Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Research

CUL4B-DDB1-COP1-mediated UTX downregulation promotes colorectal cancer progression

verfasst von: Dakui Luo, Min Chen, Qingguo Li, Kangjunjie Wang, Kaihua Wang, Junqiang Li, Guoxiang Fu, Zezhi Shan, Qi Liu, Yufei Yang, Lei Liang, Yanlei Ma, Yi Qin, Jun Qin, Daming Gao, Xinxiang Li

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

Abstract

Background

UTX (encoded by KDM6A), a histone demethylase for H3K27me2/3, is frequently mutated in human cancers. However, its functional and regulatory mechanisms in colorectal cancer (CRC) remain unclear.

Methods

Immunohistochemistry staining was used to investigate the clinical relevance of UTX in CRC. Additionally, we generated a spontaneous mouse CRC model with conditional Utx knockout to explore the role of UTX in the colorectal tumorigenesis. Post-translational regulation of UTX was determined by co-immunoprecipitation and immunoblot analyses.

Results

Herein, we identify that downregulation of UTX, mediated by the Cullin 4B-DNA Damage Binding Protein-1-Constitutive Photomorphogenesis Protein 1 (CUL4B-DDB1-COP1) complex, promotes CRC progression. Utx deletion in intestinal epithelial cells enhanced the susceptibility to tumorigenesis in AOM/DSS-induced spontaneous mouse CRC model. However, this effect is primarily alleviated by GSK126, an inhibitor of histone methyltransferase EZH2. Mechanistically, EMP1 and AUTS2 are identified as putative UTX target genes mediating UTX functions in limiting intestinal tumorigenesis. Notably, the CUL4B-DDB1-COP1 complex is identified as the functional E3 ligase responsible for targeting UTX for degradation in CRC cells. Thus, Cop1 deficiency in mouse intestinal tissue results in UTX accumulation and restricts tumorigenesis. Furthermore, patient cohort analysis reveals that UTX expression is negatively correlated with clinical stage, favorable disease outcomes, and COP1 expression.

Conclusions

In the current study, the tumor suppressor function and regulation of UTX in CRC provide a molecular basis and the rationale to target EZH2 in UTX-deficient CRC.
Begleitmaterial
Additional file 5: Figure S4. CRL4-COP1 E3 complex modulates UTX stability. A IB analysis of WCL and IP from HEK293T cells co-transfected with HA-UTX and different Cullin family constructs. Cells were treated with MG132 (20 µM) for 12 h before they were harvested. B IB analysis of WCL from HEK293T cells co-transfected with Flag-UTX and shCUL1 or shCUL4B. C IB analysis of WCL from HCT116 cells transfected with indicated siRNA targeting different Cullins. D IB analysis of WCL derived from HEK293T cells co-transfected with various doses of Flag-CUL4B together with HA-UTX constructs. Cells were treated with or without MG132, as indicated before they were harvested. E IB analysis of WCL derived from HEK293T cells co-transfected with various doses of Flag-DDB1 and HA-UTX constructs. Cells were treated with or without MG132, as indicated before they were harvested. F IB analysis of WCL derived from HEK293T cells co-transfected with various doses of Flag-DET1 and HA-UTX constructs. Cells were treated with or without MG132, as indicated before they were harvested. G IB analysis of WCL from HCT116 cells transfected with indicated siRNA targeting different CUL4B adaptor E3 ligases. H IB analysis of WCL and IP from HEK293T cells co-transfected with HA-UTX and CUL4B complex. Cells were treated with MG132 (20 µM) for 12 h before they were harvested. I IB analysis of WCL derived from HEK293T cells co-transfected with HA-UTX and Flag-COP1 (WT or C136A/C139A or △Ring). J and K IB (H) and qPCR analysis (I) of UTX expression in HCT116 cells infected with lentiviruses expressing EV or Flag-COP1. L and M IB (J) and qPCR analysis (K) of UTX expression in LoVo cells infected with lentiviruses expressing EV or Flag-COP1. Data information: In (K and M), data are presented as mean±SEM (two-tailed Student’s t-test). ns, non-significance, ***P<0.001
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40164-023-00440-z.
Dakui Luo, Min Chen and Qingguo Li contributed equally to this work.
The original online version of this article was revised: The grants acknowledgement in Funding section is revised.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CRC
Colorectal cancer
UTX
The ubiquitously transcribed tetratricopeptide repeat on the X chromosome
CRL
Cullin-RING Ligase
COP1
Constitutive photomorphogenic 1
TMA
Tissue microarray
WT
Wild type
FUSCC
Fudan University Shanghai Cancer Center
TNM
Tumor-node-metastasis
AOM
Azoxymethane
IP
Immunoprecipitation
DSS
Dextran sodium sulfate
TCGA
The Cancer Genome Atlas
GTEx
Genotype-tissue expression
GEO
Gene Expression Omnibus
IHC
Immunohistochemistry
OS
Overall survival
DFS
Disease-free survival

Background

Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second cause of cancer-related deaths among 36 cancer types globally in 2020 [1]. The initiation and progression of CRC are commonly regarded as a complicated and heterogeneous process involving genetic mutations [2] and dysregulated epigenetic modifications [3, 4]. Histone methylation, a reversible modification associated with gene activation or silencing, is regulated by specific methyltransferases and demethylases. Global changes in histone methylation have been well-established in carcinogenesis and identified as prognostic biomarkers in a spectrum of human tumors, including CRC [58].
The ubiquitously transcribed tetratricopeptide repeat on the X chromosome (UTX, encoded by KDM6A located on the X chromosome) is a histone demethylase that catalyzes the removal of H3K27me2/3, promoting the transcription of target genes. UTX is involved in a variety of biological processes, comprising homeotic gene expression, embryonic development, and cellular reprogramming [9]. UTY, a paralog sharing 83% sequence similarity with UTX, lacks detectable histone demethylase activity [10]. Interestingly, the loss of UTX during the development could be compensated by UTY, suggesting the function of UTX during embryonic development might be independent of its demethylase activity between E9.5 and E13.5 [11]. Recent studies have identified UTX as a highly mutated tumor suppressor, with reduced expression observed in several human malignancies [1217]. It has also been recognized as an “escape from X-inactivation tumor suppressor”, with a distinct dosage effect of UTX copy number observed during tumorigenesis [17]. The mutation or deletion of both copies of UTX is required for the dysfunction of this tumor suppressor in females, while loss of one copy of UTX achieves the same effect in males. UTY provides limited compensation for UTX loss during tumorigenesis, suggesting that the tumor suppressor function of UTX is demethylase activity-dependent. Accumulating evidence indicated that the oncogenic effect of UTX loss is mediated mainly through an increase in the EZH2 level in lung cancer [16], and UTX loss sensitizes cells to EZH2 inhibition in multiple myeloma [18]. Although a previous report suggested that UTX promotes CRC progression [19], the functional and regulatory mechanisms of UTX in CRC is largely unclear.
The ubiquitin–proteasome system is a significant pathway that precisely governs protein stability [20]. Among the Cullin-RING Ligase (CRL) family [21], a typical CRL4 ligase is composed of a scaffold Cullin (CUL4A or CUL4B) protein, a linker protein-DNA Damage Binding Protein-1 (DDB1), a substrate recognition adaptor (DCAF), and a ring protein with intrinsic E3 activity (RBX1 or RBX2) [22]. The substrate specificity of CRL4 is mainly dependent on the DCAF protein loaded on the specific CRL4 complex. Constitutive photomorphogenic 1 (COP1, also known as RFWD2) was initially identified as a negative regulator of photomorphogenesis in Arabidopsis [23], and its functions have been extensively studied in the context of light signaling in plants [24]. Notably, mammalian COP1 possesses intrinsic E3 ubiquitin ligase activity, enabling it to either promote protein ubiquitination or serve as a DCAF protein that forms a CRL4 ligase complex to ubiquitinate substrates [25]. Intriguingly, both classic oncoprotein and tumor suppressor proteins, such as c-Jun, ETS transcription factors, and P53, have been identified as ubiquitination-degradation substrates of COP1, indicating a complex and context-dependent role of COP1 in cancer [2628]. However, the role of COP1 in CRC has not yet been reported.
In the current study, we found that UTX is negatively correlated with clinical stage, and low UTX expression is associated with poor patient survival. Subsequently, we validated the tumor suppressor role of UTX in CRC by specifically depleting the Utx gene in intestinal epithelial cells and inducing de novo colorectal tumorigenesis. Our mechanistic study showed that UTX loss promoted colorectal oncogenesis partially via transcriptionally repression of EMP1 and AUTS2. Experiments performed with mouse CRC model and human CRC organoids further suggested that UTX deficiency confers enhanced sensitivity to EZH2 inhibitor GSK126 in CRC. Next, we certified that UTX is subject to CRL4-COP1 complex-mediated degradation, partially explaining the oncogenic role of COP1 in CRC.

Methods

Human CRC tissue microarray (TMA) construction

A human TMA was obtained from CRC patients who underwent radical surgery between January 2007 and November 2009 at Fudan University Shanghai Cancer Center (FUSCC), Shanghai, China. Human CRC TMA was carried out as described previously [29]. The clinicopathological characteristics of patients, such as age, gender, tumor location, histological type, tumor-node-metastasis (TNM) stage, grade, venous and nervous invasion, pretreatment serum CEA level, and survival data, were collected from the FUSCC database.

