Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2024, Vol. 18 Issue (5) : 878-895    https://doi.org/10.1007/s11684-024-1057-7
ING5 inhibits aerobic glycolysis of lung cancer cells by promoting TIE1-mediated phosphorylation of pyruvate dehydrogenase kinase 1 at Y163
Haihua Zhang1, Xinli Liu2, Junqiang Li3, Jin Meng4, Wan Huang5, Xuan Su1, Xutao Zhang6, Guizhou Gao1, Xiaodong Wang1, Haichuan Su3(), Feng Zhang2(), Tao Zhang1()
. Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China
. Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an 710038, China
. Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China
. Department of Pharmacy, the Medical Security Centre, Chinese PLA General Hospital, Beijing 100091, China
. National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi’an 710038, China
. Aerospace Clinical Medical Center, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710038, China
 Download: PDF(6707 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Aerobic glycolysis is critical for tumor growth and metastasis. Previously, we have found that the overexpression of the inhibitor of growth 5 (ING5) inhibits lung cancer aggressiveness and epithelial–mesenchymal transition (EMT). However, whether ING5 regulates lung cancer metabolism reprogramming remains unknown. Here, by quantitative proteomics, we showed that ING5 differentially regulates protein phosphorylation and identified a new site (Y163) of the key glycolytic enzyme PDK1 whose phosphorylation was upregulated 13.847-fold. By clinical study, decreased p-PDK1Y163 was observed in lung cancer tissues and correlated with poor survival. p-PDK1Y163 represents the negative regulatory mechanism of PDK1 by causing PDHA1 dephosphorylation and activation, leading to switching from glycolysis to oxidative phosphorylation, with increasing oxygen consumption and decreasing lactate production. These effects could be impaired by PDK1Y163F mutation, which also impaired the inhibitory effects of ING5 on cancer cell EMT and invasiveness. Mouse xenograft models confirmed the indispensable role of p-PDK1Y163 in ING5-inhibited tumor growth and metastasis. By siRNA screening, ING5-upregulated TIE1 was identified as the upstream tyrosine protein kinase targeting PDK1Y163. TIE1 knockdown induced the dephosphorylation of PDK1Y163 and increased the migration and invasion of lung cancer cells. Collectively, ING5 overexpression—upregulated TIE1 phosphorylates PDK1Y163, which is critical for the inhibition of aerobic glycolysis and invasiveness of lung cancer cells.

Keywords ING5      aerobic glycolysis      PDK1      phosphorylation      lung cancer      TIE1     
Corresponding Author(s): Haichuan Su,Feng Zhang,Tao Zhang   
Just Accepted Date: 26 July 2024   Online First Date: 14 September 2024    Issue Date: 29 October 2024
 Cite this article:   
Haihua Zhang,Xinli Liu,Junqiang Li, et al. ING5 inhibits aerobic glycolysis of lung cancer cells by promoting TIE1-mediated phosphorylation of pyruvate dehydrogenase kinase 1 at Y163[J]. Front. Med., 2024, 18(5): 878-895.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-024-1057-7
https://academic.hep.com.cn/fmd/EN/Y2024/V18/I5/878
Fig.1  Quantitative analysis of phospho-proteome upon ING5 overexpression. (A) Quantitative ratio (L/H) distribution of phosphorylated protein influenced by ING5 overexpression. The figure shows the protein ratio (log (protein ratio), with base = 2) of each differentially expressed protein. Proteins with upregulated phosphorylation are located right of zero of the x-axis, whereas proteins with downregulated phosphorylation are located left of zero of the x-axis. (B) Enrichment and clustering analysis of biological processes based on Gene Ontology annotation. (C) Enrichment and clustering analysis of molecular function based on Gene Ontology annotation. (D) Enrichment and clustering analysis of cellular components based on Gene Ontology annotation. (E) Enrichment and clustering analysis of KEGG pathways. (F) Enrichment and clustering analysis of protein domain. (G) ING5 participated in the modified protein interaction network of the pathway map05230 central carbon metabolism in cancer. The circle represents the protein; the size of the circle represents the number of interacting proteins, and the different colors represent the Q1–Q4 proteins. (H) Difference ratio information of all phosphorylation sites. The horizontal coordinate is the ratio value rank, and the vertical coordinate is the log2 ratio value. The Q1, Q2, Q3, and Q4 sites are distinguished by color, and the PDK1-Y163 site is separately marked.
