu M, Shen M, Dong S, Ye G, Shi X, et al. The emerging regulatory roles of long non-coding RNAs implicated in cancer metabolism. Mol Ther. 2021;29:2209–18.CrossRef Xu Y, Qiu M, Shen M, Dong S, Ye G, Shi X, et al. The emerging regulatory roles of long non-coding RNAs implicated in cancer metabolism. Mol Ther. 2021;29:2209–18.CrossRef
10.
Zurück zum Zitat Kim JW, Dang CV. Multifaceted roles of glycolytic enzymes. Trends Biochem Sci. 2005;30:142–50.CrossRef Kim JW, Dang CV. Multifaceted roles of glycolytic enzymes. Trends Biochem Sci. 2005;30:142–50.CrossRef
11.
Zurück zum Zitat Almaguel FA, Sanchez TW, Ortiz-Hernandez GL, Casiano CA. Alpha-enolase: emerging tumor-associated antigen, cancer biomarker, and oncotherapeutic target. Front Genet. 2020;11:614726.CrossRef Almaguel FA, Sanchez TW, Ortiz-Hernandez GL, Casiano CA. Alpha-enolase: emerging tumor-associated antigen, cancer biomarker, and oncotherapeutic target. Front Genet. 2020;11:614726.CrossRef
13.
Zurück zum Zitat Yu S, Li N, Huang Z, Chen R, Yi P, Kang R, et al. A novel lncRNA, TCONS_00006195, represses hepatocellular carcinoma progression by inhibiting enzymatic activity of ENO1. Cell Death Dis. 2018;9:1184.CrossRef Yu S, Li N, Huang Z, Chen R, Yi P, Kang R, et al. A novel lncRNA, TCONS_00006195, represses hepatocellular carcinoma progression by inhibiting enzymatic activity of ENO1. Cell Death Dis. 2018;9:1184.CrossRef
14.
Zurück zum Zitat Detterbeck FC, Boffa DJ, Kim AW, Tanoue LT. The eighth edition lung cancer stage classification. Chest. 2017;151:193–203.CrossRef Detterbeck FC, Boffa DJ, Kim AW, Tanoue LT. The eighth edition lung cancer stage classification. Chest. 2017;151:193–203.CrossRef
15.
Zurück zum Zitat Tang J, Yan T, Bao Y, Shen C, Yu C, Zhu X, et al. LncRNA GLCC1 promotes colorectal carcinogenesis and glucose metabolism by stabilizing c-Myc. Nat Commun. 2019;10:3499.CrossRef Tang J, Yan T, Bao Y, Shen C, Yu C, Zhu X, et al. LncRNA GLCC1 promotes colorectal carcinogenesis and glucose metabolism by stabilizing c-Myc. Nat Commun. 2019;10:3499.CrossRef
16.
Zurück zum Zitat Hua Q, Jin M, Mi B, Xu F, Li T, Zhao L, et al. LINC01123, a c-Myc-activated long non-coding RNA, promotes proliferation and aerobic glycolysis of non-small cell lung cancer through miR-199a-5p/c-Myc axis. J Hematol Oncol. 2019;12:91.CrossRef Hua Q, Jin M, Mi B, Xu F, Li T, Zhao L, et al. LINC01123, a c-Myc-activated long non-coding RNA, promotes proliferation and aerobic glycolysis of non-small cell lung cancer through miR-199a-5p/c-Myc axis. J Hematol Oncol. 2019;12:91.CrossRef
17.
Zurück zum Zitat Shen M, Zhao X, Zhao L, Shi L, An S, Huang G, et al. Met is involved in TIGAR-regulated metastasis of non-small-cell lung cancer. Mol Cancer. 2018;17:88.CrossRef Shen M, Zhao X, Zhao L, Shi L, An S, Huang G, et al. Met is involved in TIGAR-regulated metastasis of non-small-cell lung cancer. Mol Cancer. 2018;17:88.CrossRef
18.
Zurück zum Zitat Jia W, Zhao X, Zhao L, Yan H, Li J, Yang H, et al. Non-canonical roles of PFKFB3 in regulation of cell cycle through binding to CDK4. Oncogene. 2018;37:1685–98.CrossRef Jia W, Zhao X, Zhao L, Yan H, Li J, Yang H, et al. Non-canonical roles of PFKFB3 in regulation of cell cycle through binding to CDK4. Oncogene. 2018;37:1685–98.CrossRef
19.
Zurück zum Zitat Hua Q, Mi B, Xu F, Wen J, Zhao L, Liu J, et al. Hypoxia-induced lncRNA-AC020978 promotes proliferation and glycolytic metabolism of non-small cell lung cancer by regulating PKM2/HIF-1α axis. Theranostics. 2020;10:4762–78.CrossRef Hua Q, Mi B, Xu F, Wen J, Zhao L, Liu J, et al. Hypoxia-induced lncRNA-AC020978 promotes proliferation and glycolytic metabolism of non-small cell lung cancer by regulating PKM2/HIF-1α axis. Theranostics. 2020;10:4762–78.CrossRef
20.