Generation of genetically-modified mice

All mice were raised in a specific pathogen-free environment at the Shanghai Institute of Biochemistry and Cell Biology. Utxf/f and Villin-Cre mice were provided by Dr. Charlie Degui Chen and Dr. Anning Lin (Shanghai Institute of Biochemistry and Cell Biology, CAS), respectively. As described previously [15], the exons 11 to 14 of Utx were deleted using a targeting vector, which disrupts the Utx H3K27 demethylase domain by triggering a frameshift mutation. Cop1f/f mice were purchased from Laboratory Animal Center, East China Normal University. Utx−/y or Utx−/+ mice were generated by crossing Utxf/f mice with Villin-Cre mice, while Utx−/− mice were embryonic lethal. Cop1−/+ mice were generated by crossing Cop1f/f mice with Villin-Cre mice, while Cop1−/− mice were embryonic lethal. All mice were maintained in a C57BL/6 background. The primers for genotyping are listed in Additional file 1: Table S1.

Induction of colitis, CRC, and treatments

We combined the carcinogen azoxymethane (AOM) treatment with repeated administration of dextran sodium sulfate (DSS) in drinking water to induce colorectal tumors. wild-type (WT), Utx−/y, and Utx−/+ mice were used for AOM/DSS treatment. 8–10-week-old mice were injected intraperitoneally with AOM (10 mg/kg, Sigma, A5486). After one week, DSS was solubilized into the drinking water to feed mice for 5 days (2%, MP Biomedicals, 160,110). Subsequently, regular drinking water was given for 14 days. The DSS treatment was repeated for another two cycles. Body weights were recorded during the DSS treatment. The mice were sacrificed 90 days after the AOM injection, and colorectums were obtained for further analysis. Gross inspection, total tumor number, tumor burden, and tumor size were recorded. The samples were fixed as ‘‘swiss-rolls,’’ and a histopathological examination was conducted.
For drug treatment, 2 weeks after AOM/DSS treatment, WT and Utx−/y mice were randomized into two groups. Then, the mice were treated with GSK126 (50 mg/kg, dissolved in SBE-β-CD) or vehicle control daily for additional 2 weeks before being sacrificed for analysis.

Immunoblotting (IB) and immunoprecipitation (IP)

Cells were lysed using EBC lysis buffer (50 mM Tris–HCl, pH 8.0, 120 mM NaCl, 0.5% Nonidet P-40) with protease inhibitors (1:100, Selleck) and phosphatase inhibitors (1:100, Selleck). Intestinal/tumor tissues from human or mice were lysed using Minute™ Total Protein Extraction Kit (Invent Biotechnologies, Inc.) with protease inhibitors (1:100, Selleck) and phosphatase inhibitors (1:100, Selleck). The protein concentrations of lysates were determined using the Bio-Rad protein assay kit on a spectrophotometer (Thermo Scientific). An equivalent of 30 μg total lysate from each sample was used for IB. The cell lysates were incubated with anti-FLAG M2 agarose beads or anti-HA agarose beads for 2–4 h for IP. The pellets were washed with NETN buffer (20 mM Tris–HCl, pH 8.0, 100 mM NaCl, 0.5% Nonidet P-40, 1 mM EDTA) before analysis by SDS-PAGE and IB with specific antibodies.

Public database analysis

Datasets from The Cancer Genome Atlas (TCGA) (https://​portal.​gdc.​cancer.​gov), Genotype-tissue expression (GTEx) (https://​www.​gtexportal.​org/​home/​index.​html) and the Gene Expression Omnibus (GEO, https://​www.​ncbi.​nlm.​nih.​gov/​sites/​GDSbrowser?​acc=​GDS2947) were utilized to determine COP1 mRNA expression in colon cancer, colorectal adenomas and normal colon tissues.

Immunohistochemistry (IHC)

IHC staining was performed with indicated antibodies using the standard protocol. The TMA was incubated with an anti-UTX antibody (1:400 dilution, 33510S, Cell Signaling Technology) and an anti-COP1 antibody (1:100 dilution, A300-894A, Bethyl). Mouse tissues were incubated with anti-UTX antibody (1:100 dilution, GTX121246, GeneTex), anti-COP1 antibody (1:100 dilution, A300-894A, Bethyl), Ki67 (1:100 dilution, ab15580, Abcam), anti-EZH2 antibody (1:200 dilution, ab191080, Abcam), and anti-H3K27me3 antibody (1:100 dilution, ab192985, Abcam; 1:100 dilution, A2363, Abclonal), respectively, as indicated. The IHC stained tissue sections were scored by two independent pathologists blinded to the clinicopathological features. The proportion of staining was graded as 0 (< 5%), 1 (5–25%), 2 (26–50%), 3 (51–75%), and 4 (> 75%) based on the percentages of the positive staining areas vs. the whole area. The staining intensity was graded as 0 (negative), 1 (weak), 2 (medium,) or 3 (strong). The immunoreactivity score (IRS) was calculated by multiplying the score of staining proportion with staining intensity. IRS ≤ 4 was considered as low, and > 4 was considered as high.

Cell proliferation, colony formation, and soft agar colony formation assays

HCT116 cells were seeded into 96-well plates at a density of 3000 cells/well. Then, the cells were incubated with CCK-8 reagent (C0039, Beyotime Biotechnology, Shanghai, China) for 2 h, and absorbance was measured at 450 nm. For colony formation assay, HCT116 cells were seeded in the 96-well plates with 500 cells/well and incubated for 10 days. For the soft agar colony formation assay, 6-well plates were covered with lower agar (1.5 mL, 0.8%), and 500 cells were seeded in the upper agar (1.5 mL, 0.4%). After 2 weeks, the colonies were stained with 1% crystal violet (Beyotime).

ChIP-seq assay and data analysis

The distal colorectum samples of 2-month-old WT and Utx−/y mice (each sample mixed with three animals) were collected for ChIP-seq analysis service by Active Motif Inc. using the antibody against H3K27me3 (Active Motif). The data were deposited in the NCBI’s SRA (Accession: PRJNA752794).

RNA-seq analysis

The distal colorectum mRNAs from 2-month-old WT mice and Utx−/y mice (each RNA sample mixed with three animals) were obtained for transcriptome analysis service by Sangon Biotech. The data were deposited in the NCBI’s SRA (Accession: PRJNA734666).

siRNA screening for putative target genes

siRNA-based screening was performed for the selected 55 genes in HCT116 cells. The siRNAs were selected from Dharmacon Human ON-TARGET plus siRNA Library. To eliminate the off-target effects, four independent siRNAs (SMARTpools) were used for each gene. siNC and siTOX were selected as the negative and positive controls in the screening assay. 0.15 μL lipofectamine RNAiMAX reagent (Thermo) was added to the siRNA-loaded 96-well plates using Multidrop Combi (Thermo). Then, HCT116 cells were seeded in the 96-well plates at a density of 1500 cells/well. The number of cells was counted in the following 4 days using Ensight (Perkin Elmer). Subsequently, CTG (G756A, Promega) was added and measured on an Envision plate reader (Perkin Elmer).

Total RNA extraction and quantitative PCR

Total RNA was extracted from cells or tissues using TRIzol (Invitrogen) according to the manufacturer’s instructions. The cDNA was synthesized using ABScript II RT Master Mix (ABclonal Technology). Quantitative PCR was performed using a SYBR.
Kit (Takara) according to the manufacturer’s protocol. Human GAPDH or mouse β-Actin was used as an internal control. The primer sequences are listed in Additional file 1: Table S1.

ChIP-qPCR assays

The ChIP assay was performed using truChIP Chromatin Shearing Kit (Covaris) and EZ-ChIP kit (Millipore). The procedure was according to the kit instruction manual provided by the manufacturer. Primer sequences can be found in Additional file 1: Table S1.

Plasmid construction

HA-UTX and different mutants were cloned into pcDNA3.1 vector. FLAG-UTX was generated by inserting UTX cDNA into pCMV-FLAG and pRLenti-CMV-MCS-3FLAG-PGK-Puro vectors. The Flag-tagged of DET1, DDB1, RBX1, Cullin1, Cullin2, Cullin3, Cullin4A, Cullin4B, and Cullin5 were generated by inserting cDNAs into pCMV-FLAG vector. The Flag-tagged and HA-tagged coding sequence of human COP1 WT or the relevant isoforms and the mutants were inserted into the lentiviral vector pLex-MCS-CMV-puro (Addgene, USA) to generate COP1 expression plasmids. Myc-Ub was provided by Dr. Cory Ronggui Hu (Shanghai Institute of Biochemistry and Cell Biology, CAS). pLKO.1-TRC cloning vector (Addgene: 10,878) was used to express shRNAs against COP1 or UTX. Briefly, the oligos targeting COP1, UTX, CUL1 and CUL4B were cloned into the pLKO.1-TRC vector to generate shRNA constructs.

