Mechanism study of serum extracellular nano-vesicles miR-412-3p targeting regulation of TEAD1 in promoting malignant biological behavior of sub-centimeter lung nodules
Abstract
OBJECTIVE:
To investigate the impact and potential mechanisms of serum extracellular nano-vesicles (sEVs) miR-412-3p released from sub-centimeter lung nodules with a diameter of
METHODS:
A total of 87 participants were included and divided into a mnLC group (
RESULTS:
The expression level of sEVs-miR-412-3p in the mnLC group was significantly higher than that in the BLN and healthy groups (
CONCLUSION:
sEVs-miR-412-3p could promote the biological process of EMT, and lead to the occurrence of malignant biological behavior in sub-centimeter lung nodules. This provides evidence for the miR-412-3p/TEAD1 signaling axis as a potential therapeutic target for mnLC.
1.Introduction
Lung cancer is one of the most common and poorly prognostic cancers worldwide, and also one of the most burdensome malignant tumors worldwide, seriously endangering human health [1, 2]. Therefore, the early diagnosis and treatment of lung cancer are of great significance for improving the quality of life of patients and reducing mortality [3, 4]. With the widespread application of low-dose spiral computed tomography (LDCT) in clinical practice, the detection rate of sub-centimeter lung nodules with a diameter of
Serum extracellular nano-vesicles (sEVs) are extracellular vesicles secreted by cells with a diameter ranging from 30 to 150 nm, widely present in various bodily fluids [7]. After release, sEVs are absorbed by neighboring or distant cells, and their miRNAs are involved in regulating tumor immunity and microenvironment, which may further promote tumor growth, invasion, metastasis, angiogenesis, and drug resistance [8, 9, 10]. miRNAs have become important biomarkers in non-invasive liquid biopsy of various tumors [11, 12]. Previous studies reported that sEVs-miR-519a-3p secreted by gastric cancer cells could induce M2-like macrophage-mediated angiogenesis in the liver and promote liver metastasis [13]. In lung cancer, sEVs-miR-942 derived from M2 macrophages may promote lung cancer cell invasion and migration, as well as angiogenesis, making it a new therapeutic target for metastatic lung cancer [14]. Related fibroblasts in lung cancer tissue might promote tumor progression by releasing sEVs-miR-142-5p [15]. The above studies indicate that the enhanced proliferation, migration, and invasion ability of tumor cells involve changes in the microenvironment, and sEVs are important mediators in regulating the tumor microenvironment. In particular, sEVs-mediated functional miRNAs play an important role in mediating tumor occurrence and development. However, the biological function of sEVs miRNAs released from sub-cellular lung nodules in mnLC is currently unclear. In the preliminary research of this study, sEVs were isolated from the peripheral serum of patients with pulmonary nodules, and the expression profile of miRNAs contained in them was analyzed by sequencing. Our previous studies discovered that some sEVs miRNAs could be used as new biological detection markers to distinguish between benign and malignant pulmonary nodules [16, 17], among which miR-412-3p is highly expressed in the peripheral serum of early lung cancer patients. Therefore, we will further investigate the role and mechanism of miR-412-3p in the malignant biological behavior of sub-centimeter pulmonary nodules.
2.Materials and methods
2.1Clinical sample collection
This study collected 87 participants from the Department of Respiratory, Thoracic Surgery, and Oncology at the Affiliated Zhongda Hospital of Southeast University from January 2022 to June 2023. They were divided into an mnLC group (
2.2Purification and identification of sEVs
Referring to our previous research [16, 17], 4–5 mL of peripheral blood was extracted from all participants, and sEVs were purified. Observe the morphological characteristics of sEVs using a transmission electronic microscope (TEM) (Tecnai G2 Spirit, 120 KV, Dawson Creek Drive Hillsboro, OR, USA). Evaluate the particle size distribution of sEVs through nanoparticle tracking analysis (NTA). The data was processed using a particle tracking program (Zetasizer Ver. 7.03, Malvern Instruments, Ltd, UK).