Zurück zum Zitat Li L, Liang Y, Kang L, Liu Y, Gao S, Chen S, et al. Transcriptional regulation of the Warburg effect in cancer by SIX1. Cancer Cell. 2018;33:368-385.e7.CrossRef Li L, Liang Y, Kang L, Liu Y, Gao S, Chen S, et al. Transcriptional regulation of the Warburg effect in cancer by SIX1. Cancer Cell. 2018;33:368-385.e7.CrossRef
21.
Zurück zum Zitat Díaz-Ramos A, Roig-Borrellas A, García-Melero A, López-Alemany R. α-Enolase, a multifunctional protein: its role on pathophysiological situations. J Biomed Biotechnol. 2012;2012:156795.CrossRef Díaz-Ramos A, Roig-Borrellas A, García-Melero A, López-Alemany R. α-Enolase, a multifunctional protein: its role on pathophysiological situations. J Biomed Biotechnol. 2012;2012:156795.CrossRef
22.
Zurück zum Zitat Wang J, Maldonado MA. The ubiquitin-proteasome system and its role in inflammatory and autoimmune diseases. Cell Mol Immunol. 2006;3:255–61.PubMed Wang J, Maldonado MA. The ubiquitin-proteasome system and its role in inflammatory and autoimmune diseases. Cell Mol Immunol. 2006;3:255–61.PubMed
23.
Zurück zum Zitat Dai J, Zhou Q, Chen J, Rexius-Hall ML, Rehman J, Zhou G. Alpha-enolase regulates the malignant phenotype of pulmonary artery smooth muscle cells via the AMPK-Akt pathway. Nat Commun. 2018;9:3850.CrossRef Dai J, Zhou Q, Chen J, Rexius-Hall ML, Rehman J, Zhou G. Alpha-enolase regulates the malignant phenotype of pulmonary artery smooth muscle cells via the AMPK-Akt pathway. Nat Commun. 2018;9:3850.CrossRef
24.
Zurück zum Zitat Fu QF, Liu Y, Fan Y, Hua SN, Qu HY, Dong SW, et al. Alpha-enolase promotes cell glycolysis, growth, migration, and invasion in non-small cell lung cancer through FAK-mediated PI3K/AKT pathway. J Hematol Oncol. 2015;8:22.CrossRef Fu QF, Liu Y, Fan Y, Hua SN, Qu HY, Dong SW, et al. Alpha-enolase promotes cell glycolysis, growth, migration, and invasion in non-small cell lung cancer through FAK-mediated PI3K/AKT pathway. J Hematol Oncol. 2015;8:22.CrossRef
25.
Zurück zum Zitat Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: new biological insights and recent therapeutic advances. CA Cancer J Clin. 2011;61:91–112.CrossRef Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: new biological insights and recent therapeutic advances. CA Cancer J Clin. 2011;61:91–112.CrossRef
26.
Zurück zum Zitat Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacol Res. 2019;150:104511.CrossRef Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacol Res. 2019;150:104511.CrossRef
27.
Zurück zum Zitat Vencken SF, Greene CM, McKiernan PJ. Non-coding RNA as lung disease biomarkers. Thorax. 2015;70:501–3.CrossRef Vencken SF, Greene CM, McKiernan PJ. Non-coding RNA as lung disease biomarkers. Thorax. 2015;70:501–3.CrossRef
28.
Zurück zum Zitat Zheng R, Yao Q, Li X, Xu B. Long noncoding ribonucleic acid SNHG18 promotes glioma cell motility via disruption of α-enolase nucleocytoplasmic transport. Front Genet. 2019;10:1140.CrossRef Zheng R, Yao Q, Li X, Xu B. Long noncoding ribonucleic acid SNHG18 promotes glioma cell motility via disruption of α-enolase nucleocytoplasmic transport. Front Genet. 2019;10:1140.CrossRef
29.
Zurück zum Zitat Cappello P, Principe M, Bulfamante S, Novelli F. Alpha-Enolase (ENO1), a potential target in novel immunotherapies. Front Biosci. 2017;22:944–59.CrossRef Cappello P, Principe M, Bulfamante S, Novelli F. Alpha-Enolase (ENO1), a potential target in novel immunotherapies. Front Biosci. 2017;22:944–59.CrossRef
30.
Zurück zum Zitat Stencel K, Chmielewska I, Milanowski J, Ramlau R. Non-small-cell lung cancer: new rare targets-new targeted therapies-state of the art and future directions. Cancers. 2021;13:1829.CrossRef Stencel K, Chmielewska I, Milanowski J, Ramlau R. Non-small-cell lung cancer: new rare targets-new targeted therapies-state of the art and future directions. Cancers. 2021;13:1829.CrossRef
31.
32.
Zurück zum Zitat Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol Rev. 2016;96:1297–325.CrossRef Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol Rev. 2016;96:1297–325.CrossRef
Metadaten
Titel
AL355338 acts as an oncogenic lncRNA by interacting with protein ENO1 to regulate EGFR/AKT pathway in NSCLC
verfasst von
Qian Hua
Dongliang Wang
Lin Zhao
Zhihui Hong
Kairu Ni
Yizhen Shi
Zengli Liu
Baoming Mi
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2021
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02232-z

Weitere Artikel der Ausgabe 1/2021

Cancer Cell International 1/2021 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.