Cell culture and transfection

HCT116, HT-29, SW620, SW480, LoVo, DLD1, and HEK293T cells were kindly provided by Cell Bank, Chinese Academy of Science and were cultured in DMEM containing 10% FBS (Invitrogen) at 37 °C in 5% CO2. Plasmids were transiently transfected with polyethylenimine (Sigma) or Lipofectamine 3000 (Invitrogen). Cells were transfected with siRNA using Lipofectamine 3000 (Invitrogen) according to the manufacturer’s protocol.
The siRNAs targeting indicated genes were synthesized by Biotend (Shanghai, China), and the sequences are listed in Additional file 1: Table S2.

Human primary CRC organoid culture

Organoids were cultured in Advanced DMEM/F12 medium (GIBCO, 12,634–010), containing 500 ng/mL R-spondin 1 (Sino Biological, 11,083-HNAS), 100 ng/mL Noggin (Sino Biological, 50,688-M02H), 50 ng/mL EGF (Sino Biological, 50,482-MNCH), 100 ng/mL FGF-10 (Sino Biological, 10,573-HNAE), 1X HEPES (GIBCO, 15,630,080), 1X Glutamax (GIBCO, 35,050,061), 100 μg/mL Normocin (InvivoGen, ant-nr-1), 1X Gentamicin/amphotericin B (GIBCO, R01510), 1X B27 (Invitrogen, 17,504–044), 1X N2 (Invitrogen, 17,504–048), 1 mM n-Acetylcysteine (Sigma-Aldrich, A9165), 10 mM nicotinamide (Sigma-Aldrich, N0636), 0.5 μM A-83–01 (Tocris, 2939), 10 μM Y-27632 (Sigma-Aldrich, Y0503), 10 nM Gastrin, 1 μM prostaglandin E2 (Sigma-Aldrich, P6532), and 3 μM SB202190 (Sigma-Aldrich, S7067). The culture medium was refreshed every 3 days.

Organoid cytotoxicity assay

Organoids were harvested, passaged, and seeded in a 48-well cell culture plate. Each well contained about 100 ± 50 organoids in 20 μL Matrigel with 300 μL culture medium. Once the organoids grew to 100 μm in diameter, the organoids culture medium was replaced with 300 μL drug-containing culture medium with GSK126. The drug-containing culture medium was refreshed every 3 days. Organoids were photographed every 3 days during the 9-day drug treatment period. The size of live organoids was measured using ImageJ 1.53a (National Institutes of Health, USA).

TdT-UTP nick end labeling (TUNEL) assay

TUNEL assay was performed with a colorimetric TUNEL apoptosis assay kit according to the manufacturer’s instructions (C1091, Beyotime).

5-ethynyl-2’-deoxyuridine (EdU) assay

EdU assay was conducted using BeyoClick™ EdU Cell Proliferation Kit with DAB (C0085S, Beyotime).

Xenograft tumorigenesis assay

HCT116 cells expressing ectopic COP1, ectopic COP1 plus UTX mutant (V607A/P608A/V1205A/P1206A), and empty vector were injected into the flank of each nude mouse (1 × 107 cells each mouse) to evaluate the tumorigenesis. Tumor volumes were measured and recorded every 3 days.

Antibodies and chemical inhibitors

Primary antibodies used in this study are as follows: UTX (33510S, Cell Signaling Technology; GTX121246, GeneTex), COP1 (A300-894A, Bethyl), CUL1 (SC-17775, Santa Cruz Biotechnology), CUL2 (A5308, Abclonal), CUL3 (2759, Cell Signaling Technology), CUL4A (AB60215a, Abgent), CUL4B (A6198, Abclonal), CUL5 (SC-373822, Santa Cruz Biotechnology), EMP1 (ab230445 and ab202975, Abcam), AUTS2 (25,001–1-AP, Proteintech), Ki67 (ab15580, Abcam), EZH2 (ab191080, Abcam), VEGFC (A2556, Abclonal), Caspase-9 (A0281, Abclonal), Bcl2 (15,071, Cell Signaling Technology), DDB2 (ab181136, Abcam), DCAF7 (ab138490, Abcam), DCAF13 (ab195121, Abcam), H3K27me3 (ab192985, Abcam; A2363, Abclonal), H3 (ab1791, Abcam), P27 (610,241, BD), HA-Tag (51,064–2-AP, Proteintech; SC-7392, Santa Cruz Biotechnology), Flag-Tag (F7425-0.2MG, Sigma; F3165-1MG, Sigma), GFP (66,002–1-Ig, Proteintech), Myc-Tag (16,286–1-AP, Proteintech), α-tubulin (SC-23948, Santa Cruz Biotechnology), and Vinculin (V4505, Sigma). The chemical inhibitors MG132 (S2619), MG101 (S7386), MLN4924 (S7109), Baf-A1 (S1413), and GSK126 (S7061) were purchased from Selleck.

Statistical analysis

Statistical evaluation was conducted using SPSS 25.0 (SPSS Inc., Chicago, IL, USA) and GraphPad Prism v.8 (La Jolla, CA, USA). The experimental data were analyzed using a two-tailed Student’s t-test, one-way analysis of variance (ANOVA) with Dunnett’s multiple comparisons test, Pearson’s correlation coefficients test, and log-rank test. P < 0.05 was considered significant.

Results

Low UTX expression predicts poor survival in human CRC

To investigate the putative role of UTX in CRC, a TMA containing 260 CRC samples was generated to examine UTX expression. Immunochemistry (IHC) staining results indicated significantly lower UTX expression in stages II and III CRC patients than in stage I (Fig. 1A and B). Specifically, low UTX expression was commonly observed in male patients (P = 0.003) and patients with advanced T stage disease (P = 0.045) (Table 1). Importantly, low UTX expression was significantly associated with poor overall survival (OS) (P = 0.048, Fig. 1C) and disease-free survival (DFS) (P = 0.019, Fig. 1D) in the current CRC cohort. Multivariable Cox regression analysis further revealed that UTX expression is an independent predictor for risk stratification of DFS in CRC patients (P = 0.037, Tables 2 and 3). Therefore, low UTX expression is associated with advanced CRC stages and unfavorable clinical outcomes, which is consistent with the tumor suppressor role of UTX reported in other cancer types [16, 17].
Table 1
Clinicopathological characteristics of CRC patients according to UTX expression
 
UTX expression
 
 
High group (n = 104), %
Low group (n = 156), %
P valuve
Characteristics
   
Age
  
0.325
  ≤ 60
67 (42.4)
91 (57.6)
 
  > 60
37 (36.3)
65 (63.7)
 
Gender
  
0.003
 Male
52 (32.7)
107 (67.3)
 
 Female
52 (51.5)
49 (48.5)
 
Tumor Location
  
0.085
 Colon
42 (34.4)
80 (65.6)
 
 Rectum
62 (44.9)
76 (55.1)
 
Preoperative Serum CEA
  
0.916
 Elevated
36 (40.0)
54 (60.0)
 
 Normal
65 (40.4)
96 (59.6)
 
 Unknown
3 (33.3)
6 (66.7)
 
pT stage
  
0.045
 2
23 (56.1)
18 (43.9)
 
 3
23 (42.6)
31 (57.4)
 
 4
58 (35.2)
107 (64.8)
 
pN stage
  
0.743
 0
48 (42.1)
66 (57.9)
 
 1
27 (36.5)
47 (63.5)
 
 2
29 (40.3)
43 (59.7)
 
TNM stage
  
0.283
 I
12 (60.0)
8 (40.0)
 
 II
28 (36.4)
49 (63.6)
 
 III
48 (39.0)
75 (61.0)
 
 IV
16 (40.0)
24 (60.0)
 
Historical type
  
1.000
 Adenocarcinoma
98 (40.0)
147 (60.0)
 
 Mucinous adenocarcinoma
6 (40.0)
9 (60.0)
 
Grade
  
0.499
 Well-Moderately differentiated
77 (40.7)
112 (59.3)
 
 Moderately-Poor differentiated
19 (34.5)
36 (65.5)
 
 Unknown
8 (50.0)
8 (50.0)
 
Vessel Invasion
  
0.408
 Yes
39 (45.3)
47 (54.7)
 
 No
64 (37.6)
106 (62.4)
 
 Unknown
1 (25.0)
3 (75.0)
 
Nerve Invasion
  
0.404
 Yes
14 (34.1)
27 (65.9)
 
 No
90 (41.1)
129 (58.9)
 
Bold emphasis indicates a statistically significant p-value
Table 2
Univariate and multivariate analysis of prognostic factors for disease-free survival in CRC patients
Variable
Univariate analysis
Multivariate analysis
 
HR (95% CI)
P value
HR (95% CI)
P value
UTX expression
 Low expression
Reference
 
Reference
 
 High expression
0.595 (0.383–0.925)
0.021
0.619 (0.394–0.972)
0.037
Age
  ≤ 60
Reference
 
NA
 
  > 60
1.140 (0.763–1.703)
 
0.522
 
Sex
 Male
Reference
 
NA
 
 Female
0.762 (0.503–1.155)
 
0.200
 
Tumor Location
 Colon
Reference
 
NA
 
 Return
0.809 (0.544–1.202)
 
0.293
 
Preoperative Serum CEA
 Normal
Reference
 
Reference
 
 Elevated
2.606 (1.731–3.922)
 < 0.001
1.985 (1.302–3.024)
0.001
pT stage
 T2
Reference
 