2.3Cell culture
Normal lung epithelial cell line (BEAS-2B) and lung adenocarcinoma cell line (A549, NCI-1299, PC-9, and H1975) (ATCC, Virginia, USA), human renal epithelial cell line 293T (Wuhan Punosai, CL-0005). Lung adenocarcinoma cell lines were cultured in RPMI-1640 (A549, NCI-1299, PC-9) and DMEM complete medium (BEAS-2B, 293T, and H1975) containing 10% fetal bovine serum (FBS; Sigma Aldrich, St Louis, MO), 100 U/mL penicillin, and Cultivate streptomycin with a concentration of 100
2.4RT-qPCR
Total sEVs and BEAS-2B cells, as well as lung cancer A549, NCI-1299, PC-9, and H1975 cells, were extracted using Trizol LS (Invitrogen Life Technologies, MA, USA) reagent. After extraction, the total RNA concentration was measured using Nanodrop2000, and gene expression levels were quantitatively analyzed using Applied Biosystems 7500 Fast. Relative quantitative determination of each gene using the double standard curve method, with a relative expression level of 2 for each gene-
2.5Cell transfection
Synthesize mimics and inhibitors of miR-412-3p (synthesized by Shanghai Shenggong). When the cell fusion reaches 90%, it is passaged. Inoculate cells into a 6-well plate and culture for 24 h. When the cells fuse to 70%, intervene according to the reagent instructions. After transfection, continue to culture for 48 h and collect cells for subsequent experiments.
2.6Marking of sEVs
Follow the product manual of sEVs green fluorescent labeling dye (PKH67) for subsequent operations (Shanghai Noning Biotechnology Co., Ltd.). Firstly, quantify the extracted sEVs, then add dye working solution and cover the centrifuge tube tightly. Mix well with a vortex oscillator for 1 min, then let it stand and incubate for 10 min; Add 10 mL of PBS to the incubated sEVs dye complex and mix well. Extract sEVs again using the sEVs extraction method to remove excess dye; Take 200
2.7CCK-8 cell viability experiment
Inoculate logarithmic growth phase cells in a 96 well plate, and transfect them when the cell growth fusion rate reaches 50%. Count 2000 cells per well, and set up 3 wells in each group. After transfection, continue to culture for 24, 48, 72, and 96 h. Add CCK-8 reagent 10
2.8Clone formation assay
Take each group of cells with a logarithmic growth phase, digest them with 0.25% trypsin, and blow them into individual cells. Inoculate them on a 6-well plate with 700 cells per well, and culture in a 37∘C incubator with a CO2 content of 5%. Change the culture medium every three days until 21 days after cultivation, wash with PBS three times, then add 4% formaldehyde, fix for 10 min, and wash again three times. Add 1 mL of 1% crystal violet for staining for 15 min, wash three times with PBS solution, observe, take photos, and calculate the number of clones formed.
2.9Transwell test
Take each group of cells in the logarithmic growth phase, digest them with 0.25% trypsin, blow them into a single cell suspension, adjust the cell count to 5
2.10Stem cell sphere-forming assay
Digest and treat A549 and PC-9 lung cancer cells with 0.25% trypsin, centrifuge cell suspension at 300
2.11Double luciferase reporter gene detection
Using software (https://www.targetscan.org/vert_80/), one of the downstream target genes predicted to bind to miR-412-3p in lung cancer cells is transcriptional enhancer-associated domain 1 (TEAD1). Based on the binding sites of miR-412-3p and TEAD1, wild-type vectors (TEAD1 Wt) and mutant vectors (TEAD1 Mut) of TEAD1 were constructed. TEAD1 Wt+miR-412-3p mimics and TEAD1 Mut+miR-412-3p mimics were transfected into A549 lung cancer cells and 293T cells using liposomes, respectively. After transfection for 48 h, the detection was performed using a luciferase reporter gene detection kit. The operation steps are shown in the instructions [16].
