Colorectal cancer (CRC) is a common malignant tumor in digestive tract with highly invasive and metastatic capacity. Drug sensitivity remains a significant obstacle to successful chemotherapy in CRC patients. The present study aimed to explore genes related to cetuximab (CTX) sensitivity in CRC by clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9. Celigo image cytometer was used to detect suitable cells and optimal dosage of CTX. Inhibition rate of CTX on Caco-2 cells was evaluated by cell counting kit-8 (CCK-8) method before and after transfection. 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyl tetrazolium bromide (MTT) was performed to explore suitable concentration of puromycin and multiplicity of infection (MOI). CRISPR-Cas9, sequencing data quality analysis and cell viability test were used for the selection of genes related to CTX sensitivity in CRC cells. Finally, the selected genes associated with CTX sensitivity in CRC cells were further validated by colony formation and CCK-8 assays. In the present study, Caco-2 cells had a better prolificacy, and CTX 100 μg/ml exhibited a good inhibition trend on the 7th and 14th days of infection. MTT assay indicated that the minimum lethal concentration of puromycin was 2.5 μg/ml. Forty-six candidate genes were preliminarily screened via sequencing data quality analysis. Subsequently, we found that knockout of any of the four genes (MMP15, MRPL48, CALN1 and HADHB) could enhance CTX sensitivity in Caco-2 cells, which was further confirmed by colony formation assay. In summary, MMP15, MRPL48, CALN1 and HADHB genes are related to the mediation of CTX sensitivity in CRC.

Colorectal cancer (CRC) is a highly invasive and metastatic malignant tumor of the digestive tract, with high mortality [1]. Most CRC cells are currently assumed to be stem cells or stem cell-like cells. These stem cells are the outcomes of progressive accumulation of genetic and epigenetic changes that activate oncogenes and inactivate tumor-suppressor genes [2]. Due to the low early diagnosis rate, most patients are diagnosed as middle or advanced CRC, resulting in difficult treatment and poor prognosis [3]. At present, chemotherapy and radiotherapy are typical therapies for CRC. Systemic treatment of metastatic CRC usually involves chemotherapy backbone (fluoropyrimidines, oxaliplatin and irinotecan chemotherapies form) combined with biotherapy [4]. However, despite therapeutic advancements, the prognosis for metastatic CRC patients remains poor, with a median overall survival of 18–21 months [5]. Drug resistance is the main reason for the failure of chemotherapy in cancers.

Apart from chemotherapy and radiotherapy, advances have been achieved by the addition of targeted agents. Cetuximab (CTX) (Erbitux) is a monoclonal antibody that targets epidermal growth factor receptor to impede the phosphorylation of receptor-associated kinases and related signaling cascade [6], and plays an anti-tumor role at both cellular and genetic levels [7]. Cetuximab was also demonstrated to enhance overall survival when combined with radiotherapy alone [8]. Segelov et al. have clarified that the median survival of advanced CRC patients can be extended from 6–7 to 24–30 months after treatment with combined targeted drugs such as CTX and chemotherapy [9]. However, after a period of CTX treatment, numerous patients become insensitive to CTX with disease progression. CTX can kill cancer cells that are sensitive to drugs, but the remaining resistant cells can cause tumor recurrence and even metastasis. Currently, the vital challenge for CRC treatment is to improve sensitivity to CTX. Therefore, it is of great significance to find new CTX-sensitized targets in CRC.

Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 is a special structure found in the genomes of bacteria and archaea that protect them from phages and plasmids [10]. Scientists have transformed CRISPR-Cas9 into a simple, efficient and accurate genome editing tool, providing an effective method for finding functional genes [11]. For CRC, the use of CRISPR-Cas9 to explore genes related to CTX sensitivity has not been reported. Therefore, in the present study, we screened novel genes involved in CTX sensitivity in CRC cells based on genome-scale CRISPR-Cas9 knockout (GeCKO) screening and coupled with cell viability test.

Cell culture

HT-29 and Caco-2 cells were obtained from the Cell Biology Institute of Chinese Academy of Sciences. The cells were cultured in DMEM (Gibco, Invitrogen, U.S.A.), supplemented with 10% fetal bovine serum (HyClone Laboratories, Logan, UT), 100 U/ml penicillin and 100 µg/ml streptomycin (Invitrogen, CA, U.S.A.) at 37°C in a humidified atmosphere of 5% CO2.

CRISPR-Cas9 knockout screening

To identify genes associated with enhanced sensitivity of CRC to CTX, GeCKO screening with a pooled GeCKO library was performed. We applied a GeCKO library of 123411 single-guide RNAs (sgRNAs) targeting 20914 human genes, including 19050 annotated protein-coding genes and 1864 microRNA expression genes, with an average coverage of six sgRNAs per gene. A single lentiviral vector system (lentiCRISPRv2) of GeCKO library was used to transfect HT-29 and Caco-2 cells at a low multiplicity of infection (MOI) to ensure that most cells gained only one viral construct. After 72 h of transduction, HT-29 and Caco-2 cells were screened with puromycin, and the cells that survived after 7 days were successfully transfected cells. A total of 3 × 107 cells were directly harvested as day 0 group after puromycin selection. Then the remaining cells were treated with dimethyl sulfoxide (DMSO) vehicle and 100 μg/ml CTX for 7 and 14 days, respectively.