Fig.2  Y163 is a new phosphorylation site in PDK1 upregulated by ING5 overexpression, which positively correlated with better prognosis in patients with lung cancer. (A) LC–MS/MS spectrum of peptide HNDVIPTMAQGVIEYK of PDK1. (B) Phosphorylation of PDK1 Y163 is upregulated in ING5-overexpressing lung cancer cells by Western blot. Phosphorylation of PDK1 Y376 shows no significant change between the control and high-ING5-expression group. Actin was used as an internal loading control. (C) Phosphorylation of PDK1 Y163 is downregulated in ING5-knockdown lung cancer cells. Phosphorylation of PDK1 Y376 shows no significant change between the control and ING5 knockdown groups. (D, E) Representative images of NSCLC and adjacent noncancerous tissues with p-PDK1 Y163 staining. Scale bar, 200 μmol/L and 50 μmol/L. (F) The expression of p-PDK1 Y163 in NSCLC was lower than that in adjacent noncancerous specific antibody (P < 0.001). (G) Kaplan–Meier overall survival (OS) analysis based on NSCLC patients with high and low p-PDK1 Y163 levels (HR = 0.596, 95% CI = 0.396–0.979, P = 0.041, log-rank test).
Clinicopathological variables Total (n) p-PDK1 (Y163) expression (IHC)
Low High χ2 P value
Age
< 60 45 20 25 0.085 0.771
≥ 60 53 22 31
Gender
Male 55 21 34 1.107 0.293
Female 43 21 22
Tumor location
Left lung 43 18 25 0.005 0.943
Right lung 54 23 31
Tumor invasion
T1 20 10 10 9.033 0.029*
T2 51 17 34
T3 22 10 12
T4 5 5 0
Lymph node metastases
N0 44 16 28 1.139 0.286
N1–N3 53 25 28
Metastasis
No 97 55 42 0.750 0.387
Yes 1 1 0
Clinical stage
I–II 52 19 33 2.398 0.122
III–IV 45 24 22
Pathological grade
Well and moderate 84 38 46 1.347 0.246
Poorly and not 14 4 10
Tab.1  Correlation of p-PDK1 Y163 expression with clinicopathological features of NSCLC
Fig.3  Y163 phosphorylation negatively regulates PDK1 kinase activity, and it is necessary for the ING5-reversed Warburg effect. (A) LC–MS/MS spectrum of peptide YHGHSMSDPGVSTR of PDHA1. Phosphorylation of PDHA1 S293 is downregulated by ING5 overexpression by quantitative phospho-proteomics. (B) ING5 overexpression decreased the phosphorylation of PDHA1 S293 by Western blot. Actin was used as an internal loading control. (C) ING5 overexpression (ING5) increased the enzyme activity of PDH. (D) ING5 overexpression enhanced cancer cell oxygen consumption. (E) ING5 overexpression downregulated HIF-1α and LDHA by Western blot. Actin was used as an internal loading control. (F) ING5 overexpression inhibited LDH activity. (G) ING5 overexpression decreased cancer cell lactic acid (LD) production. (H) Phosphorylation of PDK1 Y163 was decreased by Y163F mutation. ING5 overexpression—decreased PDHA1 S293 phosphorylation was impaired by PDK1 Y163F mutation. Actin was used as an internal loading control. (I) ING5 overexpression—increased PDH enzyme activity was abolished by PDK1 Y163F mutation. (J) ING5 overexpression—enhanced oxygen consumption was decreased by PDK1 Y163F mutation. (K) PDK1 Y163F phospho-dead mutation impaired the downregulating effects of ING5 overexpression on HIF-1α and LDHA levels. (L) ING5 overexpression—downregulated LDH activity was impaired by PDK1 Y163F mutation. (M) ING5 overexpression–decreased lactic acid production was impaired by PDK1 Y163F mutation. Data of (C, D, F, G, I, J, L, and M) are shown as the mean plus standard error of three independent experiments (n = 3 samples). Statistical analysis was performed by one-way ANOVA. *P < 0.05 and **P < 0.01 versus the NC group.