2.12Western blot
Collect transfected A549 and PC-9 cells and shake them on ice with lysis buffer for lysis. Quantify with BCA protein, separate by SDA-PVDF electrophoresis, and transfer to PVDF membrane. Seal the PVDF membrane at room temperature using 8% skim milk for 2 h. The ratio of primary antibodies: CD81 (1:1000, ab155760, Abcam), TSG101 (1:1000, ab225877, Abcam), Calnexin (1:1000, ab92573, Abcam), E-cadherin (1:10000, ab40772, Abcam), N-cadherin (1:5000, ab76011, Abcam), TEAD1 (1:1000, D3F7L, CST),
2.13Statistical analysis
Statistical analysis was conducted using the SPSS 20.0 (SPSS Inc., Chicago, USA) software system, and the experimental data was expressed as mean
3.Results
3.1The expression level of miR-412-3p in sEVs of patients with sub-centimeter pulmonary nodules
Figure 1.
TEM intuitively confirmed the presence of sEVs of all three groups of participants, with a diameter of 40
3.2The effect of miR-412-3p on the viability and stemness of lung cancer cells
Figure 2.
Research has reported that over 85% of malignant lung nodules are non-small cell lung cancer (NSCLC), with the majority being lung adenocarcinoma [18]. Therefore, lung adenocarcinoma cell lines (A549, NCI-1299, PC-9, and H1975) were selected as experimental cell lines in this experiment. The results showed that miR-412-3p was highly expressed in sEVs in the mnLC group, further exploring the impact of miR-412-3p on the malignant biological function of lung cancer cells. The RT-qPCR detection results showed that compared with normal lung cancer epithelial cells (BEAS-2B), the expression level of miR-412-3p in lung cancer cells (A549, NCI-1299, PC-9, and H1975) was significantly increased, with the highest expression level in PC-9 cells and the lowest expression level in A549 cells (
The synthesized miR-412-3p mimics and miR-412-3p inhibitor were transfected into A549 and PC-9 cells, respectively. RT-qPCR detection results showed that compared with the miR-NC mimics group, the transfected miR-412-3p mimics group significantly promoted the expression level of miR-412-3p in lung cancer A549 cells (
Figure 3.
Further cloning experiments showed that compared with the miR-NC mimics group, the miR-412-3p mimics group significantly promoted the cloning ability of lung cancer in A549 cells (
The results of the stem cell sphere-forming assay showed that compared with the miR-NC mimics group, the miR-412-3p mimics group significantly promoted the stemness ability of lung cancer A549 cells (
3.3The effect of miR-412-3p on the migration and invasion ability of lung cancer cells
Figure 4.
Transwell test results showed that compared with the miR-NC mimics group, the miR-412-3p mimics group significantly promoted the migration and invasion of lung cancer A549 cells (
Further WB analysis showed that in A549 cells, transfection of miR-412-3p mimics significantly inhibited the expression of E-cadherin protein in lung cancer cells compared to miR-NC mimics, while promoting the expression of N-cadherin and Vimentin proteins (
3.4The effect of miR-412-3p on TEAD1 protein in lung cancer cells
Figure 5.
Using software (https://www.targetscan.org/vert_80/), it is predicted that one of the downstream target genes that miR-412-3p may bind to in lung cancer cells is TEAD1, and there is a binding site for miR-412-3p in the 3’UTR of its mRNA (Fig. 5A). Transfect wild-type (TEAD1 Wt) and mutant (TEAD1 Mut) luciferase reporter gene vectors into A549 cells, respectively. At the same time, co-transfect with miR-412-3p mimics and detects the activity of the luciferase reporter gene in the cells for 48 h. The results showed that in the TEAD1 Wt vector, transfection of miR-412-3p mimics significantly inhibited cell luciferase activity compared to miR-NC mimics (
Further validation was conducted in 293T human renal epithelial cells, and similar results were obtained. In the wild-type (TEAD1 Wt) vector, compared with the miR-NC mimics group, the transfected miR-412-3p mimics group significantly inhibited cell luciferase activity (
3.5MiR-412-3p affects the proliferation and migration ability of lung cancer cells by regulating the target gene TEAD1
Figure 6.
Next, we will further investigate whether miR-412-3p affects the proliferation and migration ability of lung cancer cells by regulating TEAD1. Based on the above research results, it can be inferred that miR-412-3p negatively regulates the expression of TEAD1 in lung cancer cells. To further validate this inference, the constructed blank vector (pcDNA-NC) and overexpressing TEAD1 vector (pcDNA-TEAD1) were transfected into lung cancer cell A549 cells, respectively, and miR-412-3p mimics were co-transfected. The results of clone formation and migration experiments showed that compared with miR-NC mimics, transfection of miR-412-3p mimics significantly promoted the clone formation (
3.6sEVs promote the proliferation, stemness, and migration ability of lung cancer cells
Figure 7.