Cell growth curve assay

HT-29 and Caco-2 cells were plated in triplicate in 96-well plates at a density of 2000 cells per well. After culturing at 37°C in a 5% CO2 incubator for another day, Celigo image cytometer (Cyntellect Inc., CA, U.S.A.) was used to capture cell images once a day for 4 days. Cell numbers in each well were quantified by Celigo image cytometer, and cell growth curves were generated for each cell.

MTT assay

To explore the most suitable concentration of puromycin, 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyl tetrazolium bromide (MTT) (Sigma Chemical Company, MO, U.S.A.) was used to assess the proliferation of Caco-2 cells. Briefly, Caco-2 cells were seeded in 96-well plates in triplicate at the density of 2000 cells per well, followed by addition of puromycin at various concentrations (0, 1.25, 2.5, 5, 10 μg/ml). Then the culture medium was discarded, and 20 µl of MTT (5 mg/ml) were added into each well. After 7 days of incubation, the MTT crystals were dissolved in DMSO and the absorbance at 490 nm was measured.

Cell proliferation analysis via Celigo image cytometer

To find out the best dosage of CTX, Celigo image cytometer was used to quantify the cell number. The Caco-2 cells were placed in 96-well plates in triplicate and then adding CTX at a range of concentrations (0, 50, 100, 200, 400, 600, 800, 1200 μg/ml). After culturing at 37°C in a 5% CO2 incubator for another day, Celigo image cytometer was used to capture cell images for 14 days. Cell numbers in each well were quantified by Celigo image cytometer, and cell growth curves were generated for each cell.

Determination of optimal MOI

Caco-2 cells with and without puromycin were divided into group A and group B, respectively. Cells were infected with different MOI values (0.25, 0.5, 1, 2, 10) and seeded in 96-well plates at the density of 6000 cells per well. Puromycin (2.5 μg/ml) was then added to each well in group A only. Forty-eight hours later, the culture medium was discarded, and 20 µl of MTT (5 mg/ml) were added to each well. Following a 4-h incubation, the crystals were dissolved by DMSO and the absorbance at 490 nm was read. The functional MOI was defined as the MTT value of group A/the MTT value of group B [12].

Sequencing data quality analysis

NEBNext® Ultra™ DNA Library Prep Kit for Illumina® kit (QIAGEN Company, Hilden, Germany) was used for the construction of library. Briefly, more than 50 ng PCR products purified by gel cut or magnetic beads were adopted to construct the library directly. End Prep Enzyme Mix was used for end repair, including 5′-end phosphorylation and 3′-end of ‘A’ plus, with addition of sequencing adapter at both ends, followed by purification via AxyPrep Mag PCR Clean-up. Finally, P5 and P7 primers were used for amplification, in which the index containing 6-bp base at the end of P7 was used for subsequent sample discrimination. After purification, the quality and concentration of library were measured by Agilent 2100 bioanalyzer (Agilent Technologies, CA, U.S.A.) and Qubit 2.0 fluorophotometer (Invitrogen, CA, U.S.A.), respectively. Subsequently, 2× 150 bp paired-end sequencing (PE) was performed according to Illumina HiSeq instruction (Illumina, CA, U.S.A.), and the sequence information was read by HiSeq Control Software (HCS) + OLB + GAPipeline-1.6.

Cell viability test

The infected cells in logarithmic phase were seeded in 96-well plates and were treated with 100 μl of CTX 100 μg/ml or DMSO, respectively. Meanwhile, normal control (NC) group was set up and treated with CTX 100 μg/ml or DMSO, respectively. Five days later, 100 μl of CellTiter-Glo (thawed 24 h at 4°C in advance) was added to each well and shook on an oscillated instrument for 10 min to induce cell lysis. Subsequently, an enzyme mark instrument was adopted to measure the absorbance of each well in Luminescent mode. The PC interference background, multiples of inhibited proliferation and sensitization multiples of each group of cells were calculated by absorbance of each well.

  • Day 5 proliferation multiples = Cell number of day 5/Cell number day 1

  • PC background interference = Day 5 proliferation multiples of infected cell treated with DMSO/Day 5 proliferation multiples of normal cell treated with DMSO

  • Multiples of inhibited proliferation = Day 5 proliferation multiples of infected cell treated with DMSO/Day 5 proliferation multiples of infected cell treated with CTX

  • Sensitization multiples = Multiples of inhibited proliferation of infected cell/Multiples of inhibited proliferation of normal cell

Experimental grouping

To evaluate the genes that were related to the CTX sensitivity in CRC cells, cell viability, cell growth and colony formation assays were performed. Caco-2 cells were divided into ten groups, namely, NC group (DMSO), NC+CTX 100 μg/ml group, MMP15 knockout group (DMSO), MMP15 knockout + CTX 100 μg/ml group, MRPL48 knockout group (DMSO), MRPL48 knockout + CTX 100 μg/ml group, CALN1 knockout group (DMSO), CALN1 knockout + CTX 100 μg/ml group, HADHB knockout group (DMSO) and HADHB knockout + CTX 100 μg/ml group.