Fig.4  PDK1 Y163 phosphorylation is required for ING5-inhibited invasiveness of lung cancer cells in vitro and in vivo. Cells were divided into the following groups: (1) control A549 cells (control); (2) ING5-overexpressing cells (ING5); (3) ING5-overexpressing A549 cells with PDK1 knockdown (ING5-PDK1 KD); (4) ING5-PDK1 KD cells rescued with wild-type PDK1 (ING5-PDK1 WT); (5) ING5-PDK1 KD cells rescued with phospho-dead Y163F mutant PDK1 (ING5-PDK1 Y163F). (A) PDK1 Y163F mutation significantly increased the proliferation of lung cancer A549 cells with ING5 overexpression. (B) PDK1 Y163F mutation significantly increased colony formation of lung cancer A549 cells with ING5 overexpression. Representative pictures are shown. Colony numbers were quantified. (C) Wound-healing assay was performed to show the promoting effects of PDK1 Y163 mutation on the migration of A549 ING5-overexpressing cells. A scratch wound was made on the cell surface, and cells were photographed at 0 h, 8 h, 16 h, 20 h, and 24 h. Representative pictures are shown. (D) PDK1 Y163 mutation accelerated the Transwell migration of A549 ING5-overexpressing cells. The migrated cells were photographed (100× magnification). Representative pictures are shown. (E) PDK1 Y163 mutation promoted the Transwell invasive abilities of A549 ING5-overexpressing cells. The invaded cells were photographed (100× magnification). Representative pictures are shown. (F) Effects of PDK1 Y163 mutation on the expression of EMT markers in A549 ING5-overexpressing cells. Actin was used as an internal loading control. (G) Mice were injected subcutaneously with 5 × 106 lung cancer A549 cells. At the end of the experiment, mice were sacrificed and photographed. (H) Tumor growth curves revealed that the tumor volumes were significantly higher in mice with ING5-PDK1 Y163F cells. (I) Survival analysis showed that mice with ING5 OE cells had better overall survival than those with control cells (P = 0.0007), whereas mice with ING5-PDK1 Y163F cells had worse overall survival than those with ING5-PDK1 WT cells (P = 0.0023). (J) Mice were injected through the tail vein with 5 × 106 lung cancer A549 cells. On day 45 after tumor cell injection, mice were sacrificed, and the lungs were photographed. Gross images of the lungs showed lung-metastasized tumors in different groups. (K) Representative images of HE staining for lung tissues were provided (scale bar, 200 μm). (L) Tumor index of mice from different groups. (M) Lung weight of mice from different groups. Statistical analysis was performed using two-way ANOVA, followed by post-hoc tests. *P < 0.05 and **P < 0.01 versus the NC group.
Fig.5  TIE1 was identified as an anti-tumor TPK upregulated by ING5. (A) Results of the top 10 targeted genes were shown from a TPK siRNA library-based screening of invasion assay. (B) TIE1 mRNA expression in diverse tumor and normal tissues using the GEPIA2 database. Red, tumor samples; green, normal control sample. TIE1 expression was decreased in 11 tumors: LUAD (lung adenocarcinoma), BLCA (bladder urothelial carcinoma), BRCA (breast invasive carcinoma), CESC (cervical squamous cell carcinoma and endocervical adenocarcinoma), COAD (colon adenocarcinoma), KICH (kidney chromophobe), KIRP (kidney renal papillary cell carcinoma), LUSC (lung squamous cell carcinoma), THCA (thyroid carcinoma), UCEC (uterine corpus endometrial carcinoma), and UCS (uterine carcinosarcoma). (C) The mRNA expression level of TIE1 was analyzed in LUAD and normal tissues using TCGA samples. *P < 0.05, compared with normal tissues. (D) TIE1 protein expression levels in LUAD and normal tissue were analyzed using CPTAC samples. (E) Kaplan–Meier overall survival curves showed that the high expression level of TIE1 predicts longer survival. (F) Spearman correlation analysis showed that the expression of TIE1 was positively correlated with ING5 expression. (G, H) Representative images and results of Transwell migration assays of the TIE1-knockdown (sh-TIE1-1 and sh-TIE1-2) A549 control group and A549-ING5-OE control group. (I, J) Representative images and results of Transwell invasion assays of the TIE1-knockdown (sh-TIE1-1 and sh-TIE1-2) A549 control group and A549-ING5-OE control group. Data of (G−J) are shown as the mean plus standard error of three independent experiments (n = 3 samples).
Fig.6  TIE1 directly phosphorylates PDK1 at Y163, which is promoted by ING5. (A) PDK1 Y163 is specifically phosphorylated by rTIE1 by in vitro kinase assay. An antibody that specifically recognizes p-PDK1 Y163 was used. (B) Immunofluorescence assay showed the mitochondrial location of TIE1 in lung cancer cells. Representative images are presented (600×). (C) Mitochondrial localization of TIE1 in A549 cells was confirmed by transmission electron microscopy (TEM). Scale bars: 5.0 μm (left), 1.0 μm (middle), and 500 nm (right). (D, E) ING5 overexpression-promoted PDK1Y163 phosphorylation was impaired by TIE1 knockdown by Western blot. (F, G) Expression of TIE1 and p-PDK1Y163 in ING5-overexpressing or knockdown lung cancer cells by Western blot. (H) A graphic illustration of the proposed mechanism in this study. In brief, ING5 overexpression upregulates TIE1, which phosphorylates PDK1 at Y163, leading to the dephosphorylation-dependent activation of PDHA1 and subsequent increased oxidative phosphorylation. In addition, ING5 overexpression downregulates HIF-1α/LDHA1, thereby decreasing glycolysis.