Select A549 cells with the lowest expression level of miR-412-3p from lung cancer cells (A549, NCI-1299, PC-9, and H1975) as the experimental cell model for detecting the effect of sEVs on lung cancer cells. Separate sEVs from the mnLC group, BLN group, and healthy group, and incubate them with A549 cells respectively. The changes in sEVs uptake by A549 cells can be clearly observed using immunofluorescence, and the results show that green fluorescent labeled sEVs (PKH67) can enter A549 cells (Fig. 7A). Further cellular biology functional experiments showed that compared with the sEVs of the BLN and healthy people groups, sEVs in the mnLC group significantly promoted the ability of lung cancer cells to form clones, dry and migrate (
Further WB analysis showed that compared with the sEVs of the BLN and health groups, the sEVs of the mnLC group significantly inhibited the expression of E-cadherin and TEAD1 proteins in lung cancer cells, while promoting the expression of N-cadherin and Vimentin proteins (
Figure 8.
In summary, the results of this study found that sEVs-miR-412-3p was highly expressed in the serum of mnLC patients, suggesting that miR-412-3p may play an important role in the occurrence of early lung cancer. The results of in vitro cell experiments confirmed our hypothesis: miR-412-3p targeted regulation of the TEAD1 signaling pathway plays an important role in early lung cancer (Fig. 8). The high expression of miR-412-3p in lung cancer cells promotes the expression of N-cadherin and Vimentin proteins, inhibits the expression of E-cadherin proteins, induces the epithelial-mesenchymal transition (EMT) process in lung cancer cells, and enhances their proliferation, stemness, and migration abilities by inhibiting TEAD1.
4.Discussion
Lung cancer is one of the most widely distributed malignant tumors worldwide, with an age-standardized 5-year survival rate of only 10% to 20% [19]. The EMT process is related to tumor occurrence and development, and tumor cells enhance their migration and invasion abilities through the EMT process [20]. During the process, EMT not only manifests as changes in cellular morphology, but is also accompanied by changes in related molecular markers such as E-cadherin, N-cadherin, and Vimentin [21]. Early lung cancer screening for high-risk populations has become an important link in reducing lung cancer mortality and reducing the burden of lung cancer in various countries. With the widespread application of LDCT in early lung cancer screening, the detection rate of sub-centimeter lung nodules with a diameter of
The sEVs are important mediators of intercellular communication, carrying abundant miRNAs and proteins that play important roles in information transmission between tumor cells and their microenvironment. Meanwhile, sEVs participate in regulating tumor cell proliferation, stemness, migration, and invasion ability, and can serve as a potential biomarker for early diagnosis of tumors [22, 23, 24]. Previous research reported fibrinogen
miR-412-3p is a newly identified class of non-coding miRNAs that are involved in regulating the occurrence and development of colorectal and renal cancer [29, 30]. In colorectal cancer, miR-412-3p acts as a pro-oncogene and promotes tumor progression by negatively regulating the expression of MYL9 which may mediate canceration through the TGF
The results of this study’s cell function experiment showed that transfection of miR-412-3p mimics significantly promoted the proliferation, stemness, migration, and invasion ability of lung cancer cells, while transfection of miR-412-3p inhibitor significantly inhibited the biological functions of lung cancer cells mentioned above, suggesting that miR-412-3p, as an oncogenic gene, significantly promote the progression of lung cancer. Therefore, we speculate that sEVs-miR-412-3p released by mnLC may significantly promote the proliferation of lung cancer cells, enhance the stem cell characteristics of lung cancer cells themselves, and enhance their ability to invade and migrate surrounding tissues. Biological information software prediction and luciferase reporter gene results show that one of the target genes of miR-412-3p is TEAD1, and miR-412-3p binds and negatively regulates the expression of TEAD1. Previous studies have reported that TEAD1 could regulate cell proliferation and stem cell function [33, 34], and reducing TEAD1 expression should significantly affect Hippo, Wnt, and TGF-
In summary, this study constraints that miR-412-3p targets negative regulation of TEAD1 and plays an important role in the malignant proliferation, stemness, invasion, and migration ability, as well as EMT process of lung cancer cells, exhibiting a progression in promoting the malignant biological behavior of sub-centimeter lung nodules. Therefore, drugs or inhibitors targeting the miR-412-3p/TEAD1 signaling axis may be an effective target for inhibiting the malignant biological behavior of mnLC in the future.