Cell counting kit-8 assay

After transfection, Caco-2 cells were seeded in 96-well plates in triplicate at the density of 2 × 103 cells per well, followed by addition of CTX 100 μg/ml. After culturing for 24, 48, 72, 96 and 120 h, cell counting kit-8 (CCK-8) solution 100 μl (Sigma) was added into each well, and plates were incubated at 37°C for 1 h. Microplate reader was used to test optical density at the wavelength of 450 nm.

Colony formation assay

Colony formation assay was adopted to evaluate the effects of Caco-2 cells proliferation in vitro. Cells were plated in a six-well plate containing 1 × 103 cells per well in triplicate. After 2 weeks, the cells were fixed with 4% paraformaldehyde for 15 min and stained with 0.1% Crystal Violet. The visible colonies were counted.

Statistical analysis

Statistical analysis was performed by SPSS (version 20.0) and the experimental measurement data were expressed as mean ± SD. Statistical differences between groups were determined by one-way or two-way ANOVA. Differences were considered significant when P<0.05.

Growth curve results

From Figure 1A,B, the proliferation multiples of Caco-2 and HT-29 cells significantly increased in the first 4 days. However, on the 3rd day, the proliferation multiples of Caco-2 cells were higher than 2, while that of HT-29 cells was lower than 2, indicating that infected Caco-2 cells had a better prolificacy. Therefore, Caco-2 cells were selected for the following experiments.

Caco-2 cells had a better prolificacy

Figure 1
Caco-2 cells had a better prolificacy

Proliferation multiples of the infected (A) Caco-2 and (B) HT-29 cells.

Figure 1
Caco-2 cells had a better prolificacy

Proliferation multiples of the infected (A) Caco-2 and (B) HT-29 cells.

Close modal

Optimal concentration of puromycin

MTT assay results revealed that the percentage of viable Caco-2 cells treated with 1.25, 2.5, 5 and 10 μg/ml puromycin for 2 days were 16.3, 7.9, 7.6 and 7.6%, respectively. Therefore, the optimal lethal concentration of puromycin was 2.5 μg/ml (Figure 2).

The OD value of Caco-2 cells treated with puromycin at different concentrations (0, 1.25, 2.5, 5, 10 μg/ml)

Figure 2
The OD value of Caco-2 cells treated with puromycin at different concentrations (0, 1.25, 2.5, 5, 10 μg/ml)
Figure 2
The OD value of Caco-2 cells treated with puromycin at different concentrations (0, 1.25, 2.5, 5, 10 μg/ml)
Close modal

Optimal dosage of CTX

As shown in Figure 3, CTX suppressed Caco-2 cell proliferation in a dose-dependent manner, and the IC50 of CTX was 245.7 μg/ml. On the 14th day, when the CTX concentration was 50, 100, 200, 400, 600, 800 and 1200 μg/ml, the inhibitory rate was 11.67, 42.33, 43.67, 56.33, 57.67, 76.67 and 97.46%, respectively, indicating that CTX had a significant inhibitory function on Caco-2 cells in a dose-dependent manner. In the case that three CTX concentrations of 100, 200, 400 μg/ml had similar inhibitory effect and considering the high cost of CTX, CTX 100 μg/ml was selected as the optimal concentration for subsequent experiments.

The number of Caco-2 cells after CTX treatment at different concentrations (0, 50, 100, 200, 400, 600, 800, 1200 μg/ml)

Figure 3
The number of Caco-2 cells after CTX treatment at different concentrations (0, 50, 100, 200, 400, 600, 800, 1200 μg/ml)
Figure 3
The number of Caco-2 cells after CTX treatment at different concentrations (0, 50, 100, 200, 400, 600, 800, 1200 μg/ml)
Close modal

Optimal MOI results

After 48 h of infection with Caco-2 cells, we found that the fluorescence rate augmented with the increase in infection intensity in group A, but not in group B. When MOI was 0.5, the fluorescence rate was 43% in group A, and 86% in group B, with 50% Functional MOI. MOI = 0.5 was selected for the subsequent experiments (Table 1).

Table 1
The effect of different MOI on Functional MOI
Cell number/wellMOIMTT test value of group BMTT test value of group AFunctional MOI
6000 10 0.60 0.44 0.74 
6000 0.57 0.40 0.69 
6000 0.89 0.44 0.50 
6000 0.5 0.86 0.43 0.50 
6000 0.25 0.97 0.28 0.29 
Cell number/wellMOIMTT test value of group BMTT test value of group AFunctional MOI
6000 10 0.60 0.44 0.74 
6000 0.57 0.40 0.69 
6000 0.89 0.44 0.50 
6000 0.5 0.86 0.43 0.50 
6000 0.25 0.97 0.28 0.29 

Sequencing data quality analysis results

Sequencing data quality analysis was used to screen genes meeting the screening criteria (|FC Caco2| < 2, FCERBITUX ≤ −2; BF Caco2 < 0, BFERBITUX > 2) between Day 7 vs Day 0 and Day 14 vs Day 0. A total of 16 genes were obtained from the intersection in accordance with 209 genes acquired from the preliminary selection of Day 7 vs Day 0 and 307 genes obtained from the preliminary selection of Day 14 vs Day 0. In addition, another 30 genes were selected according to the screening criteria (day 14 ERBITUX logFC was sorted from small to large, Cacao2 logFC was less than 0.5) (Table 2). However, previous report has reported that BCR and GEM genes are closely related to CTX sensitivity, so we only focused on the remaining 44 genes [13].