Reference
 
 T3
2.621 (0.845–8.127)
0.095
2.542 (0.805–8.029)
0.112
 T4
6.546 (2.398–17.872)
 < 0.001
4.074 (1.466–11.321)
0.007
pN stage
 0
Reference
 
Reference
 
 1
3.157 (1.836–5.427)
 < 0.001
0.777 (0.378–1.599)
0.494
 2
4.434 (2.621–7.501)
 < 0.001
0.932 (0.463–1.875)
0.843
TNM stage
 I + II
Reference
 
Reference
 
 III + IV
6.041 (3.298–11.067)
 < 0.001
6.111 (2.469–15.122)
 < 0.001
Histologic type
 Adenocarcinoma
Reference
 
NA
 
 Mucinous adenocarcinoma
0.870 (0.354–2.141)
0.762
  
Grade
 Well-Moderately differentiated
Reference
 
NA
 
 Moderately-Poor differentiated
1.268 (0.512–3.141)
0.608
  
Vessel invasion
 No
Reference
 
NA
 
 Yes
1.240 (0.171–8.976)
0.831
  
Nerve invasion
 No
Reference
 
Reference
 
 Yes
1.767 (1.107–2.821)
0.017
1.123 (0.689–1.830)
0.642
Bold emphasis indicates a statistically significant p-value
Table 3
Univariate and multivariate analysis of prognostic factors for overall survival in CRC patients
Variable
Univariate analysis
Multivariate analysis
 
HR (95% CI)
P value
HR (95% CI)
P value
UTX expression
 Low expression
Reference
 
Reference
 
 High expression
0.615 (0.377–1.001)
0.051
0.683 (0.414–1.125)
0.134
Age
  ≤ 60
Reference
 
NA
 
  > 60
1.233 (0.793–1.918)
0.353
  
Sex
 Male
Reference
 
NA
 
 Female
1.233 (0.793–1.918)
0.308
  
Tumor location
 Colon
Reference
 
NA
 
 Return
0.699 (0.450–1.086)
0.110
  
Preoperative serum CEA
 Normal
Reference
 
Reference
 
 Elevated
2.364 (1.501–3.722)
 < 0.001
1.689 (1.060–2.694)
0.028
pT stage
 T2
Reference
   
 T3
4.245 (0.930–19.377)
0.062
3.915 (0.840–18.251)
0.082
 T4
10.531 (2.579–42.994)
0.001
6.871 (1.649–28.622)
0.008
pN stage
 0
Reference
 
Reference
 
 1
2.858 (1.580–5.172)
0.001
0.586 (0.280–1.227)
0.156
 2
4.143 (2.334–7.353)
 < 0.001
0.708 (0.346–1.451)
0.346
TNM stage
 I + II
Reference
 
Reference
 
 III + IV
6.493 (3.240–13.011)
 < 0.001
8.739 (3.227–23.667
 < 0.001
Histologic type
 Adenocarcinoma
Reference
 
NA
 
 Mucinous adenocarcinoma
0.857 (0.314–2.344)
 
0.764
 
Grade
 Well-Moderately differentiated
Reference
 
NA
 
 Moderately-Poor differentiated
1.749 (0.548–5.583)
 
0.345
 
Vessel invasion
 No
Reference
 
NA
 
 Yes
0.990 (0.136–7.201)
0.992
  
Nerve invasion
 No
Reference
 
Reference
 
 Yes
1.573 (0.931–2.659)
0.091
0.938 (0.543–1.620)
0.818
Bold emphasis indicates a statistically significant p-value

UTX depletion promotes CRC tumorigenesis in vivo

Next, we generated mice with specifically depleted Utx alleles in intestinal epithelial cells by crossing mice expressing Villin-Cre and Floxed Utx (Utxf/f or Utxf/y) mice (Additional file 2: Fig. S1A). When we crossed Utx−/+ and Utx−/y mice, among a total of 327 little mid-born expressing Villin-Cre, only mice with Utx+/y; Villin-Cre (n = 103), Utx−/+; Villin-Cre (n = 115) and Utx−/y; Villin-Cre (n = 109) genotypes were born. This could be due to the fact that the deletion of both copies of Utx in intestinal epithelial cells results in the embryonic lethality in female mice, while the homologous Uty gene may partially compensate for Utx loss in male mice. Western blotting confirmed the efficiency of UTX depletion in male mice’s large intestines (Additional file 2: Fig. S1B). Over a 16 months period, no abnormalities were detected in the appearance and structure of the large intestine in the Utx−/y genotype mice, where Utx was depleted in the intestinal epithelial cells (Additional file 2: Fig. S1C and D).
Subsequently, AOM was combined with DSS to induce colitis and carcinoma (Fig. 2A). IHC was applied to determine the levels of UTX and H3K27me3. Consistent with a previous finding [15], Utx deletion led to increased H3K27me3 levels in intestinal tumors (Additional file 2: Fig. S1E).
During AOM/DSS treatment, four Utx−/y mice and one Utx−/+ mouse while no deaths were observed in WT mice. As expected, the mice with different genotypes developed tumors located mainly in the distal to middle colorectum following AOM/DSS treatment. Utx−/+ and Utx−/y mice showed marginal evidence of greater body weight loss than WT mice during AOM/DSS treatment (Fig. 2B), suggesting that the loss of Utx facilitates colitis and intestinal damage. We also observed that Utx−/+ and Utx−/y mice had more macroscopic tumors and a higher average tumor load than WT mice (Fig. 2C–E). Moreover, Utx−/y mice showed a more pronounced tumor load than Utx−/+ mice. Importantly, Utx−/y mice formed larger tumors than WT mice (P = 0.049) (Fig. 2F). And the large intestines of Utx−/y mice exhibited higher proliferative rates as compared with WT mice, as evidenced by the Ki67 signals (Fig. 2G). These results demonstrated that loss of Utx promoted tumorigenesis in the intestinal epithelial cells, and the tumor suppressive role of Utx is dose-dependent.

UTX regulates CRC cell proliferation, in part, through governing EMP1 and AUTS2 expression

In order to investigate the mechanism of UTX function in CRC, UTX-knockdown HCT116 cells were generated using two independent shRNA constructs (Fig. 3A). As expected, the depletion of UTX facilitated the proliferation of HCT116 cells, as evidenced by Cell Counting Kit-8 (CCK8) assay and anchorage-independent soft agar assay (Fig. 3B and C). Since UTX is an epigenetic enzyme regulating H3K27me3, its function is expected to involve gene-specific H3K27me3 levels and subsequent changes in transcription. Thus, we performed ChIP-seq and transcriptome analysis of normal colorectal tissues from WT and Utx knockout mice (8-weeks-old, male) to elucidate how UTX deficiency drives an oncogenic program. This analysis identified 92 genes with > 1.5-fold increment of H3K27me3 modifications and > 33.3% decrease in transcripts upon Utx depletion. Among these genes, 55 genes were further selected for a siRNA-based screening in human CRC cell line HCT116 to identify conserved target genes mediating the growth inhibitory function of human UTX based on their gene function and literature evidence. Out of the 55 genes tested in the screening, the depletion of 9 genes (comprising FRMPD1, HPSE2, EMP1, SELL, IFIT2, SLC8A1, SLC44A5, LIMCH1, and AUTS2) significantly enhanced cell proliferation. Next, we employed qPCR to verify the expression changes in these 9 genes in mice intestinal tissues and HCT116 cells upon UTX depletion or knockdown, and we validated their function in suppressing HCT116 cell proliferation. Eventually, AUTS2 and EMP1 were identified as the significant UTX target genes as their expression showed dependence on UTX depletion in both mice tissue and HCT116 cells (Fig. 3D–F). The increased H3K27me3 modifications in Emp1 and Auts2 gene locus upon Utx depletion were displayed by genomic snapshots (Additional file 3: Fig. S2A). To investigate whether Utx directly regulates Emp1 and Auts2, we performed chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR). Consistently, Utx depletion caused significantly an increased H3K27me3 around the promoter of Emp1 and Auts2 (Additional file 3: Fig. S2B). Typically, transcriptome sequencing revealed 401 significantly downregulated and 211 significantly upregulated genes upon Utx depletion (Additional file 3: Fig. S2C). Surprisingly, gene ontology (GO) analysis reflected that most biological processes affected were associated with immune processes (Additional file 3: Fig. S2D). Knockdown of EMP1 or AUTS2 significantly enhanced HCT116 cell proliferation in vitro (Fig. S3A-E). To further strengthen the potential functional role of EMP1 and AUTS2 as UTX’s major downstream genes to regulate cell growth, re-expression of EMP1 and AUTS2 was effectuated in human colorectal cancer cell line HCT116 post-shUTX treatment. Interestingly, the overexpression of EMP1 or AUTS2 largely rescued the increased growth phenotype caused by UTX depletion in the anchorage-independent soft agar assay (Additional file 4: Fig. S3F and G). Additionally, we analyzed UTX, EMP1, and AUTS2 mRNA expression in CRC patients derived from the TCGA database, and found a positive correlation between UTX-EMP1 and UTX-AUTS2 expression levels (Additional file 4: Fig. S3H). It is worth noting that AUTS2 is a component of the PRC1 complex, which regulates the development of neuronal gene expression [30]. Although several studies reported its possible function in lymphoma or leukemia, the mechanistic role of AUTS2 in cancer remains largely unclear.
EMP1 (Epithelial Membrane Protein1) has been implicated in various cancer types and is regarded to promote cancer progression by modulating cell growth and metastasis [3134]. Unlike other cancer types, EMP1 has been implicated in CRC as a putative tumor suppressor, which may inhibit cell migration and promote apoptosis [35]. To explore the underlying mechanisms by which EMP1 inhibited HCT116 cell proliferation, the western blotting analysis demonstrated downregulation of EMP1 upregulated VEGFC, Bcl-2 expression and downregulated Caspase-9 expression (Additional file 4: Fig S3A), consistent with the previous studies [35, 36]. Moreover, overexpression of EMP1 partially counteracted the suppressive role of UTX knockdown on apoptosis in HCT116 cells, as indicated by TUNEL assay and western blot (Fig. 3G–I). Taken together, these results suggested that EMP1 and AUTS2 play roles in mediating colorectal tumorigenesis driven by the loss of UTX.