Funding
This research was supported by Jiangsu Funding Program for Excellent Postdoctoral Talent (2022ZB799).
Author contributions
Conception: YD, NP, HZ.
Interpretation or analysis of data: YD, NP, SD, HZ.
Preparation of the manuscript: YD, NP, SD, HZ.
Revision for important intellectual content: YD, NP, HZ.
Supervision: HZ.
Acknowledgments
None.
Conflict of interest
Nothing to declare.
References
[1] | F. Abu Rous, E.K. Singhi, A. Sridhar, M.S. Faisal and A. Desai, Lung Cancer Treatment Advances in 2022, Cancer Invest 41: ((2023) ), 12–24. |
[2] | S.J. Adams, E. Stone, D.R. Baldwin, R. Vliegenthart, P. Lee and F.J. Fintelmann, Lung cancer screening, Lancet401 ((2023) ), 390–408. |
[3] | S. Huang, J. Yang, N. Shen, Q. Xu and Q. Zhao, Artificial intelligence in lung cancer diagnosis and prognosis: Current application and future perspective, Semin Cancer Biol 89: ((2023) ), 30–37. |
[4] | P. Li, S. Liu, L. Du, G. Mohseni, Y. Zhang and C. Wang, Liquid biopsies based on DNA methylation as biomarkers for the detection and prognosis of lung cancer, Clin Epigenetics 14: ((2022) ), 118. |
[5] | Z. Wu, F. Wang, W. Cao, C. Qin, X. Dong, Z. Yang, Y. Zheng, Z. Luo, L. Zhao, Y. Yu, Y. Xu, J. Li, W. Tang, S. Shen, N. Wu, F. Tan, N. Li and J. He, Lung cancer risk prediction models based on pulmonary nodules: A systematic review, Thorac Cancer 13: ((2022) ), 664–677. |
[6] | T.J. Kim, C.H. Kim, H.Y. Lee, M.J. Chung, S.H. Shin, K.J. Lee and K.S. Lee, Management of incidental pulmonary nodules: current strategies and future perspectives, Expert Rev Respir Med 14: ((2020) ), 173–194. |
[7] | R. Kalluri and V.S. LeBleu, The biology, function, and biomedical applications of exosomes, Science, 367: ((2020) ), eaau6977. |
[8] | R. Isaac, F.C.G. Reis, W. Ying and J.M. Olefsky, Exosomes as mediators of intercellular crosstalk in metabolism, Cell Metab 33: ((2021) ), 1744–1762. |
[9] | B. Li, Y. Cao, M. Sun and H. Feng, Expression, regulation, and function of exosome-derived miRNAs in cancer progression and therapy, Faseb J 35: ((2021) ), e21916. |
[10] | J. Chen, F. Cao, Y. Cao, S. Wei, X. Zhu and W. Xing, Targeted therapy of lung adenocarcinoma by the nanoplatform based on milk exosomes loaded with paclitaxel, J Biomed Nanotechnol 18: ((2022) ), 1075–1083. |
[11] | V.C. Kok and C.C. Yu, Cancer-Derived Exosomes: Their Role in Cancer Biology and Biomarker Development, Int J Nanomedicine 15: ((2020) ), 8019–8036. |
[12] | F.G. Kugeratski, K. Hodge, S. Lilla, K.M. McAndrews, X. Zhou, R.F. Hwang, S. Zanivan and R. Kalluri, Quantitative proteomics identifies the core proteome of exosomes with syntenin-1 as the highest abundant protein and a putative universal biomarker, Nat Cell Biol 23: ((2021) ), 631–641. |
[13] | S. Qiu, L. Xie, C. Lu, C. Gu, Y. Xia, J. Lv, Z. Xuan, L. Fang, J. Yang, L. Zhang, Z. Li, W. Wang, H. Xu, B. Li and Z. Xu, Gastric cancer-derived exosomal miR-519a-3p promotes liver metastasis by inducing intrahepatic M2-like macrophage-mediated angiogenesis, J Exp Clin Cancer Res 41: ((2022) ), 296. |