Table 2
Candidate genes screened by sequencing data quality analysis
Entrez IDEntryCaco2 logFCERBITUX logFC
The intersecting genes of Day 7 vs Day 0 and Day 14 vs Day 0 
83698 CALN1 −0.151 −2.598 
1690 COCH 0.244 −2.701 
2824 GPM6B −0.041 −1.119 
25960 GPR124 0.137 −1.054 
4324 MMP15 −0.630 −3.279 
79570 NKAIN1 −0.232 −2.910 
403277 OR5K3 0.340 −1.837 
340990 OTOG −0.289 −3.768 
29944 PNMA3 0.072 −2.672 
5706 PSMC6 −0.344 −1.244 
29127 RACGAP1 0.137 −5.228 
2030 SLC29A1 −0.141 −1.166 
84851 TRIM52 0.264 −1.300 
51322 WAC −0.741 −3.744 
56949 XAB2 −0.556 −2.979 
80032 ZNF556 −0.433 −4.791 
The genes that met the screening criteria (screening criteria: day 14 ERBITUX logFC was sorted from small to large, Caco2 logFC was less than 0.5) 
4897 NRCAM −0.243 −5.262 
3554 IL1R1 0.053 −5.206 
402415 XKRX −0.030 −5.132 
613 BCR 0.041 −4.895 
51642 MRPL48 −0.102 −4.776 
5422 POLA1 −0.172 −4.679 
7579 ZSCAN20 −0.164 −3.717 
84455 EFCAB7 −0.051 −3.718 
90522 YIF1B 0.218 −3.371 
85480 TSLP 0.071 −3.486 
79065 ATG9A −0.203 −3.243 
130497 OSR1 −0.223 −3.196 
200132 TCTEX1D1 −0.231 −3.177 
64221 ROBO3 −0.188 −3.206 
92421 CHMP4C −0.194 −3.155 
1373 CPS1 0.013 −3.309 
79083 MLPH 0.105 −3.114 
23589 CARHSP1 −0.037 −3.149 
3646 EIF3E 0.165 −2.956 
2734 GLG1 −0.128 −2.984 
3032 HADHB 0.182 −2.920 
2669 GEM −0.198 −2.900 
58524 DMRT3 0.176 −2.873 
1608 DGKG −0.195 −2.814 
51116 MRPS2 −0.241 −3.365 
84530 SRRM4 0.228 −3.255 
138882 OR1N2 0.192 −3.145 
161424 NOP9− −0.185 −3.106 
5588 PRKCQ −0.227 −2.856 
4762 NEUROG1 0.012 −2.999 
Entrez IDEntryCaco2 logFCERBITUX logFC
The intersecting genes of Day 7 vs Day 0 and Day 14 vs Day 0 
83698 CALN1 −0.151 −2.598 
1690 COCH 0.244 −2.701 
2824 GPM6B −0.041 −1.119 
25960 GPR124 0.137 −1.054 
4324 MMP15 −0.630 −3.279 
79570 NKAIN1 −0.232 −2.910 
403277 OR5K3 0.340 −1.837 
340990 OTOG −0.289 −3.768 
29944 PNMA3 0.072 −2.672 
5706 PSMC6 −0.344 −1.244 
29127 RACGAP1 0.137 −5.228 
2030 SLC29A1 −0.141 −1.166 
84851 TRIM52 0.264 −1.300 
51322 WAC −0.741 −3.744 
56949 XAB2 −0.556 −2.979 
80032 ZNF556 −0.433 −4.791 
The genes that met the screening criteria (screening criteria: day 14 ERBITUX logFC was sorted from small to large, Caco2 logFC was less than 0.5) 
4897 NRCAM −0.243 −5.262 
3554 IL1R1 0.053 −5.206 
402415 XKRX −0.030 −5.132 
613 BCR 0.041 −4.895 
51642 MRPL48 −0.102 −4.776 
5422 POLA1 −0.172 −4.679 
7579 ZSCAN20 −0.164 −3.717 
84455 EFCAB7 −0.051 −3.718 
90522 YIF1B 0.218 −3.371 
85480 TSLP 0.071 −3.486 
79065 ATG9A −0.203 −3.243 
130497 OSR1 −0.223 −3.196 
200132 TCTEX1D1 −0.231 −3.177 
64221 ROBO3 −0.188 −3.206 
92421 CHMP4C −0.194 −3.155 
1373 CPS1 0.013 −3.309 
79083 MLPH 0.105 −3.114 
23589 CARHSP1 −0.037 −3.149 
3646 EIF3E 0.165 −2.956 
2734 GLG1 −0.128 −2.984 
3032 HADHB 0.182 −2.920 
2669 GEM −0.198 −2.900 
58524 DMRT3 0.176 −2.873 
1608 DGKG −0.195 −2.814 
51116 MRPS2 −0.241 −3.365 
84530 SRRM4 0.228 −3.255 
138882 OR1N2 0.192 −3.145 
161424 NOP9− −0.185 −3.106 
5588 PRKCQ −0.227 −2.856 
4762 NEUROG1 0.012 −2.999 