Utx-deficient CRC exhibits sensitivity to EZH2 inhibitor treatment

Since the tumor suppressor role of UTX is related to H3K27me3 demethylation activity, tumors with low UTX activity may be driven by EZH2-related gene inhibition and are therefore sensitive to EZH2 inhibitors. Previous studies indicated that loss of UTX sensitizes the cells or tissues to EZH2 inhibition in multiple cancer settings [16, 18]. To explore whether UTX loss would sensitize CRC to EZH2 inhibition, spontaneous CRC was induced in WT and Utx−/y mice through AOM/DSS treatment, followed by administration of EZH2 inhibitor GSK126 or vehicle control for additional two weeks (Fig. 4A). Compared to vehicle control, GSK126 treatment did not affect tumor number and tumor load in the WT mice. In contrast, treatment with GSK126 resulted in significant tumor regression in Utx−/y mice (Fig. 4B–D) This observation is consistent with the finding that GSK126 treatment decreases the levels of H3K27me3 in both WT and Utx−/y mice (Fig. 4E). To further explore the correlation between UTX expression and sensitivity to EZH2 inhibitor, three human CRC organoids were treated with different doses of GSK126 and their growth was examined. As indicated in Fig. 4F and G, organoid #Pv24 was the most sensitive organoid to GSK126 treatment with the lowest UTX expression, while organoid #Pv11 was the most resistant organoid to GSK126 treatment with the highest UTX expression, indicating an intrinsic reverse correlation between UTX expression and sensitivity to EZH2 inhibitor in human CRC. The expression of UTX target genes EMP1 or AUTS2 at protein and mRNA levels was increased post-GSK126 treatment (Additional file 4: Fig. S3I and J). These results suggested that UTX expression determines the sensitivity of CRC to EZH2 inhibitor, and low UTX expression may serve as a biomarker to identify CRC patients who can benefit from EZH2 inhibitor therapy.

CRL4-COP1 E3 ligase complex ubiquitylates UTX for degradation

Since the downregulation of UTX promoted CRC occurrence and progression, it is crucial to understand how UTX expression is regulated in CRC cells. In order to investigate the pathway governing UTX protein stability, we observed that treatment with the proteasome inhibitor MG132 or MG101 and the neddylation inhibitor MLN4924, but not lysosomal inhibitor BafA1, resulted in a marked increase in endogenous UTX abundance (Fig. 5A). This finding indicated that UTX is degraded via the ubiquitination-proteasome system, and the Cullin-RING E3 ligase (CRL) complexes are likely involved in mediating UTX ubiquitination, as MLN4924 inhibits CRL activity, which is well-documented. We then assessed how UTX interacts with and responds to various Cullin proteins. Although UTX interacted with CUL1 and CUL4B (Additional file 5: Fig. S4A), their expression levels were elevated only upon CUL4B knockdown (Additional file 5: Fig. S4B). Subsequently, we conducted a screening by transfecting siRNAs targeting all six Cullins in HCT116 cells and found that only CUL4B knockdown upregulated UTX protein levels (Additional file 5: Fig. S4C). Furthermore, we found that endogenous UTX protein levels were increased upon depletion of CUL4B in CRC cell line SW620 (Fig. 5B) Additionally, ectopic expression of CUL4B, DDB1 and DET1, the components of the CRL4 complex, reduced UTX expression in a dose-dependent manner (Additional file 5: Fig. S4D-F). These results strongly indicate that the CUL4B-based CRL4 complex is responsible for governing UTX protein stability.
Then, we tested siRNAs targeting several substrate recognition subunits of CUL4B E3 ligases (DDB2, COP1, DCAF7, and DCAF13) in HCT116 cells and found that COP1 deficiency increased UTX protein levels (Additional file 5: Fig. S4G). Knockdown of COP1 upregulated the ectopic UTX protein levels, while overexpression of COP1 decreased ectopic UTX protein levels (Fig. 5C and D). We also discovered that COP1 splice variants interacted with UTX (Fig. 5E). Furthermore, UTX interacted with each component of the complex, including COP1, DET1, Cul4B, DDB1 and RBX1 (Additional file 5: Fig. S4H). Consistently, ectopic expression of a mutation or deletion of the COP1 RING domain (C136A/C139A), which abolishes its intrinsic E3 ubiquitin ligase activity, decreased the abundance of UTX to the same extent (Additional file 5: Fig. S4I). Importantly, the depletion of endogenous COP1 diminished the interaction between exogenous CUL4B and UTX (Fig. 5F), further confirming that COP1 is the substrate recognition subunit of the CRL4 complex that modulates UTX protein stability. Moreover, an inverse correlation was observed between UTX and COP1 proteins in CRC cell lines (Fig. 5G). Importantly, depletion of COP1 in HCT116 and LoVo cells caused a distinct increase in endogenous UTX expression and reduced H3K27me3 expression (Fig. 5H). In contrast, ectopic expression of COP1 reduced the endogenous UTX expression in SW620, HCT116, and LoVo cells without affecting the transcription of UTX gene (Fig. 5I, J and Additional file 5: Fig S4J-M). Consistently, COP1 also promoted ubiquitination of UTX transfected in HEK293T cells (Fig. 5K).
Typically, COP1 interacts with “VP” motifs of its substrates, which are termed destruction “degron” sequences [25]. Thus, we mutated each of the eight conserved “VP” motifs (VP to AA) to identify the putative COP1-degron in UTX (Fig. 5L). Strikingly, only mutation in degron 3 (V607A/P608A) or degron 7 (V1205A/P1206A) impaired COP1-mediated UTX degradation (Fig. 5M and Additional file 6: Fig S5A). Taken together, these results indicate that the CRL4-COP1 complex governs UTX stability and H3K27me3 levels in CRC cells, with COP1 recognizing specific degron sequences in UTX to facilitate its degradation.

COP1 promotes CRC partially via degrading UTX

Since COP1 negatively regulates UTX expression in intestinal tissues, we speculated that COP1 might act as an oncoprotein in CRC. Analysis of TCGA, GTEx, and GEO databases revealed that COP1 mRNA expression was upregulated in colon cancer or adenomas compared to normal colon tissues (Additional file 6: Fig. S5B and C). Immunoblotting of six pairs of CRC tumors and adjacent normal tissues confirmed that COP1 expression was upregulated in CRC tumor tissues (Additional file 6: Fig. S5D). Next, we detected the COP1 expression in paired CRC and normal (N = 104) specimens in TMA using IHC staining. The results demonstrated that COP1 expression was upregulated in CRC tissues compared to normal tissues (Fig. 6A and B). Moreover, siRNA-mediated COP1 deficiency dramatically impaired the colony formation capacity of HCT116 and LoVo cells (Fig. 6C and D). Therefore, COP1 is a putative oncoprotein in CRC tumor cells. Next, we generated mice with specifically deleted Cop1 alleles in intestinal epithelial cells by crossing flox Cop1 (Cop1f/f) mice with Villin-Cre mice (Additional file 6: Fig. S5E). When we crossed Villin-Cre;Cop1−/+ and Villin-Cre;Cop1−/+ mice, Villin-Cre;Cop1−/− mice were unavailable over the 2-year period, which could be ascribed to embryonic lethality due to the deletion of both copies of Cop1 in intestinal epithelial cells. Next, we utilized WT and Cop1−/+ mice to generate a spontaneous CRC model following AOM/DSS treatment. Initially, eight male mice were used in each group, but one mouse in the WT group died during AOM/DSS treatment. Subsequently, we observed that Cop1−/+ mice displayed fewer macroscopic tumors and a low average tumor load than WT mice (Fig. 6E–G). These observations further support the notion of an oncogenic role of COP1 in spontaneous CRC, consistent with the findings of elevated COP1 levels in clinical CRC cases.
To investigate the physiological role of COP1 in regulating UTX, we detected the protein expression of UTX in large intestines of WT and Cop1−/+ mice. Reduced COP1 expression increased the protein levels of UTX in mouse intestinal tissues, as detected by both immunoblotting and IHC staining (Fig. 7A and B). Then, we stained 260 CRC tumor tissues by the IHC method to determine the protein expression of COP1 and UTX. As indicated, the COP1 signal was high in most CRC tumors and significantly correlated with lower UTX expression (r = -0.257, P < 0.001, Fig. 7C and D). These findings suggest COP1 may facilitate CRC growth via UTX degradation. To investigate this hypothesis, we performed xenograft tumorigenesis experiment using HCT116 cells that ectopically expressed HA-COP1 and Flag-UTX mutant (V607A/P608A/V1205A/P1206A) resistant to COP1-mediated degradation (Additional file 6: Fig. S5F). The ectopic expression of COP1 dramatically promoted tumorigenesis compared to parental HCT116 cells, while expression of non-degradable UTX essentially reversed the tumor-promoting effect derived from COP1 overexpression (Fig. 7E–G). Importantly, this reversal was associated with increased H3K27me3 levels upon COP1 overexpression and reduced H3K27me3 levels after overexpression of non-degradable UTX in the tumors from xenograft experiment (Fig. 7H). These results suggested that COP1-mediated UTX degradation is crucial for COP1-driven CRC tumorigenesis.