[14] | K. Wei, Z. Ma, F. Yang, X. Zhao, W. Jiang, C. Pan, Z. Li, X. Pan, Z. He, J. Xu, W. Wu, Y. Xia and L. Chen, M2 macrophage-derived exosomes promote lung adenocarcinoma progression by delivering miR-942, Cancer Lett 526: ((2022) ), 205–216. |
[15] | Y. Jiang, K. Wang, X. Lu, Y. Wang and J. Chen, Cancer-associated fibroblasts-derived exosomes promote lung cancer progression by OIP5-AS1/ miR-142-5p/ PD-L1 axis, Mol Immunol 140: ((2021) ), 47–58. |
[16] | Y. Deng, R. Xue, N. Patel, W. Xu and H. Zhang, Serum extracellular nano-vesicles miR-153-3p to Identify micronodular lung cancer from sub-centimeter lung nodules, J Biomed Nanotechnol 18: ((2022) ), 705–717. |
[17] | N. Patel, W. Xu, Y. Deng, J. Jin and H. Zhang, Cross-scale integration of nano-sized extracellular vesicle-based biomarker and radiomics features for predicting suspected sub-solid pulmonary nodules, J Biomed Nanotechnol 17: ((2021) ), 1109–1122. |
[18] | Y. Su, H.B. Fang and F. Jiang, An epigenetic classifier for early stage lung cancer, Clin Epigenetics 10: ((2018) ), 68. |
[19] | C. Allemani, T. Matsuda, V. Di Carlo, R. Harewood, M. Matz, M. Nikšić, A. Bonaventure, M. Valkov, C.J. Johnson, J. Estève, O.J. Ogunbiyi, E.S.G. Azevedo, W.Q. Chen, S. Eser, G. Engholm, C.A. Stiller, A. Monnereau, R.R. Woods, O. Visser, G.H. Lim, J. Aitken, H.K. Weir and M.P. Coleman, Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries, Lancet 391: ((2018) ), 1023–1075. |
[20] | A. Dongre and R.A. Weinberg, New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer, Nat Rev Mol Cell Biol 20: ((2019) ), 69–84. |
[21] | R. Kalluri and R.A. Weinberg, The basics of epithelial-mesenchymal transition, J Clin Invest 119: ((2009) ), 1420–1428. |
[22] | Q.F. Han, W.J. Li, K.S. Hu, J. Gao, W.L. Zhai, J.H. Yang and S.J. Zhang, Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer, Mol Cancer 21: ((2022) ), 207. |
[23] | F. Man, J. Wang and R. Lu, Techniques and applications of animal- and plant-derived exosome-based drug delivery system, J Biomed Nanotechnol 16: ((2020) ), 1543–1569. |
[24] | D. He, X. Xu, L. Li, C. Chen, K. Gong, Q. Guo, F. Liu, Y. Wang, Y. Duan and H. Li, Functional exosome-mediated delivery of triptolide endowed with targeting properties as chemotherapy carriers for ovarian carcinoma, J Biomed Nanotechnol 17: ((2021) ), 426–438. |
[25] | M. Kuang, Y. Peng, X. Tao, Z. Zhou, H. Mao, L. Zhuge, Y. Sun and H. Zhang, FGB and FGG derived from plasma exosomes as potential biomarkers to distinguish benign from malignant pulmonary nodules, Clin Exp Med 19: ((2019) ), 557–564. |
[26] | M. Kuang, X. Tao, Y. Peng, W. Zhang, Y. Pan, L. Cheng, C. Yuan, Y. Zhao, H. Mao, L. Zhuge, Z. Zhou, H. Chen and Y. Sun, Proteomic analysis of plasma exosomes to differentiate malignant from benign pulmonary nodules, Clin Proteomics 16: ((2019) ), 5. |