Cell viability by CellTiter-Glo luminescence assay

To measure the PC background interference, multiples of inhibited proliferation and sensitization multiples of the 44 genes, CellTiter-Glo luminescence assay was used to determine the proliferation of cells. Genes with PC background interference multiples between 0.7 and 1 (closer to 1 indicated less background interference), multiples of inhibited proliferation greater than 1.5, and sensitization multiples greater than 1.2 were selected. There were four genes meeting the selection criteria, namely MMP15, MRPL48, CALN1 and HADHB (Table 3).

Table 3
Cell viability test results of cells with different genes knocked out
TargetDay 5 proliferation multiplePC background interferenceProliferation inhibition multipleSensitizing multiple
NC 10.43 1.00 1.31 1.00 
NC + CTX 7.94    
MMP15 7.48 0.72 1.56 1.19 
MMP15 + CTX 4.80    
MRPL48 7.28 0.70 1.61 1.23 
MRPL48 + CTX 4.51    
CALN1 7.69 0.74 1.69 1.29 
CALN1 + CTX 4.54    
HADHB 8.33 0.80 1.71 1.30 
HADHB + CTX 4.88    
TargetDay 5 proliferation multiplePC background interferenceProliferation inhibition multipleSensitizing multiple
NC 10.43 1.00 1.31 1.00 
NC + CTX 7.94    
MMP15 7.48 0.72 1.56 1.19 
MMP15 + CTX 4.80    
MRPL48 7.28 0.70 1.61 1.23 
MRPL48 + CTX 4.51    
CALN1 7.69 0.74 1.69 1.29 
CALN1 + CTX 4.54    
HADHB 8.33 0.80 1.71 1.30 
HADHB + CTX 4.88    

CTX combined with genes knockout suppressed CRC cell viability and proliferation

From Figure 4A,B, knockout of MMP15, MRPL48, CALN1 and HADHB genes suppressed Caco-2 cell viability and proliferation. On day 7, the inhibitory rate of Caco-2 cells in MMP15 knockout + CTX 100 μg/ml, MRPL48 knockout + CTX 100 μg/ml, CALN1 knockout + CTX 100 μg/ml and HADHB knockout + CTX 100 μg/ml groups were 46.8, 52.68, 50.26 and 56.61, respectively, which were significantly higher than that in NC + CTX 100 μg/ml group (20.64%). These findings revealed that MMP15, MRPL48, CALN1 and HADHB genes knockout might facilitate the sensitivity of CRC cell line Caco-2 to CTX.

Caco-2 cells were divided into NC group, NC + CTX 100 μg/ml group, MMP15 knockout group, MMP15 knockout + CTX 100 μg/ml group, MRPL48 knockout group, MRPL48 knockout + CTX 100 μg/ml group, CALN1 knockout group, CALN1 knockout +CTX 100 μg/ml group, HADHB knockout group and HADHB knockout + CTX 100 μg/ml group

Figure 4
Caco-2 cells were divided into NC group, NC + CTX 100 μg/ml group, MMP15 knockout group, MMP15 knockout + CTX 100 μg/ml group, MRPL48 knockout group, MRPL48 knockout + CTX 100 μg/ml group, CALN1 knockout group, CALN1 knockout +CTX 100 μg/ml group, HADHB knockout group and HADHB knockout + CTX 100 μg/ml group

CCK-8 assays were used to determine the (A) viability and (B) proliferation of Caco-2 cells. (C) Colony formation assay showed that CTX 100 μg/ml combined with knockout of MMP15, MRPL48, CALN1 and HADHB genes impaired the colony formation capacity of Caco-2 cells. ****P<0.0001 vs NC group; ##P<0.01, ####P<0.0001 vs gene knockout alone group.

Figure 4
Caco-2 cells were divided into NC group, NC + CTX 100 μg/ml group, MMP15 knockout group, MMP15 knockout + CTX 100 μg/ml group, MRPL48 knockout group, MRPL48 knockout + CTX 100 μg/ml group, CALN1 knockout group, CALN1 knockout +CTX 100 μg/ml group, HADHB knockout group and HADHB knockout + CTX 100 μg/ml group

CCK-8 assays were used to determine the (A) viability and (B) proliferation of Caco-2 cells. (C) Colony formation assay showed that CTX 100 μg/ml combined with knockout of MMP15, MRPL48, CALN1 and HADHB genes impaired the colony formation capacity of Caco-2 cells. ****P<0.0001 vs NC group; ##P<0.01, ####P<0.0001 vs gene knockout alone group.