Discussion

The oncogenic function and regulation of EZH2, the major H3K27 methyltransferase, in CRC have been well-documented [3740]. Conversely, the functional and regulatory mechanism of the H3K27me3 histone demethylase UTX in CRC is yet unclear. In the present study, we found that UTX protein levels were downregulated in advanced stages of CRC as examined by immunostaining. Moreover, low UTX expression was associated with poor patient survival, indicating a putative tumor suppressor role of UTX in human CRC (Fig. 1). Next, we observed that UTX loss significantly increased tumor numbers and burden in the AOM/DSS-induced CRC model. These results suggested that UTX functions as a tumor suppressor in CRC, and reduced UTX may drive the progression of the disease. Since reduced UTX expression in intestinal tissue significantly increased the H3K27me3 levels, which are enriched in CRC with more advanced disease stages, we speculated that targeting H3K27me3 may be an effective way to treat UTX loss-related CRC. Typically, spontaneous colorectal tumors derived from UTX-deficient intestines or human CRC organoids with low UTX expression exhibited sensitivity to EZH2 inhibitors, consistent with previous reports in other cancer types [16, 18]. Therefore, the EZH2 inhibitor could be a promising therapeutic option for CRC patients with UTX deficiency.
The combined analysis of UTX-depleted mice intestine and human CRC cells identified EMP1 and AUTS2 as the target genes modulated by UTX since their expression decreased upon UTX depletion and increased upon treatment with the EZH2 inhibitor GSK126. Notably, the depletion of EMP1 and AUTS2 promoted CRC cell proliferation, a phenotype similar to UTX knockdown. Reportedly, EMP1 has a possible cross-talk with the EGFR signaling pathway [41] and negatively regulates cell growth and metastasis in CRC [35], and its downregulation is observed in multiple cancers [36, 42]. In contrast, AUTS2 is frequently disrupted in patients with neurological disorders [30] but is less studied in cancer. Here, we characterized UTX as the epigenetic regulator of EMP1 and AUTS2 expression. Furthermore, the increased protein level of VEGFC and BCL2 as well as reduced cell apoptosis caused by depletion of UTX could be rescued by ectopic expression of EMP1, suggesting that EMP1 is a significant effector gene mediating the tumor-suppressing function of UTX in CRC. However, during our screening process, we only focused on putative target genes mutually modulated by UTX in human CRC cells and mice intestines, particularly those that affect cell growth. Other putative UTX target genes (FRMPD1, HPSE2, SELL, IFIT2, SLC8A1, SLC44A5, and LIMCH1) were identified in ChIP-seq and RNA-seq analyses. These genes may also modulate CRC via various mechanisms other than regulating cell growth. Some specific human UTX target genes that mediate its tumor suppressor function in CRC might not have been discovered in the mouse model.
According to the COSMIC (the Catalogue of Somatic Mutations in Cancer) database and previous reports, genomic deletion or mutation of UTX is not a frequent event in CRC. Therefore, it is essential to understand how UTX expression is impaired in CRC. The current study indicated that the CUL4B-DDB1-COP1 E3 ligase complex governs UTX protein stability. Depletion of COP1 in human CRC cells or mice intestinal tissue caused a marked increase in UTX expression and restricted tumorigenesis. The expression of COP1 was inversely correlated with UTX expression in human CRC specimens. Furthermore, we observed a remarkably enhanced growth of tumors upon ectopic expression of COP1 in the in vivo tumorigenesis assays. Importantly, ectopic expression of a non-degradable UTX mutant reversed the tumor-promoting effect of COP1 overexpression. Mechanistically, we have characterized that UTX is a substrate for the CUL4B-DDB1-COP1 E3 complex, but not for the COP1 intrinsic RING motif. We also identified two putative “VP” degron motifs in UTX that mediate COP1 recognition. Thus, the current study revealed the oncogenic role of COP1 in CRC, at least partially, by degrading UTX and inducing epigenetic changes. Additionally, it uncovers a critical layer of UTX regulation in CRC.

Conclusions

The current study has provided insights into the tumor suppressor function of UTX in CRC. We identified its downstream effector genes, EMP1 and AUTS2, which play crucial roles in mediating its tumor-suppressing effects. Additionally, we observed the downregulation of UTX caused by the CRL4-COP1 complex in CRC (Fig. 7I). Thus, our findings would shed new light on the epigenetic reprogramming and novel therapeutic option of CRC with UTX deficiency.

Acknowledgements

We are grateful to Dr. Charlie Degui Chen, Anning Lin, and Cory Ronghui Hu for generously sharing mouse strains and reagents. We thank Dr. Cong Tan and Shujuan Ni, specialist pathologists from the Department of Pathology, Fudan University Shanghai Cancer Center, for pathological evaluation. We also thank the Chemical Biology Core Facility of the Center for Excellence in Molecular Cell Science, CAS, for technical assistance.

Declarations

All animal experiments were approved by the Institutional Animal Care and Use Committee of the Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. The CRC specimens were obtained from patients who underwent surgery at Fudan University Shanghai Cancer Center. The study protocol was approved by the Ethics Committee of Fudan University Shanghai Cancer Center.
Not applicable.

Competing interests

Junqiang Li has ownership of D1 Medical Technology (Shanghai) Co., Ltd, Shanghai. Other authors do not have any competing interest.
Open Access This 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.
Anhänge