1 M Yuan, Y Zhao, HT Arkenau, T Lao, L Chu, Q Xu. Signal pathways and precision therapy of small-cell lung cancer. Signal Transduct Target Ther 2022; 7(1): 187–204
https://doi.org/10.1038/s41392-022-01013-y
2 Y Doyon, C Cayrou, M Ullah, AJ Landry, V Côté, W Selleck, WS Lane, S Tan, XJ Yang, J Côté. ING tumor suppressor proteins are critical regulators of chromatin acetylation required for genome expression and perpetuation. Mol Cell 2006; 21(1): 51–64
https://doi.org/10.1016/j.molcel.2005.12.007
3 S Anwar, A Shamsi, T Mohammad, A Islam, MI Hassan. Targeting pyruvate dehydrogenase kinase signaling in the development of effective cancer therapy. Biochim Biophys Acta Rev Cancer 2021; 1876(1): 188568–188592
https://doi.org/10.1016/j.bbcan.2021.188568
4 S Cui, X Liao, C Ye, X Yin, M Liu, Y Hong, M Yu, Y Liu, H Liang, CY Zhang, X Chen. ING5 suppresses breast cancer progression and is regulated by miR-24. Mol Cancer 2017; 16(1): 89–100
https://doi.org/10.1186/s12943-017-0658-z
5 S Ghafouri-Fard, M Taheri, A Baniahmad. Inhibitor of growth factors regulate cellular senescence. Cancers (Basel) 2022; 14(13): 3107–3118
https://doi.org/10.3390/cancers14133107
6 J Archambeau, A Blondel, R Pedeux. Focus-ING on DNA integrity: implication of ING proteins in cell cycle regulation and DNA repair modulation. Cancers (Basel) 2019; 12(1): 58–76
https://doi.org/10.3390/cancers12010058
7 F Wang, AY Wang, C Chesnelong, Y Yang, A Nabbi, S Thalappilly, V Alekseev, K Riabowol. ING5 activity in self-renewal of glioblastoma stem cells via calcium and follicle stimulating hormone pathways. Oncogene 2018; 37(3): 286–301
https://doi.org/10.1038/onc.2017.324
8 G Ormaza, JA Rodríguez, de Opakua A Ibáñez, N Merino, M Villate, I Gorroño, M Rábano, I Palmero, M Vilaseca, R Kypta, MDM Vivanco, AL Rojas, FJ Blanco. The tumor suppressor ING5 is a dimeric, bivalent recognition molecule of the histone H3K4me3 mark. J Mol Biol 2019; 431(12): 2298–2319
https://doi.org/10.1016/j.jmb.2019.04.018
9 F Zhang, X Zhang, J Meng, Y Zhao, X Liu, Y Liu, Y Wang, Y Li, Y Sun, Z Wang, Q Mei, T Zhang. ING5 inhibits cancer aggressiveness via preventing EMT and is a potential prognostic biomarker for lung cancer. Oncotarget 2015; 6(18): 16239–16252
https://doi.org/10.18632/oncotarget.3842
10 XL Liu, XT Zhang, J Meng, HF Zhang, Y Zhao, C Li, Y Sun, QB Mei, F Zhang, T Zhang. ING5 knockdown enhances migration and invasion of lung cancer cells by inducing EMT via EGFR/PI3K/Akt and IL-6/STAT3 signaling pathways. Oncotarget 2017; 8(33): 54265–54276
https://doi.org/10.18632/oncotarget.17346
11 XL Liu, J Meng, XT Zhang, XH Liang, F Zhang, GR Zhao, T Zhang. ING5 inhibits lung cancer invasion and epithelial-mesenchymal transition by inhibiting the WNT/β-catenin pathway. Thorac Cancer 2019; 10(4): 848–855
https://doi.org/10.1111/1759-7714.13013
12 T Zhang, J Meng, X Liu, X Zhang, X Peng, Z Cheng, F Zhang. ING5 differentially regulates protein lysine acetylation and promotes p300 autoacetylation. Oncotarget 2018; 9(2): 1617–1629
https://doi.org/10.18632/oncotarget.22176
13 O Warburg. On the origin of cancer cells. Science 1956; 123(3191): 309–314
https://doi.org/10.1126/science.123.3191.309
14 D Hanahan, RA Weinberg. Hallmarks of cancer: the next generation. Cell 2011; 144(5): 646–674
https://doi.org/10.1016/j.cell.2011.02.013
15 RV Pusapati, A Daemen, C Wilson, W Sandoval, M Gao, B Haley, AR Baudy, G Hatzivassiliou, M Evangelista, J Settleman. mTORC1-dependent metabolic reprogramming underlies escape from glycolysis addiction in cancer cells. Cancer Cell 2016; 29(4): 548–562
