China Oncology ›› 2024, Vol. 34 ›› Issue (11): 987-997.doi: 10.19401/j.cnki.1007-3639.2024.11.001
• Article • Previous Articles Next Articles
WU Zhibai1(), XU Guiqin1, ZHANG Li1, YANG Zhaojuan1, LIU Yun1, JIAO Kun1,2, CHEN Zehong1, XU Chen1, ZUO You1, ZHENG Ningqian1, YE Zhiqian1, LIU Yongzhong1(
)
Received:
2024-06-26
Revised:
2024-09-12
Online:
2024-11-30
Published:
2024-12-11
Contact:
LIU Yongzhong
Share article
CLC Number:
WU Zhibai, XU Guiqin, ZHANG Li, YANG Zhaojuan, LIU Yun, JIAO Kun, CHEN Zehong, XU Chen, ZUO You, ZHENG Ningqian, YE Zhiqian, LIU Yongzhong. Mechanism study of KCMF1 promoting proliferation and NF-κB signaling transduction in colorectal cancer cells[J]. China Oncology, 2024, 34(11): 987-997.
Fig. 1
High expression of KCMF1 is associated with poor prognosis of patients with CRC A: mRNA levels of KCMF1 in different types of tumor and normal tissues from the TCGA and GTEx databases. TPM: Transcripts per million; NS: No significance; *: P<0.05, assessed by Student’s t-tests; **: P<0.01, assessed by Student’s t-tests; ***: P<0.001, assessed by Student’s t-tests. B: The correlation between KCMF1 expression and the prognosis of CRC patients in GSE39582 dataset from the GEO database. **: P<0.01, assessed by log-rank tests. C: Representative images of immunohistochemical staining for KCMF1 in CRC tissues and their paired adjacent tissues. D: Immunohistochemical staining scores in the 90 colorectal tumor tissues and their paired adjacent tissues. ***: P<0.001, assessed by Student’s t-tests. E: The survival probability of 90 patients with high or low expression of KCMF1 in colorectal tumors; The KCMF1 scores are ranked by High and Low, with KCMF1-High group (n=45) and KCMF1-Low group (n=45). ***: P<0.001, assessed by log-rank tests. F: The correlation between KCMF1 expression and TNM stage in patients with CRC. *: P<0.05, assessed by Student-Newman-Keuls."
Fig. 2
Effects of KCMF1 knockdown on the proliferation of CRC cells in vitro A: Expression level of KCMF1 protein in HCT116 and HCT15 cells with or without shKCMF1 expression. B, C: The proliferation of HCT116 and HCT15 cells with or without shKCMF1 expression was assessed by MTT assay (B) and colony formation assay (C). ***: P<0.001, assessed by Student’s t-tests. D: Expression levels of the apoptosis-related proteins in HCT116 and HCT15 cells with or without shKCMF1. E: Flow cytometry analysis of the percentage of apoptotic cells in the indicated cells treated with or without Z-VAD-FMK (20 µm) for 48 h. NS: No significance; ***; P<0.001, assessed by Student-Newman-Keuls. F: Expression levels of the cell cycle-related proteins in HCT116 and HCT15 cells with or without shKCMF1. G: Flow cytometry analysis of the cell cycle distribution of HCT116 and HCT15 cells with or without shKCMF1. **: P<0.01, assessed by Student-Newman-Keuls; ***: P<0.001, assessed by Student-Newman-Keuls."
Fig. 3
Effects of KCMF1 knockdown on the signaling pathways in human CRC cells A: Volcano plots show differentially expressed genes affected by KCMF1 knockdown in HCT116 cells. The significant different genes are determined with the following criteria: |Log2 FC|>1 and P<0.05. FC: Fold change. B: GO analysis of the down-regulated genes in HCT116 cells with KCMF1 knockdown. P<0.05 and Log2 FC≤-1 were used as the criteria to define down-regulated genes. C: Enrichment analysis of the KCMF1-regulated genes with HALLMARK gene signatures, the significantly enriched signatures were ranked by P value, the bar plot shows the top 10 signatures. D: GSEA of the transcriptional profiles of KCMF1-koncokdown HCT116 cells and control cells with the TNFα-NF-κB signature. E: Spearman correlation analysis was performed between BIOCARTA_NFKB_PATHWAY and KCMF1 expression based on TCGA database cohort. F: The TCGA cohort were classified into four groups by both KCMF1 expression and NF-κB activity and subjected to Kaplan-Meier overall survival analysis. The activity of NF-κB signaling was determined by the GSVA score of the gene set BIOCARTA_NFKB_SIGNALING. **: P<0.01, assessed by log-rank tests."