Supplementary Information

Additional file 5: Figure S4. CRL4-COP1 E3 complex modulates UTX stability. A IB analysis of WCL and IP from HEK293T cells co-transfected with HA-UTX and different Cullin family constructs. Cells were treated with MG132 (20 µM) for 12 h before they were harvested. B IB analysis of WCL from HEK293T cells co-transfected with Flag-UTX and shCUL1 or shCUL4B. C IB analysis of WCL from HCT116 cells transfected with indicated siRNA targeting different Cullins. D IB analysis of WCL derived from HEK293T cells co-transfected with various doses of Flag-CUL4B together with HA-UTX constructs. Cells were treated with or without MG132, as indicated before they were harvested. E IB analysis of WCL derived from HEK293T cells co-transfected with various doses of Flag-DDB1 and HA-UTX constructs. Cells were treated with or without MG132, as indicated before they were harvested. F IB analysis of WCL derived from HEK293T cells co-transfected with various doses of Flag-DET1 and HA-UTX constructs. Cells were treated with or without MG132, as indicated before they were harvested. G IB analysis of WCL from HCT116 cells transfected with indicated siRNA targeting different CUL4B adaptor E3 ligases. H IB analysis of WCL and IP from HEK293T cells co-transfected with HA-UTX and CUL4B complex. Cells were treated with MG132 (20 µM) for 12 h before they were harvested. I IB analysis of WCL derived from HEK293T cells co-transfected with HA-UTX and Flag-COP1 (WT or C136A/C139A or △Ring). J and K IB (H) and qPCR analysis (I) of UTX expression in HCT116 cells infected with lentiviruses expressing EV or Flag-COP1. L and M IB (J) and qPCR analysis (K) of UTX expression in LoVo cells infected with lentiviruses expressing EV or Flag-COP1. Data information: In (K and M), data are presented as mean±SEM (two-tailed Student’s t-test). ns, non-significance, ***P<0.001
Literatur
1.
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(3):209–49.CrossRefPubMed 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(3):209–49.CrossRefPubMed
2.
Zurück zum Zitat Stoffel EM, Koeppe E, Everett J, Ulintz P, Kiel M, Osborne J, et al. Germline genetic features of young individuals with colorectal cancer. Gastroenterology. 2018;154(4):897–905.PubMed Stoffel EM, Koeppe E, Everett J, Ulintz P, Kiel M, Osborne J, et al. Germline genetic features of young individuals with colorectal cancer. Gastroenterology. 2018;154(4):897–905.PubMed
3.
Zurück zum Zitat Jung G, Hernandez-Illan E, Moreira L, Balaguer F, Goel A. Epigenetics of colorectal cancer: biomarker and therapeutic potential. Nat Rev Gastroenterol Hepatol. 2020;17(2):111–30.PubMedPubMedCentral Jung G, Hernandez-Illan E, Moreira L, Balaguer F, Goel A. Epigenetics of colorectal cancer: biomarker and therapeutic potential. Nat Rev Gastroenterol Hepatol. 2020;17(2):111–30.PubMedPubMedCentral
4.
Zurück zum Zitat Orouji E, Raman AT, Singh AK, Sorokin A, Arslan E, Ghosh AK, et al. Chromatin state dynamics confers specific therapeutic strategies in enhancer subtypes of colorectal cancer. Gut. 2022;71(5):938–49.PubMed Orouji E, Raman AT, Singh AK, Sorokin A, Arslan E, Ghosh AK, et al. Chromatin state dynamics confers specific therapeutic strategies in enhancer subtypes of colorectal cancer. Gut. 2022;71(5):938–49.PubMed
5.
Zurück zum Zitat Clarke TL, Tang R, Chakraborty D, Van Rechem C, Ji F, Mishra S, et al. Histone lysine methylation dynamics control EGFR DNA copy-number amplification. Cancer Discov. 2020;10(2):306–25.PubMed Clarke TL, Tang R, Chakraborty D, Van Rechem C, Ji F, Mishra S, et al. Histone lysine methylation dynamics control EGFR DNA copy-number amplification. Cancer Discov. 2020;10(2):306–25.PubMed
6.
Zurück zum Zitat Chung C, Sweha SR, Pratt D, Tamrazi B, Panwalkar P, Banda A, et al. Integrated metabolic and epigenomic reprograming by H3K27M mutations in diffuse intrinsic pontine gliomas. Cancer Cell. 2020;38(3):334–49.PubMedPubMedCentral Chung C, Sweha SR, Pratt D, Tamrazi B, Panwalkar P, Banda A, et al. Integrated metabolic and epigenomic reprograming by H3K27M mutations in diffuse intrinsic pontine gliomas. Cancer Cell. 2020;38(3):334–49.PubMedPubMedCentral
7.
Zurück zum Zitat Qian Y, Li Y, Zheng C, Lu T, Sun R, Mao Y, et al. High methylation levels of histone H3 lysine 9 associated with activation of hypoxia-inducible factor 1alpha (HIF-1alpha) predict patients’ worse prognosis in human hepatocellular carcinomas. Cancer Genet. 2020;245:17–26.PubMed Qian Y, Li Y, Zheng C, Lu T, Sun R, Mao Y, et al. High methylation levels of histone H3 lysine 9 associated with activation of hypoxia-inducible factor 1alpha (HIF-1alpha) predict patients’ worse prognosis in human hepatocellular carcinomas. Cancer Genet. 2020;245:17–26.PubMed
8.
Zurück zum Zitat Yang GJ, Zhu MH, Lu XJ, Liu YJ, Lu JF, Leung CH, et al. The emerging role of KDM5A in human cancer. J Hematol Oncol. 2021;14(1):30.PubMedPubMedCentral Yang GJ, Zhu MH, Lu XJ, Liu YJ, Lu JF, Leung CH, et al. The emerging role of KDM5A in human cancer. J Hematol Oncol. 2021;14(1):30.PubMedPubMedCentral
10.
Zurück zum Zitat Shpargel KB, Sengoku T, Yokoyama S, Magnuson T. UTX and UTY demonstrate histone demethylase-independent function in mouse embryonic development. PLoS Genet. 2012;8(9): e1002964.PubMedPubMedCentral Shpargel KB, Sengoku T, Yokoyama S, Magnuson T. UTX and UTY demonstrate histone demethylase-independent function in mouse embryonic development. PLoS Genet. 2012;8(9): e1002964.PubMedPubMedCentral
11.
Zurück zum Zitat Welstead GG, Creyghton MP, Bilodeau S, Cheng AW, Markoulaki S, Young RA, et al. X-linked H3K27me3 demethylase Utx is required for embryonic development in a sex-specific manner. Proc Natl Acad Sci U S A. 2012;109(32):13004–9.PubMedPubMedCentral Welstead GG, Creyghton MP, Bilodeau S, Cheng AW, Markoulaki S, Young RA, et al. X-linked H3K27me3 demethylase Utx is required for embryonic development in a sex-specific manner. Proc Natl Acad Sci U S A. 2012;109(32):13004–9.PubMedPubMedCentral
12.
Zurück zum Zitat van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, Greenman C, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41(5):521–3.PubMedPubMedCentral van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, Greenman C, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41(5):521–3.PubMedPubMedCentral
13.
Zurück zum Zitat Mar BG, Bullinger L, Basu E, Schlis K, Silverman LB, Dohner K, et al. Sequencing histone-modifying enzymes identifies UTX mutations in acute lymphoblastic leukemia. Leukemia. 2012;26(8):1881–3.PubMedPubMedCentral Mar BG, Bullinger L, Basu E, Schlis K, Silverman LB, Dohner K, et al. Sequencing histone-modifying enzymes identifies UTX mutations in acute lymphoblastic leukemia. Leukemia. 2012;26(8):1881–3.PubMedPubMedCentral
14.
Zurück zum Zitat Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487(7406):239–43.PubMedPubMedCentral Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487(7406):239–43.PubMedPubMedCentral
15.
Zurück zum Zitat Zheng L, Xu L, Xu Q, Yu L, Zhao D, Chen P, et al. Utx loss causes myeloid transformation. Leukemia. 2018;32(6):1458–65.PubMed Zheng L, Xu L, Xu Q, Yu L, Zhao D, Chen P, et al. Utx loss causes myeloid transformation. Leukemia. 2018;32(6):1458–65.PubMed
16.
Zurück zum Zitat Wu Q, Tian Y, Zhang J, Tong X, Huang H, Li S, et al. In vivo CRISPR screening unveils histone demethylase UTX as an important epigenetic regulator in lung tumorigenesis. Proc Natl Acad Sci U S A. 2018;115(17):E3978–86.PubMedPubMedCentral Wu Q, Tian Y, Zhang J, Tong X, Huang H, Li S, et al. In vivo CRISPR screening unveils histone demethylase UTX as an important epigenetic regulator in lung tumorigenesis. Proc Natl Acad Sci U S A. 2018;115(17):E3978–86.PubMedPubMedCentral
17.
Zurück zum Zitat Li X, Zhang Y, Zheng L, Liu M, Chen CD, Jiang H. UTX is an escape from X-inactivation tumor-suppressor in B cell lymphoma. Nat Commun. 2018;9(1):2720.PubMedPubMedCentral Li X, Zhang Y, Zheng L, Liu M, Chen CD, Jiang H. UTX is an escape from X-inactivation tumor-suppressor in B cell lymphoma. Nat Commun. 2018;9(1):2720.PubMedPubMedCentral
18.
Zurück zum Zitat Ezponda T, Dupere-Richer D, Will CM, Small EC, Varghese N, Patel T, et al. UTX/KDM6A loss enhances the malignant phenotype of multiple myeloma and sensitizes cells to EZH2 inhibition. Cell Rep. 2017;21(3):628–40.PubMedPubMedCentral Ezponda T, Dupere-Richer D, Will CM, Small EC, Varghese N, Patel T, et al. UTX/KDM6A loss enhances the malignant phenotype of multiple myeloma and sensitizes cells to EZH2 inhibition. Cell Rep. 2017;21(3):628–40.PubMedPubMedCentral
19.
Zurück zum Zitat Tang X, Cai W, Cheng J, Lu P, Ma S, Chen C, et al. The histone H3 lysine-27 demethylase UTX plays a critical role in colorectal cancer cell proliferation. Cancer Cell Int. 2019;19:144.PubMedPubMedCentral Tang X, Cai W, Cheng J, Lu P, Ma S, Chen C, et al. The histone H3 lysine-27 demethylase UTX plays a critical role in colorectal cancer cell proliferation. Cancer Cell Int. 2019;19:144.PubMedPubMedCentral