https://doi.org/10.1016/j.ccell.2016.02.018
16 A Erdem, S Marin, DA Pereira-Martins, R Cortés, A Cunningham, MG Pruis, Boer B de, den Heuvel FAJ van, M Geugien, ATJ Wierenga, AZ Brouwers-Vos, EM Rego, G Huls, M Cascante, JJ Schuringa. The glycolytic gatekeeper PDK1 defines different metabolic states between genetically distinct subtypes of human acute myeloid leukemia. Nat Commun 2022; 13(1): 1105–1120
https://doi.org/10.1038/s41467-022-28737-3
17 L Lv, Q Lei. Proteins moonlighting in tumor metabolism and epigenetics. Front Med 2021; 15(3): 383–403
https://doi.org/10.1007/s11684-020-0818-1
18 A Cenigaonandia-Campillo, R Serna-Blasco, L Gómez-Ocabo, S Solanes-Casado, N Baños-Herraiz, LD Puerto-Nevado, JA Cañas, MJ Aceñero, J García-Foncillas, Ó Aguilera. Vitamin C activates pyruvate dehydrogenase (PDH) targeting the mitochondrial tricarboxylic acid (TCA) cycle in hypoxic KRAS mutant colon cancer. Theranostics 2021; 11(8): 3595–3606
https://doi.org/10.7150/thno.51265
19 SM Hong, YK Lee, I Park, SM Kwon, S Min, G Yoon. Lactic acidosis caused by repressed lactate dehydrogenase subunit B expression down-regulates mitochondrial oxidative phosphorylation via the pyruvate dehydrogenase (PDH)-PDH kinase axis. J Biol Chem 2019; 294(19): 7810–7820
https://doi.org/10.1074/jbc.RA118.006095
20 I Adant, M Bird, B Decru, P Windmolders, M Wallays, Witte P de, D Rymen, P Witters, P Vermeersch, D Cassiman, B Ghesquière. Pyruvate and uridine rescue the metabolic profile of OXPHOS dysfunction. Mol Metab 2022; 63: 101537–101550
https://doi.org/10.1016/j.molmet.2022.101537
21 S Imanaka, H Shigetomi, H Kobayashi. Reprogramming of glucose metabolism of cumulus cells and oocytes and its therapeutic significance. Reprod Sci 2022; 29(3): 653–667
https://doi.org/10.1007/s43032-021-00505-6
22 TN Seyfried, G Arismendi-Morillo, P Mukherjee, C Chinopoulos. On the origin of ATP synthesis in cancer. iScience 2020; 23(11): 101761–101781
https://doi.org/10.1016/j.isci.2020.101761
23 J Fan, C Shan, HB Kang, S Elf, J Xie, M Tucker, TL Gu, M Aguiar, S Lonning, H Chen, M Mohammadi, LM Britton, BA Garcia, M Alečković, Y Kang, S Kaluz, N Devi, Meir EG Van, T Hitosugi, JH Seo, S Lonial, M Gaddh, M Arellano, HJ Khoury, FR Khuri, TJ Boggon, S Kang, J Chen. Tyr phosphorylation of PDP1 toggles recruitment between ACAT1 and SIRT3 to regulate the pyruvate dehydrogenase complex. Mol Cell 2014; 53(4): 534–548
https://doi.org/10.1016/j.molcel.2013.12.026
24 V De Rosa, F Iommelli, C Terlizzi, E Leggiero, R Camerlingo, GG Altobelli, R Fonti, L Pastore, S Del Vecchio. Non-canonical role of PDK1 as a negative regulator of apoptosis through macromolecular complexes assembly at the ER-mitochondria interface in oncogene-driven NSCLC. Cancers (Basel) 2021; 13(16): 4133–4146
https://doi.org/10.3390/cancers13164133
25 V Calleja, M Laguerre, G de Las Heras-Martinez, PJ Parker, J Requejo-Isidro, B Larijani. Acute regulation of PDK1 by a complex interplay of molecular switches. Biochem Soc Trans 2014; 42(5): 1435–1440
https://doi.org/10.1042/BST20140222
26 T Hitosugi, J Fan, TW Chung, K Lythgoe, X Wang, J Xie, Q Ge, TL Gu, RD Polakiewicz, JL Roesel, GZ Chen, TJ Boggon, S Lonial, H Fu, FR Khuri, S Kang, J Chen. Tyrosine phosphorylation of mitochondrial pyruvate dehydrogenase kinase 1 is important for cancer metabolism. Mol Cell 2011; 44(6): 864–877
https://doi.org/10.1016/j.molcel.2011.10.015