Fig. 4
Knockdown of KCMF1 inhibits the activation of the NF-κB signaling in human CRC cells A: mRNA levels of KCMF1, BCL-XL, XIAP and CIAP in HCT116 and HCT15 cells with or without KCMF1 knockdown. *: P<0.05, assessed by Student-Newman-Keuls; **: P<0.01, assessed by Student-Newman-Keuls; ***: P<0.001, assessed by Student-Newman-Keuls. B: Expression of the phosphorylated proteins related to NF-κB signaling in HCT116 and HCT15 cells with or without KCMF1 knockdown. C: Activity of the NF-κB-reporter in HCT116 and HCT15 cells with KCMF1 knockdown. **: P<0.01, assessed by Student-Newman-Keuls; ***: P<0.001, assessed by Student-Newman-Keuls. D: Immunofluorescence of p65 (green) in HCT116 and HCT15 cells treated with TNFα (10 ng/mL) for 5 min after KCMF1 knockdown. DAPI stains nuclear DNA (blue). Scale=20 microns. E: Quantification of p65 nuclear and cytosolic localization was assessed in the indicated cells, as shown by a ratio of nuclear to cytoplasmic fluorescence. **: P<0.01, assessed by Student-Newman-Keuls. F: p65 cellular localization in HCT116 and HCT15 cells with or without KCMF1 knockdown."
[1] | SUNG H, FERLAY J, SIEGEL R L, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71(3): 209-249. |
[2] |
GUINNEY J, DIENSTMANN R, WANG X, et al. The consensus molecular subtypes of colorectal cancer[J]. Nat Med, 2015, 21(11): 1350-1356.
doi: 10.1038/nm.3967 pmid: 26457759 |
[3] | LAURENT-PUIG P, AGOSTINI J, MALEY K. Colorectal oncogenesis[J]. Bull Cancer, 2010, 97(11): 1311-1321. |
[4] | TAIXIANG W, MUNRO A J, GUANJIAN L. Chinese medical herbs for chemotherapy side effects in colorectal cancer patients[J]. Cochrane Database Syst Rev, 2005, 2005(1): CD004540. |
[5] | RONNEKLEIV-KELLY S M, KENNEDY G D. Management of stage Ⅳ rectal cancer: palliative options[J]. World J Gastroenterol, 2011, 17(7): 835-847. |
[6] | XIE Y H, CHEN Y X, FANG J Y. Comprehensive review of targeted therapy for colorectal cancer[J]. Signal Transduct Target Ther, 2020, 5(1): 22. |
[7] |
GIANNAKIS M, MU X J, SHUKLA S A, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma[J]. Cell Rep, 2016, 17(4): 1206.
doi: S2211-1247(16)31395-X pmid: 27760322 |
[8] |
HERSHKO A, CIECHANOVER A. The ubiquitin system[J]. Annu Rev Biochem, 1998, 67: 425-479.
pmid: 9759494 |
[9] |
SWATEK K N, KOMANDER D. Ubiquitin modifications[J]. Cell Res, 2016, 26(4): 399-422.
doi: 10.1038/cr.2016.39 pmid: 27012465 |
[10] | QI J F, RONAI Z A. Dysregulation of ubiquitin ligases in cancer[J]. Drug Resist Updat, 2015, 23: 1-11. |
[11] |
BEILKE S, OSWALD F, GENZE F, et al. The zinc-finger protein KCMF1 is overexpressed during pancreatic cancer development and downregulation of KCMF1 inhibits pancreatic cancer development in mice[J]. Oncogene, 2010, 29(28): 4058-4067.
doi: 10.1038/onc.2010.156 pmid: 20473331 |
[12] |
HONG J H, KAUSTOV L, COYAUD E, et al. KCMF1 (potassium channel modulatory factor 1) Links RAD6 to UBR4 (ubiquitin N-recognin domain-containing E3 ligase 4) and lysosome-mediated degradation[J]. Mol Cell Proteomics, 2015, 14(3): 674-685.