Close modal

Clonal formation results

To verify the sensitization effect of the above four gene knockout and evaluate its long-term sensitization effect, the clonal formation ability of Caco-2 cells was further tested through cell cloning experiment. As shown in Figure 4C, MMP15, MRPL48, CALN1 and HADHB genes knockout alone reduced the cloning potential of Caco-2 cells. On this basis, the addition of CTX 100 μg/ml inhibited the clonal formation of Caco-2 cells more significantly than the gene knockout alone group (P<0.0001).

In the present study, GeCKO screening and cell viability test results revealed that MMP15, MRPL48, CALN1 and HADHB genes might be involved in drug sensitivity to CTX in CRC cell (Caco-2). This finding suggested that MMP15, MRPL48, CALN1 and HADHB genes were potential targets for the treatment of CRC, which fills in the gap of genes responsible for CTX sensitivity in CRC.

CRISPR-Cas9 forms a complex by using nuclease Cas9 protein and sgRNA [14]. To our knowledge, sgRNA determines the specificity of target sequence through base complementary pairing; Cas9 protein acts as nuclease to cut genomic DNA complementary to the spacers on the sgRNA, causing double-stranded DNA damage, thereby introducing genetic mutations through mechanisms [15]. As the third generation of gene editing technology, CRISPR-Cas9 has attracted wide attention due to its advantages of simple operation, flexible design, low price and multi-point editing. With the continuous development and improvement of CRISPR-Cas9 system, a series of drug-resistance genes or virus-resistance genes have been screened [16]. Chen and Zhang have found that CRISPR-Cas9 can revert resistance gene mutations [17]. Zhang et al. also believed that CRISPR-Cas9 could be used to construct drug-resistant cancer cell lines for drug screening [18]. In this study, we preliminarily obtained 44 candidate genes in Caco-2 CRC cells related to CTX resistance by CRISPR-Cas9. Subsequently, based on the selection criteria of PC background interference multiples between 0.7 and 1, multiples of inhibited proliferation greater than 1.5, and sensitization multiples greater than 1.2, four genes including MMP15, MRPL48, CALN1 and HADHB were selected via CellTiter-Glo luminescence assay.

Metalloproteinases (MMPs) is a family of proteolytic enzymes that can maintain the dynamic equilibrium of extracellular matrix by degrading and remodeling extracellular matrix [19]. Several studies have shown that MMPs are related to inflammation, atherosclerosis, liver cirrhosis, connective tissue disease and cancer [20,21]. As the active center of mitochondrial ribosomes, mitochondrial ribosomal proteins (MRPs) are the basis for the normal expression of mitochondrial DNA and responsible for translation of encoding proteins of mitochondrial DNA [22]. Wang et al. have noticed that down-regulation of MRPL18 gene expression can cause metabolic disorders in mouse insulinoma cells (MIN6) [23]. It is well known that changes in the concentration and activity of intracellular Ca2+ have a certain effect on the generation of drug resistance in tumors [24]. Human CALN1 gene is only expressed in the brain, especially in the hippocampus and cerebral cortex. Sequence alignment done by Wu et al. has showed that CALN1 gene is highly similar to the ubiquitous calcium mediator protein, calmodulin [25]. Calmodulin is the principle mediator of the Ca2+ signal found in many eukaryotic cells [26]. The proliferation, migration and metastasis of tumor cells require a large amount of energy, which means they need a more vigorous metabolism than normal cells. In addition to sugars that provide energy for the active proliferation of tumor cells, fatty acids can also produce ATP through β-oxidation [27]. HADHB mainly participates in the last three steps of fatty acid β-oxidation, including hydration of 2-dienoyl-CoA, dehydrogenation of fatty acid-2-hydroxyacyl-CoA, and thiolysis of β-ketoacyl CoA [28]. With a 3-year follow-up of 91 patients diagnosed with hilar cholangiocarcinom, Zhang et al. have found that HADHB not only shows high expression in hilar cholangiocarcinom, but also have a close relationship with tumor infiltration and lymphatic metastasis [29]. Nevertheless, no studies have shown the relationship between MMP15, MRPL48, CALN1 and HADHB genes and CTX sensitivity.

On the basis of our prophase research, we speculated that MMP15, MRPL48, CALN1 and HADHB genes may be related to CTX sensitivity in CRC. To verify the idea, CCK-8 assay was performed on cells that had MMP15, MRPL48, CALN1 and HADHB genes knocked out. Results showed that MMP15, MRPL48, CALN1 and HADHB genes knockout alone remarkably suppresed viability and proliferation of Caco-2 cells, and the addition of CTX 100 μg/ml inhibited cell viability and proliferation more significantly than the gene knockout alone group. Moreover, the sensitization effect of the above four gene knockout was further confirmed by clonal formation assay. Taken together, these data revealed that MMP15, MRPL48, CALN1 and HADHB genes knockout might facilitate the sensitivity of CRC cell line Caco-2 to CTX.

In summary, the present study confirms that MMP15, MRPL48, CALN1 and HADHB genes are responsible for CTX sensitivity in CRC, which is the first time to explore genes related to CTX sensitivity by CRISPR-Cas9, providing a theoretical basis for further research on drug-sensitivity mechanism of CRC.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported by the National Natural Science Foundation of China [grant number 81572291]; the Zhejiang Provincial Natural Science Foundation of China [grant number LY19H160022]; and the Wenzhou Scientific Research Project [grant number Y20190424].

T.-t.H. contributed to the conception and design of the present study. J.-w.Y. and Y.Y. contributed to the experiment and analysis of the data. Y.-y.C., H.-b.X. and Y.-q.X. contributed to the analysis and interpretation of the data. L.-c.Y. wrote the first draft of manuscript. All authors critically revised and approved the content of the final manuscript.

CCK-8

cell counting kit-8

CRC

colorectal cancer

CRISPR

clustered regularly interspaced short palindromic repeats

CTX

cetuximab (Erbitux)

DMSO

dimethyl sulfoxide

GeCKO

genome-scale CRISPR-Cas9 knockout

MOI

multiplicity of infection

MTT

3-(4,5-dimethylthiazol-2-yl)2,5-diphenyl tetrazolium bromide

NC

normal control

sgRNA

single-guide RNA

1.
Qin
H.
,
Song
C.
and
Tian
W.
(
2018
)
mir106a regulates cell proliferation and apoptosis of colon cancer cells through targeting the PTEN/PI3K/AKT signaling pathway
.
Oncol. Lett.
15
,
3197
3201
2.
Dekker
E.
,
Tanis
P.
,
Vleugels
J.
,
Kasi
P.
and
Wallace
M.
(
2019
)
Colorectal cancer
.
Lancet
394
,
1467
1480
[PubMed]
3.
Nassar
D.
and
Blanpain
C.
(
2016
)
Cancer stem cells: basic concepts and therapeutic implications
.
Annu. Rev. Pathol.
11
,
47
76
4.
Liu
X.
,
Tao
X.
,
Xiuxiu
H.
,
XiaoXiang
C.
,
Kaixuan
Z.
,
Li
S.
et al.
,
Elevated circulating miR-182 acts as a diagnostic biomarker for early colorectal cancer
.
Cancer Manag. Res.
10
,
857
865
[PubMed]
5.
Park
S.
,
Hwang
C.
,
Cho
S.
,
Lee
D.
,
Gong
J.
,
Lee
S.
et al.
(
2019
)
Systems analysis identifies potential target genes to overcome cetuximab resistance in colorectal cancer cells
.
FEBS J.
286
,
1305
1318
[PubMed]
6.
Han
Y.
,
Peng
Y.
,
Fu
Y.
,
Cai
C.
,
Guo
C.
,
Liu
S.
et al.
(
2020
)
MLH1 deficiency induces cetuximab resistance in colon cancer via Her-2/PI3K/AKT signaling
.
Adv. Sci.
7
,
2000112
7.
Dede
K.
,
Salamon
F.
,
Landherr
L.
,
Jakab
F.
and
Bursics
A.
(
2015
)
Pathologic assessment of response to chemotherapy in colorectal cancer liver metastases after hepatic resection: which method to use?
Pathol. Oncol. Res.
21
,
173
179
8.
Xiang
M.
,
Holsinger
F.C.
,
Colevas
A.D.
,
Chen
M.M.
,
Le
Q.T.
and
Beadle
B.M.
(
2018
)
Survival of patients with head and neck cancer treated with definitive radiotherapy and concurrent cisplatin or concurrent cetuximab: A Surveillance, Epidemiology, and End Results-Medicare analysis
.
Cancer
124
,
4486
4494
[PubMed]
9.
Segelov
E.
,
Thavaneswaran
S.
,
Waring
P.
,
Desai
J.
,
Robledo
K.
,
Gebski
V.
et al.
(
2016
)
Response to cetuximab with or without irinotecan in patients with refractory metastatic colorectal cancer harboring the KRAS G13D mutation: Australasian Gastro-Intestinal Trials Group ICECREAM Study
.
J. Clin. Oncol.
34
,
2258
2264
[PubMed]
10.
Strich
J.R.
and
Chertow
D.S.
(
2019
)
CRISPR-Cas biology and its application to infectious diseases
.
J. Clin. Microbiol.
57
,
e1307
18
11.
Minkenberg
B.
,
Wheatley
M.
and
Yang
Y.
(
2017
)
CRISPR/Cas9-Enabled Multiplex Genome Editing and Its Application
.
Prog. Mol. Biol. Transl. Sci.
149
,
111
132
12.
Joung
J.
,
Konermann
S.
,
Gootenberg
J.S.
,
Abudayyeh
O.O.
,
Platt
R.J.
,
Brigham
M.D.
et al.
(
2017
)
Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening
.
Nat. Protoc.
12
,
828
863
[PubMed]
13.
Jeitany
M.
,
Leroy
C.
,
Tosti
P.
,
Lafitte
M.
,
Le Guet
J.
,
Simon
V.
et al.
(
2018
)
Inhibition of DDR1-BCR signalling by nilotinib as a new therapeutic strategy for metastatic colorectal cancer
.
EMBO Mol. Med.
10
,
e7918
[PubMed]
14.
Vladimir
M.
,
Leonid
M.
,
Ekaterina
S.
,
Konstantin
K.
and
Konstantin
S.
(
2016
)
Kinetics of the CRISPR-Cas9 effector complex assembly and the role of 3′-terminal segment of guide RNA
.
Nucleic Acids Res.
6
,
2837
45
15.
Plummer
R.J.
,
Guo
Y.
and
Peng
Y.
(
2017
)
A CRISPR reimagining: new twists and turns of CRISPR beyond the genome-engineering revolution: production of SCD1 transgenic mice
.
J. Cell. Biochem.
119
,
1299
[PubMed]
16.
Bi
Y.
,
Shen
W.
,
Min
M.
and
Liu
Y.
(
2017
)
MicroRNA-7 functions as a tumor-suppressor gene by regulating ILF2 in pancreatic carcinoma
.
Int. J. Mol. Med.
39
,
900
906
[PubMed]
17.
Chen
Y.
and
Zhang
Y.
(
2018
)
Application of the CRISPR/Cas9 system to drug resistance in breast cancer
.
Adv. Sci.
5
,
1700964
[PubMed]
18.
Zhang
L.
,
Li
Y.
,
Chen
Q.
,
Xia
Y.
,
Zheng
W.
and
Jiang
X.
(
2018
)
The construction of drug-resistant cancer cell lines by CRISPR/Cas9 system for drug screening
.
Sci. Bull.
21
,
1411
1419
19.
Weinreb
R.N.
,
Robinson
M.R.
,
Dibas
M.
and
Stamer
W.D.
(
2020
)
Matrix metalloproteinases and glaucoma treatment
.
J. Ocular Pharmacol. Ther.
36
,
208
228
20.
Huang
C.Y.
,
Tseng
K.C.
,
Lin
M.N.
,
Tsai
J.P.
and
Su
C.C.
(
2015
)
Plasma levels of matrix metalloproteinase-2 and -9 in male and female patients with cirrhosis of different aetiologies
.
J. Clin. Pathol.
68
,
917
[PubMed]
21.
Shirian
J.
,
Arkadash
V.
,
Cohen
I.
,
Sapir
T.
,
Radisky
E.S.
,
Papo
N.
et al.
(
2018
)
Converting a broad matrix metalloproteinase family inhibitor into a specific inhibitor of MMP and MMP
,
FEBS Lett.
592
,
1122
1134
22.
Zhang
L.
,
Lu
P.
,
Yan
L.
,
Yang
L.
,
Wang
Y.
,
Chen
J.
et al.
(
2019
)
MRPL35 is up-regulated in colorectal cancer and regulates colorectal cancer cell growth and apoptosis
.
Am. J. Pathol.
189
,
1105
1120
[PubMed]
23.
Wang
X.
,
Li
H.
,
De Leo
D.
,
Guo
W.
,
Koshkin
V.
,
Fantus
I.G.
et al.
(
2004
)
Gene and protein kinase expression profiling of reactive oxygen species-associated lipotoxicity in the pancreatic beta-cell line MIN6
.
Diabetes
53
,
129
140
[PubMed]
24.
Büsselberg
D.
and
Florea
A.-M.
(
2017
)
Targeting intracellular calcium signaling ([Ca2+] i) to overcome acquired multidrug resistance of cancer cells: a mini-overview
.
Cancers (Basel)
9
,
48
25.
Wu
Y.Q.
,
Lin
X.
,
Liu
C.M.
,
Jamrich
M.
and
Shaffer
L.G.
(
2001
)
Identification of a human brain-specific gene, calneuron 1, a new member of the calmodulin superfamily
.
Mol. Genet. Metab.
72
,
343
350
[PubMed]
26.
Munk
M.
,
Alcalde
J.
,
Lorentzen
L.
,
Villalobo
A.
,
Berchtold
M.W.
and
Panina
S.
(
2020
)
The impact of calmodulin on the cell cycle analyzed in a novel human cellular genetic system
.
Cell Calcium
88
,
102207
[PubMed]
27.
Liu
Y.
,
Neumann
D.
,
Glatz
J.F.C.
and
Luiken
J.
(
2018
)
Molecular mechanism of lipid-induced cardiac insulin resistance and contractile dysfunction
.
Prostaglandins Leukot. Essent. Fatty Acids
136
,
131
141
[PubMed]
28.
Das
A.M.
,
Illsinger
S.
,
Lücke
T.
,
Hartmann
H.
,
Ruiter
J.P.
,
Steuerwald
U.
et al.
(
2006
)
Isolated mitochondrial long-chain ketoacyl-CoA thiolase deficiency resulting from mutations in the HADHB gene
.
Clin. Chem.
52
,
530
534
[PubMed]
29.
Zhang
H.
,
Liao
J.
,
Yang
X.
,
Aimin
D.U.
,
Wenlong
Y.U.
and
Gastroenterology
D.O.
(
2017
)
Clinical significance of HADHA and HADHB expression in hilar cholangiocarcinoma
.
Chin. Clin. Oncol.
22
,
607
611

Author notes

*

These authors contributed equally to this work.

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).