27 Z Tang, C Li, B Kang, G Gao, C Li, Z Zhang. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res 2017; 45(W1): W98–W102
https://doi.org/10.1093/nar/gkx247
28 T Li, J Fan, B Wang, N Traugh, Q Chen, JS Liu, B Li, XS Liu. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res 2017; 77(21): e108–e110
https://doi.org/10.1158/0008-5472.CAN-17-0307
29 B Győrffy. Discovery and ranking of the most robust prognostic biomarkers in serous ovarian cancer. Geroscience 2023; 45(3): 1889–1898
https://doi.org/10.1007/s11357-023-00742-4
30 A Gautam, JC Waldrep, CL Densmore, N Koshkina, S Melton, L Roberts, B Gilbert, V Knight. Growth inhibition of established B16-F10 lung metastases by sequential aerosol delivery of p53 gene and 9-nitrocamptothecin. Gene Ther 2002; 9(5): 353–357
https://doi.org/10.1038/sj.gt.3301662
31 F Chen, Y Zhang, DS Chandrashekar, S Varambally, CJ Creighton. Global impact of somatic structural variation on the cancer proteome. Nat Commun 2023; 14(1): 5637
https://doi.org/10.1038/s41467-023-41374-8
32 E Atas, M Oberhuber, L Kenner. The implications of PDK1-4 on tumor energy metabolism, aggressiveness and therapy resistance. Front Oncol 2020; 10: 583217–583225
https://doi.org/10.3389/fonc.2020.583217
33 T Wang, F Chuffart, E Bourova-Flin, J Wang, J Mi, S Rousseaux, S Khochbin. Histone variants: critical determinants in tumour heterogeneity. Front Med 2019; 13(3): 289–297
https://doi.org/10.1007/s11684-018-0667-3
34 PW Szlosarek, S Lee, PJ Pollard. Rewiring mitochondrial pyruvate metabolism: switching off the light in cancer cells. Mol Cell 2014; 56(3): 343–344
https://doi.org/10.1016/j.molcel.2014.10.018
35 JE Burns, CD Hurst, MA Knowles, RM Phillips, SJ Allison. The Warburg effect as a therapeutic target for bladder cancers and intratumoral heterogeneity in associated molecular targets. Cancer Sci 2021; 112(9): 3822–3834
https://doi.org/10.1111/cas.15047
36 F Peng, JH Wang, WJ Fan, YT Meng, MM Li, TT Li, B Cui, HF Wang, Y Zhao, F An, T Guo, XF Liu, L Zhang, L Lv, DK Lv, LZ Xu, JJ Xie, WX Lin, EW Lam, J Xu, Q Liu. Glycolysis gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia. Oncogene 2018; 37(8): 1062–1074
https://doi.org/10.1038/onc.2017.368
37 F Dupuy, S Tabariès, S Andrzejewski, Z Dong, J Blagih, MG Annis, A Omeroglu, D Gao, S Leung, E Amir, M Clemons, A Aguilar-Mahecha, M Basik, EE Vincent, J St-Pierre, RG Jones, PM Siegel. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metab 2015; 22(4): 577–589
https://doi.org/10.1016/j.cmet.2015.08.007
38 JY Hsu, JY Chang, KY Chang, WC Chang, BK Chen. Epidermal growth factor-induced pyruvate dehydrogenase kinase 1 expression enhances head and neck squamous cell carcinoma metastasis via up-regulation of fibronectin. FASEB J 2017; 31(10): 4265–4276
https://doi.org/10.1096/fj.201700156R
39 Milić N Škorja, K Dolinar, K Miš, U Matkovič, M Bizjak, M Pavlin, M Podbregar, S Pirkmajer. Suppression of pyruvate dehydrogenase kinase by dichloroacetate in cancer and skeletal muscle cells is isoform specific and partially independent of HIF-1α. Int J Mol Sci 2021; 22(16): 8610–8635
https://doi.org/10.3390/ijms22168610
40 S Savant, Porta S La, A Budnik, K Busch, J Hu, N Tisch, C Korn, AF Valls, AV Benest, D Terhardt, X Qu, RH Adams, HS Baldwin, de Almodóvar C Ruiz, HR Rodewald, HG Augustin. The orphan receptor Tie1 controls angiogenesis and vascular remodeling by differentially regulating Tie2 in tip and stalk cells. Cell Rep 2015; 12(11): 1761–1773
https://doi.org/10.1016/j.celrep.2015.08.024