[27] | G. Yang, T. Wang, X. Qu, S. Chen, Z. Han, S. Chen, M. Chen, J. Lin, S. Yu, L. Gao, K. Peng and M. Kang, Exosomal miR-21/Let-7a ratio distinguishes non-small cell lung cancer from benign pulmonary diseases, Asia Pac J Clin Oncol 16: ((2020) ), 280–286. |
[28] | X. Li, Q. Zhang, X. Jin and L. Cao, Combining serum miRNAs, CEA, and CYFRA21-1 with imaging and clinical features to distinguish benign and malignant pulmonary nodules: a pilot study: Xianfeng Li, et al.: Combining biomarker, imaging, and clinical features to distinguish pulmonary nodules, World J Surg Oncol 15: ((2017) ), 107. |
[29] | Y. Wang, Y. Zhang, X. Su, Q. Qiu, Y. Yuan, C. Weng, S. Zou, Y. Tian, W. Han, P. Liu, X. Guo, J. Mao, X. Fu, P. Wang and W. Lin, Circular RNA circDVL1 inhibits clear cell renal cell carcinoma progression through the miR-412-3p/PCDH7 axis, Int J Biol Sci 18: ((2022) ), 1491–1507. |
[30] | K. Zhu, Y. Wang, L. Liu, S. Li and W. Yu, Long non-coding RNA MBNL1-AS1 regulates proliferation, migration, and invasion of cancer stem cells in colon cancer by interacting with MYL9 via sponging microRNA-412-3p, Clin Res Hepatol Gastroenterol 44: ((2020) ), 101–114. |
[31] | S. Deng, D. Cheng, J. Wang, J. Gu, Y. Xue, Z. Jiang, L. Qin, F. Mao, Y. Cao and K. Cai, MYL9 expressed in cancer-associated fibroblasts regulate the immune microenvironment of colorectal cancer and promotes tumor progression in an autocrine manner, J Exp Clin Cancer Res 42: ((2023) ), 294. |
[32] | X. Zhou, B.L. Updegraff, Y. Guo, M. Peyton, L. Girard, J.E. Larsen, X.J. Xie, Y. Zhou, T.H. Hwang, Y. Xie, J. Rodriguez-Canales, P. Villalobos, C. Behrens, Wistuba, II, J.D. Minna and K.A. O’Donnell, PROTOCADHERIN 7 Acts through SET and PP2A to Potentiate MAPK Signaling by EGFR and KRAS during Lung Tumorigenesis, Cancer Res 77: ((2017) ), 187–197. |
[33] | H.D. Huh, D.H. Kim, H.S. Jeong and H.W. Park, Regulation of TEAD transcription factors in cancer biology, Cells 8: ((2019) ), 600. |
[34] | A.V. Pobbati, R. Kumar, B.P. Rubin and W. Hong, Therapeutic targeting of TEAD transcription factors in cancer, Trends Biochem Sci 48: ((2023) ), 450–462. |
[35] | L. Currey, S. Thor and M. Piper, TEAD family transcription factors in development and disease, Development 148: ((2021) ), dev196675. |
[36] | T.J. Hagenbeek, J.R. Zbieg, M. Hafner, R. Mroue, J.A. Lacap, N.M. Sodir, C.L. Noland, S. Afghani, A. Kishore, K.P. Bhat, X. Yao, S. Schmidt, S. Clausen, M. Steffek, W. Lee, P. Beroza, S. Martin, E. Lin, R. Fong, P. Di Lello, M.H. Kubala, M.N. Yang, J.T. Lau, E. Chan, A. Arrazate, L. An, E. Levy, M.N. Lorenzo, H.J. Lee, T.H. Pham, Z. Modrusan, R. Zang, Y.C. Chen, M. Kabza, M. Ahmed, J. Li, M.T. Chang, D. Maddalo, M. Evangelista, X. Ye, J.J. Crawford and A. Dey, An allosteric pan-TEAD inhibitor blocks oncogenic YAP/TAZ signaling and overcomes KRAS G12C inhibitor resistance, Nat Cancer 4: ((2023) ), 812–828. |