20.
Zurück zum Zitat Pohl C, Dikic I. Cellular quality control by the ubiquitin-proteasome system and autophagy. Science. 2019;366(6467):818–22.PubMed Pohl C, Dikic I. Cellular quality control by the ubiquitin-proteasome system and autophagy. Science. 2019;366(6467):818–22.PubMed
21.
Zurück zum Zitat Cavadini S, Fischer ES, Bunker RD, Potenza A, Lingaraju GM, Goldie KN, et al. Cullin-RING ubiquitin E3 ligase regulation by the COP9 signalosome. Nature. 2016;531(7596):598–603.PubMed Cavadini S, Fischer ES, Bunker RD, Potenza A, Lingaraju GM, Goldie KN, et al. Cullin-RING ubiquitin E3 ligase regulation by the COP9 signalosome. Nature. 2016;531(7596):598–603.PubMed
22.
23.
Zurück zum Zitat Deng XW, Caspar T, Quail PH. cop1: a regulatory locus involved in light-controlled development and gene expression in Arabidopsis. Genes Dev. 1991;5(7):1172–82.PubMed Deng XW, Caspar T, Quail PH. cop1: a regulatory locus involved in light-controlled development and gene expression in Arabidopsis. Genes Dev. 1991;5(7):1172–82.PubMed
24.
Zurück zum Zitat Hoecker U. The activities of the E3 ubiquitin ligase COP1/SPA, a key repressor in light signaling. Curr Opin Plant Biol. 2017;37:63–9.PubMed Hoecker U. The activities of the E3 ubiquitin ligase COP1/SPA, a key repressor in light signaling. Curr Opin Plant Biol. 2017;37:63–9.PubMed
25.
Zurück zum Zitat Marine JC. Spotlight on the role of COP1 in tumorigenesis. Nat Rev Cancer. 2012;12(7):455–64.PubMed Marine JC. Spotlight on the role of COP1 in tumorigenesis. Nat Rev Cancer. 2012;12(7):455–64.PubMed
26.
Zurück zum Zitat Wertz IE, O’Rourke KM, Zhang Z, Dornan D, Arnott D, Deshaies RJ, et al. Human De-etiolated-1 regulates c-Jun by assembling a CUL4A ubiquitin ligase. Science. 2004;303(5662):1371–4.PubMed Wertz IE, O’Rourke KM, Zhang Z, Dornan D, Arnott D, Deshaies RJ, et al. Human De-etiolated-1 regulates c-Jun by assembling a CUL4A ubiquitin ligase. Science. 2004;303(5662):1371–4.PubMed
27.
Zurück zum Zitat Vitari AC, Leong KG, Newton K, Yee C, O’Rourke K, Liu J, et al. COP1 is a tumour suppressor that causes degradation of ETS transcription factors. Nature. 2011;474(7351):403–6.PubMed Vitari AC, Leong KG, Newton K, Yee C, O’Rourke K, Liu J, et al. COP1 is a tumour suppressor that causes degradation of ETS transcription factors. Nature. 2011;474(7351):403–6.PubMed
28.
Zurück zum Zitat Dornan D, Wertz I, Shimizu H, Arnott D, Frantz GD, Dowd P, et al. The ubiquitin ligase COP1 is a critical negative regulator of p53. Nature. 2004;429(6987):86–92.PubMed Dornan D, Wertz I, Shimizu H, Arnott D, Frantz GD, Dowd P, et al. The ubiquitin ligase COP1 is a critical negative regulator of p53. Nature. 2004;429(6987):86–92.PubMed
29.
Zurück zum Zitat Sauter G, Simon R, Hillan K. Tissue microarrays in drug discovery. Nat Rev Drug Discov. 2003;2(12):962–72.PubMed Sauter G, Simon R, Hillan K. Tissue microarrays in drug discovery. Nat Rev Drug Discov. 2003;2(12):962–72.PubMed
30.
Zurück zum Zitat Gao Z, Lee P, Stafford JM, von Schimmelmann M, Schaefer A, Reinberg D. An AUTS2-Polycomb complex activates gene expression in the CNS. Nature. 2014;516(7531):349–54.PubMedPubMedCentral Gao Z, Lee P, Stafford JM, von Schimmelmann M, Schaefer A, Reinberg D. An AUTS2-Polycomb complex activates gene expression in the CNS. Nature. 2014;516(7531):349–54.PubMedPubMedCentral
31.
Zurück zum Zitat Wang J, Li X, Wu H, Wang H, Yao L, Deng Z, et al. EMP1 regulates cell proliferation, migration, and stemness in gliomas through PI3K-AKT signaling and CD44. J Cell Biochem. 2019;120(10):17142–50.PubMed Wang J, Li X, Wu H, Wang H, Yao L, Deng Z, et al. EMP1 regulates cell proliferation, migration, and stemness in gliomas through PI3K-AKT signaling and CD44. J Cell Biochem. 2019;120(10):17142–50.PubMed
32.
Zurück zum Zitat Liu Y, Ding Y, Nie Y, Yang M. EMP1 promotes the proliferation and invasion of ovarian cancer cells through activating the MAPK pathway. Onco Targets Ther. 2020;13:2047–55.PubMedPubMedCentral Liu Y, Ding Y, Nie Y, Yang M. EMP1 promotes the proliferation and invasion of ovarian cancer cells through activating the MAPK pathway. Onco Targets Ther. 2020;13:2047–55.PubMedPubMedCentral
33.
Zurück zum Zitat Aries IM, Jerchel IS, van den Dungen RE, van den Berk LC, Boer JM, Horstmann MA, et al. EMP1, a novel poor prognostic factor in pediatric leukemia regulates prednisolone resistance, cell proliferation, migration and adhesion. Leukemia. 2014;28(9):1828–37.PubMed Aries IM, Jerchel IS, van den Dungen RE, van den Berk LC, Boer JM, Horstmann MA, et al. EMP1, a novel poor prognostic factor in pediatric leukemia regulates prednisolone resistance, cell proliferation, migration and adhesion. Leukemia. 2014;28(9):1828–37.PubMed
34.
Zurück zum Zitat Ahmat Amin MKB, Shimizu A, Zankov DP, Sato A, Kurita S, Ito M, et al. Epithelial membrane protein 1 promotes tumor metastasis by enhancing cell migration via copine-III and Rac1. Oncogene. 2018;37(40):5416–34.PubMedPubMedCentral Ahmat Amin MKB, Shimizu A, Zankov DP, Sato A, Kurita S, Ito M, et al. Epithelial membrane protein 1 promotes tumor metastasis by enhancing cell migration via copine-III and Rac1. Oncogene. 2018;37(40):5416–34.PubMedPubMedCentral
35.
Zurück zum Zitat Sun GG, Wang YD, Cui DW, Cheng YJ, Hu WN. Epithelial membrane protein 1 negatively regulates cell growth and metastasis in colorectal carcinoma. World J Gastroenterol. 2014;20(14):4001–10.PubMedPubMedCentral Sun GG, Wang YD, Cui DW, Cheng YJ, Hu WN. Epithelial membrane protein 1 negatively regulates cell growth and metastasis in colorectal carcinoma. World J Gastroenterol. 2014;20(14):4001–10.PubMedPubMedCentral
36.
Zurück zum Zitat Sun GG, Wang YD, Cui DW, Cheng YJ, Hu WN. EMP1 regulates caspase-9 and VEGFC expression and suppresses prostate cancer cell proliferation and invasion. Tumour Biol. 2014;35(4):3455–62.PubMed Sun GG, Wang YD, Cui DW, Cheng YJ, Hu WN. EMP1 regulates caspase-9 and VEGFC expression and suppresses prostate cancer cell proliferation and invasion. Tumour Biol. 2014;35(4):3455–62.PubMed
37.
Zurück zum Zitat Zhou J, Huang S, Wang Z, Huang J, Xu L, Tang X, et al. Targeting EZH2 histone methyltransferase activity alleviates experimental intestinal inflammation. Nat Commun. 2019;10(1):2427.PubMedPubMedCentral Zhou J, Huang S, Wang Z, Huang J, Xu L, Tang X, et al. Targeting EZH2 histone methyltransferase activity alleviates experimental intestinal inflammation. Nat Commun. 2019;10(1):2427.PubMedPubMedCentral
38.
Zurück zum Zitat Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419(6907):624–9.PubMed Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419(6907):624–9.PubMed
39.
Zurück zum Zitat Kim KH, Kim W, Howard TP, Vazquez F, Tsherniak A, Wu JN, et al. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med. 2015;21(12):1491–6.PubMedPubMedCentral Kim KH, Kim W, Howard TP, Vazquez F, Tsherniak A, Wu JN, et al. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med. 2015;21(12):1491–6.PubMedPubMedCentral
40.
Zurück zum Zitat Wang B, Liu Y, Liao Z, Wu H, Zhang B, Zhang L. EZH2 in hepatocellular carcinoma: progression, immunity, and potential targeting therapies. Exp Hematol Oncol. 2023;12(1):52.PubMedPubMedCentral Wang B, Liu Y, Liao Z, Wu H, Zhang B, Zhang L. EZH2 in hepatocellular carcinoma: progression, immunity, and potential targeting therapies. Exp Hematol Oncol. 2023;12(1):52.PubMedPubMedCentral
41.
Zurück zum Zitat Jain A, Tindell CA, Laux I, Hunter JB, Curran J, Galkin A, et al. Epithelial membrane protein-1 is a biomarker of gefitinib resistance. Proc Natl Acad Sci U S A. 2005;102(33):11858–63.PubMedPubMedCentral Jain A, Tindell CA, Laux I, Hunter JB, Curran J, Galkin A, et al. Epithelial membrane protein-1 is a biomarker of gefitinib resistance. Proc Natl Acad Sci U S A. 2005;102(33):11858–63.PubMedPubMedCentral
42.
Zurück zum Zitat Zhang J, Cao W, Xu Q, Chen WT. The expression of EMP1 is downregulated in oral squamous cell carcinoma and possibly associated with tumour metastasis. J Clin Pathol. 2011;64(1):25–9.PubMed Zhang J, Cao W, Xu Q, Chen WT. The expression of EMP1 is downregulated in oral squamous cell carcinoma and possibly associated with tumour metastasis. J Clin Pathol. 2011;64(1):25–9.PubMed
Metadaten
Titel
CUL4B-DDB1-COP1-mediated UTX downregulation promotes colorectal cancer progression
verfasst von
Dakui Luo
Min Chen
Qingguo Li
Kangjunjie Wang
Kaihua Wang
Junqiang Li
Guoxiang Fu
Zezhi Shan
Qi Liu
Yufei Yang
Lei Liang
Yanlei Ma
Yi Qin
Jun Qin
Daming Gao
Xinxiang Li
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00440-z

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

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.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

Update Onkologie

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