41 M Singhal, N Gengenbacher, S La Porta, S Gehrs, J Shi, M Kamiyama, DM Bodenmiller, A Fischl, B Schieb, E Besemfelder, S Chintharlapalli, HG Augustin. Preclinical validation of a novel metastasis-inhibiting Tie1 function-blocking antibody. EMBO Mol Med 2020; 12(6): e11164
https://doi.org/10.15252/emmm.201911164
42 PJ Zwiers, RM Jongman, T Kuiper, J Moser, RV Stan, JR Göthert, Meurs M van, ER Popa, G Molema. Pattern of tamoxifen-induced Tie2 deletion in endothelial cells in mature blood vessels using endo SCL-Cre-ERT transgenic mice. PLoS One 2022; 17(6): e0268986
https://doi.org/10.1371/journal.pone.0268986
43 EA Korhonen, A Murtomäki, SK Jha, A Anisimov, A Pink, Y Zhang, S Stritt, I Liaqat, L Stanczuk, L Alderfer, Z Sun, E Kapiainen, A Singh, I Sultan, A Lantta, VM Leppänen, L Eklund, Y He, HG Augustin, K Vaahtomeri, P Saharinen, T Mäkinen, K Alitalo. Lymphangiogenesis requires Ang2/Tie/PI3K signaling for VEGFR3 cell-surface expression. J Clin Invest 2022; 132(15): e155478
https://doi.org/10.1172/JCI155478
44 JH Yun, HM Lee, EH Lee, JW Park, CH Cho. Hypoxia reduces endothelial Ang1-induced Tie2 activity in a Tie1-dependent manner. Biochem Biophys Res Commun 2013; 436(4): 691–697
https://doi.org/10.1016/j.bbrc.2013.06.018
45 F Chen, J Chen, L Yang, J Liu, X Zhang, Y Zhang, Q Tu, D Yin, D Lin, PP Wong, D Huang, Y Xing, J Zhao, M Li, Q Liu, F Su, S Su, E Song. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol 2019; 21(4): 498–510
https://doi.org/10.1038/s41556-019-0299-0
46 GL Semenza. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest 2013; 123(9): 3664–3671
https://doi.org/10.1172/JCI67230
47 L Hulea, SP Gravel, M Morita, M Cargnello, O Uchenunu, YK Im, C Lehuédé, EH Ma, M Leibovitch, S McLaughlan, MJ Blouin, M Parisotto, V Papavasiliou, C Lavoie, O Larsson, M Ohh, T Ferreira, C Greenwood, G Bridon, D Avizonis, G Ferbeyre, P Siegel, RG Jones, W Muller, J Ursini-Siegel, J St-Pierre, M Pollak, I Topisirovic. Translational and HIF-1α-dependent metabolic reprogramming underpin metabolic plasticity and responses to kinase inhibitors and biguanides. Cell Metab 2018; 28(6): 817–832.e8
https://doi.org/10.1016/j.cmet.2018.09.001
48 A Brand, K Singer, GE Koehl, M Kolitzus, G Schoenhammer, A Thiel, C Matos, C Bruss, S Klobuch, K Peter, M Kastenberger, C Bogdan, U Schleicher, A Mackensen, E Ullrich, S Fichtner-Feigl, R Kesselring, M Mack, U Ritter, M Schmid, C Blank, K Dettmer, PJ Oefner, P Hoffmann, S Walenta, EK Geissler, J Pouyssegur, A Villunger, A Steven, B Seliger, S Schreml, S Haferkamp, E Kohl, S Karrer, M Berneburg, W Herr, W Mueller-Klieser, K Renner, M Kreutz. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab 2016; 24(5): 657–671
https://doi.org/10.1016/j.cmet.2016.08.011
49 D Mishra, D Banerjee. Lactate dehydrogenases as metabolic links between tumor and stroma in the tumor microenvironment. Cancers (Basel) 2019; 11(6): 750–770
https://doi.org/10.3390/cancers11060750
[1] FMD-23076-OF-ZT_suppl_1 Download
[2] FMD-23076-OF-ZT_suppl_2 Download
[1] Xiang Nie, Jiahui Fan, Yanwen Wang, Rong Xie, Chen Chen, Huaping Li, Dao Wen Wang. lncRNA ZNF593-AS inhibits cardiac hypertrophy and myocardial remodeling by upregulating Mfn2 expression[J]. Front. Med., 2024, 18(3): 484-498.
[2] Tianzhuo Wang, Huiying Guo, Lei Zhang, Miao Yu, Qianchen Li, Jing Zhang, Yan Tang, Hongquan Zhang, Jun Zhan. FERM domain-containing protein FRMD6 activates the mTOR signaling pathway and promotes lung cancer progression[J]. Front. Med., 2023, 17(4): 714-728.