doi: 10.1074/mcp.M114.042168 pmid: 25582440 |
[13] | HEO A J, KIM S B, JI C H, et al. The N-terminal cysteine is a dual sensor of oxygen and oxidative stress[J]. Proc Natl Acad Sci U S A, 2021, 118(50): e2107993118. |
[14] | CERVIA L D, SHIBUE T, BORAH A A, et al. A ubiquitination cascade regulating the integrated stress response and survival in carcinomas[J]. Cancer Discov, 2023, 13(3): 766-795. |
[15] | SINGH A, CHOUDHURY S D, SINGH P, et al. Disruption in networking of KCMF1 linked ubiquitin ligase impairs autophagy in CD8+ memory T cells of patients with renal cell carcinoma[J]. Cancer Lett, 2023, 564: 216194. |
[16] | JANG J H. FIGC, a novel FGF-induced ubiquitin-protein ligase in gastric cancers[J]. FEBS Lett, 2004, 578(1/2): 21-25. |
[17] | XI Y, XU P F. Global colorectal cancer burden in 2020 and projections to 2040[J]. Transl Oncol, 2021, 14(10): 101174. |
[18] | YANG Y, WANG H Y, CHEN Y K, et al. Current status of surgical treatment of rectal cancer in China[J]. Chin Med J, 2020, 133(22): 2703-2711. |
[19] |
VARLAND S, SILVA R D, KJOSÅS I, et al. N-terminal acetylation shields proteins from degradation and promotes age-dependent motility and longevity[J]. Nat Commun, 2023, 14(1): 6774.
doi: 10.1038/s41467-023-42342-y pmid: 37891180 |
[20] | ZOU J, MI L, YU X F, et al. Interaction of 14-3-3σ with KCMF1 suppresses the proliferation and colony formation of human colon cancer stem cells[J]. World J Gastroenterol, 2013, 19(24): 3770-3780. |
[21] |
SAKAMOTO K, MAEDA S, HIKIBA Y, et al. Constitutive NF-kappaB activation in colorectal carcinoma plays a key role in angiogenesis, promoting tumor growth[J]. Clin Cancer Res, 2009, 15(7): 2248-2258.
doi: 10.1158/1078-0432.CCR-08-1383 pmid: 19276252 |
[22] |
RAJITHA B, BELALCAZAR A, NAGARAJU G P, et al. Inhibition of NF-κB translocation by curcumin analogs induces G0/G1 arrest and downregulates thymidylate synthase in colorectal cancer[J]. Cancer Lett, 2016, 373(2): 227-233.
doi: 10.1016/j.canlet.2016.01.052 pmid: 26850372 |
[23] |
JANI T S, DEVECCHIO J, MAZUMDAR T, et al. Inhibition of NF-kappaB signaling by quinacrine is cytotoxic to human colon carcinoma cell lines and is synergistic in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or oxaliplatin[J]. J Biol Chem, 2010, 285(25): 19162-19172.
doi: 10.1074/jbc.M109.091645 pmid: 20424169 |
[24] |
SENFT D, QI J F, RONAI Z A. Ubiquitin ligases in oncogenic transformation and cancer therapy[J]. Nat Rev Cancer, 2018, 18(2): 69-88.
doi: 10.1038/nrc.2017.105 pmid: 29242641 |
[25] |
HOU Y Z, MOREAU F, CHADEE K. PPARγ is an E3 ligase that induces the degradation of NFκB/p65[J]. Nat Commun, 2012, 3: 1300.
doi: 10.1038/ncomms2270 pmid: 23250430 |
[26] | JI J X, DING K K, LUO T, et al. TRIM22 activates NF-κB signaling in glioblastoma by accelerating the degradation of IκBα[J]. Cell Death Differ, 2021, 28(1): 367-381. |
[27] |
DENG L, WANG C, SPENCER E, et al. Activation of the IkappaB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain[J]. Cell, 2000, 103(2): 351-361.
doi: 10.1016/s0092-8674(00)00126-4 pmid: 11057907 |
Viewed | ||||||
Full text |
|
|||||
Abstract |
|
|||||
沪ICP备12009617
Powered by Beijing Magtech Co. Ltd