[3] Xueqing Hu, Ujjwol Khatri, Tao Shen, Jie Wu. Progress and challenges in RET-targeted cancer therapy[J]. Front. Med., 2023, 17(2): 207-219.
[4] Jia Zhong, Hua Bai, Zhijie Wang, Jianchun Duan, Wei Zhuang, Di Wang, Rui Wan, Jiachen Xu, Kailun Fei, Zixiao Ma, Xue Zhang, Jie Wang. Treatment of advanced non-small cell lung cancer with driver mutations: current applications and future directions[J]. Front. Med., 2023, 17(1): 18-42.
[5] Jianhua Shi, Ying Cheng, Qiming Wang, Kai Li, Lin Wu, Baohui Han, Gongyan Chen, Jianxing He, Jie Wang, Haifeng Qin, Xiaoling Li. Anlotinib as third- or further-line therapy for short-term relapsed small-cell lung cancer: subgroup analysis of a randomized phase 2 study (ALTER1202)[J]. Front. Med., 2022, 16(5): 766-772.
[6] Shi-Yong Sun. Targeting apoptosis to manage acquired resistance to third generation EGFR inhibitors[J]. Front. Med., 2022, 16(5): 701-713.
[7] Yaru Tian, Hairong Tian, Xiaoyang Zhai, Hui Zhu, Jinming Yu. Bevacizumab in combination with pemetrexed and platinum for elderly patients with advanced non-squamous non-small-cell lung cancer: a retrospective analysis[J]. Front. Med., 2022, 16(4): 610-617.
[8] Xiang Wang, Minghui Wang, Lin Feng, Jie Song, Xin Dong, Ting Xiao, Shujun Cheng. Four-protein model for predicting prognostic risk of lung cancer[J]. Front. Med., 2022, 16(4): 618-626.
[9] Wenjing Zhou, Jing Zhang, Mingkun Yan, Jin Wu, Shuo Lian, Kang Sun, Baiqing Li, Jia Ma, Jun Xia, Chaoqun Lian. Orlistat induces ferroptosis-like cell death of lung cancer cells[J]. Front. Med., 2021, 15(6): 922-932.
[10] Ronghui Yang, Guoguang Ying, Binghui Li. Potential of electron transfer and its application in dictating routes of biochemical processes associated with metabolic reprogramming[J]. Front. Med., 2021, 15(5): 679-692.
[11] Na Qin, Yuancheng Li, Cheng Wang, Meng Zhu, Juncheng Dai, Tongtong Hong, Demetrius Albanes, Stephen Lam, Adonina Tardon, Chu Chen, Gary Goodman, Stig E. Bojesen, Maria Teresa Landi, Mattias Johansson, Angela Risch, H-Erich Wichmann, Heike Bickeboller, Gadi Rennert, Susanne Arnold, Paul Brennan, John K. Field, Sanjay Shete, Loic Le Marchand, Olle Melander, Hans Brunnstrom, Geoffrey Liu, Rayjean J. Hung, Angeline Andrew, Lambertus A. Kiemeney, Shan Zienolddiny, Kjell Grankvist, Mikael Johansson, Neil Caporaso, Penella Woll, Philip Lazarus, Matthew B. Schabath, Melinda C. Aldrich, Victoria L. Stevens, Guangfu Jin, David C. Christiani, Zhibin Hu, Christopher I. Amos, Hongxia Ma, Hongbing Shen. Comprehensive functional annotation of susceptibility variants identifies genetic heterogeneity between lung adenocarcinoma and squamous cell carcinoma[J]. Front. Med., 2021, 15(2): 275-291.
[12] Jiahui Xu, Qianqian Wang, Elaine Lai Han Leung, Ying Li, Xingxing Fan, Qibiao Wu, Xiaojun Yao, Liang Liu. Compound C620-0696, a new potent inhibitor targeting BPTF, the chromatin-remodeling factor in non-small-cell lung cancer[J]. Front. Med., 2020, 14(1): 60-67.
[13] Zhao Zhang, Jun Jiang, Xiaodong Wu, Mengyao Zhang, Dan Luo, Renyu Zhang, Shiyou Li, Youwen He, Huijie Bian, Zhinan Chen. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy[J]. Front. Med., 2019, 13(1): 57-68.
[14] Kohei Miyazono, Yoko Katsuno, Daizo Koinuma, Shogo Ehata, Masato Morikawa. Intracellular and extracellular TGF-β signaling in cancer: some recent topics[J]. Front. Med., 2018, 12(4): 387-411.
[15] Dalin Zhang, Liwei Qu, Bo Zhou, Guizhen Wang, Guangbiao Zhou. Genomic variations in the counterpart normal controls of lung squamous cell carcinomas[J]. Front. Med., 2018, 12(3): 280-288.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed