Open Access

MicroRNA‑24 alleviates colorectal cancer progression via a rs28382740 single nucleotide polymorphism in the long noncoding region of X‑linked inhibitor of apoptosis protein

  • Authors:
    • Yu-Feng Tian
    • Chi-Jung Huang
    • Chih-Yi Liu
    • Shung-Haur Yang
    • Chih-Sheng Hung
    • Kai-Yuan Lin
    • Ching-Long Lai
    • Chun-Chao Chang
  • View Affiliations

  • Published online on: October 4, 2024     https://doi.org/10.3892/ol.2024.14724
  • Article Number: 591
  • Copyright: © Tian et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is one of the most prevalent malignant diseases worldwide. Recurrence is associated with the poor survival of patients with CRC. Targeted therapy and precision medicine for recurrent CRC may improve the clinical outcome. Therefore, finding biomarkers that can detect CRC early, assess its prognosis and survival, and predict its treatment response is key to improving the clinical prognosis. The aim of this study was to assess CRC recurrence by analyzing molecular differences using postoperative specimens. Whole‑exome sequencing was first used to evaluate the molecular differences in CRC tissues from patients with recurrent disease, and the results were then verified with tissue array methods. The regulation of single nucleotide polymorphisms (SNPs) in long noncoding regions of interest was analyzed in the presence of target microRNAs (miRs) using luciferase assays. The results demonstrated that in patients with recurrent CRC, the G allele was mainly detected at the rs28382740 SNP in the 3'‑untranslated region of the X‑linked inhibitor of apoptosis (XIAP)‑encoding gene. From the tissue arrays, 60% (3/5) of patients with the G allele of the rs28382740 SNP were diagnosed with CRC recurrence, whilst only 10% (1/10) of patients without the G allele had recurrent CRC (P=0.077). Furthermore, XIAP levels were high in non‑CRC (50%; 2/4) and CRC (75%; 3/4) tissues of patients with recurrent disease and CRC (54.5%; 6/11) tissues of patients without recurrent disease. However, but only 9.1% (1/11) of non‑CRC tissues of nonrecurrent patients had significantly high XIAP expression levels (P=0.022). Using a luciferase assay, it was demonstrated that miR‑24s (miR‑24‑1‑5p and miR‑24‑2‑5p) targeting the rs28382740 SNP reduced XIAP levels in CRC cells with rs28382740 SNP genotype G. These results indicate that apoptosis‑related proteins, such as XIAP, may be therapeutic targets or biomarkers for tumor development. The data from the present study support an inhibitory effect of miR‑24s on XIAP expression. However, this inhibitory potency depends on the rs28382740 SNP genotype and may alleviate CRC progression by regulating the expression of XIAP.

Introduction

Colorectal cancer (CRC) accounts for about one in 10 cancer cases and deaths worldwide (1). Recurrence, which contributes to its poor prognosis by increasing 5-year CRC-related mortality to 33.6%, is frequently observed in most patients undergoing curative treatment or resection (2,3). Clinically, the delayed diagnosis and treatment failure of CRC remain the main reasons for the poor prognosis. Therefore, appropriate biomarkers that can detect CRC early, assess its prognosis and survival, and predict its treatment response are key to improving its clinical prognosis (4). Furthermore, an understanding of the postoperative follow-up program for CRC recurrence is important (5). For instance, using a liquid biopsy to monitor or evaluate recurrent CRC has been frequently reported (6). In addition, mutational status in formalin-fixed paraffin-embedded (FFPE) blocks showing high concordance may indicate the current reality or possible future prognosis of patients with CRC (7,8).

Several biomarkers modulating apoptosis have been described for their prognostic value for CRC recurrence. These biomarkers may provide new insights into monitoring CRC recurrence and therapeutic targets (912). Increased expression of an inhibitor of the apoptosis protein (IAP) family is involved in colon tumorigenesis (13). In contrast, the downregulation of IAPs by modulating molecules in the tumor microenvironment, such as tumor necrosis factor-a, may repress tumor growth (14). Therefore, certain apoptosis-related proteins, including X-linked IAP (XIAP), cIAP1, cIAP2 and survivin, may have potential as biomarkers for tumor development, including in CRC (1315). Dysregulation of these antiapoptotic molecules has been reported to promote tumorigenesis in humans (16). The most important of these is XIAP, which is an emerging therapeutic target for different human cancers (1720). Notably, inhibition of XIAP expression has been reported to control the proliferation and invasion of CRC cells, especially in metastatic CRC (21,22). Therefore, identifying potent XIAP antagonists [such as drugs, antisense oligonucleotides or microRNAs (miRs)] may have clinical implications for cancers that overexpress XIAP (2325).

Dysregulation of small noncoding RNAs, such as miRs, contributes to the pathogenesis of all types of cancer (26). Exosomal miRs in serum or plasma are potential biomarkers for the diagnosis and prognosis of CRC (27,28). Furthermore, certain miRs may be potential candidate targets for treating CRC (29) and provide an attractive antitumor approach to cancer therapy (30). These miRs may act as tumor suppressors (31).

The aim of the present study was to evaluate the molecular differences in recurrent CRC via whole-exome sequencing (WES) and assess the role of XIAP. Luciferase expression was used to assess the regulation of single nucleotide polymorphisms (SNPs) in the long noncoding region of XIAP with target miRs. It is hoped that the information obtained may lead to improved and new treatment strategies to improve the prognosis of CRC.

Materials and methods

CRC sample acquisition

Delinked FFPE tissue samples from 9 patients with CRC [nonrecurrent, n=4 and recurrent cases, n=5; American Joint Committee on Cancer (AJCC) stage II; median (range) time to recurrence, 4.1 (1.9–7.0) months; Table I] were obtained from the Department of Pathology at Taipei Veterans General Hospital (Taipei, Taiwan). The protocol for the present study was reviewed and approved by the Institutional Review Board (IRB) of Taipei Veterans General Hospital (approval no. 2017-07-030AC). In addition, paired archived FFPE samples (non-CRC and CRC tissues) from 15 patients (AJCC stage II, n=7 and AJCC stage III, n=8; Table II) for fabrication into tissue arrays were acquired from the Cathay General Hospital Biobank (Taipei, Taiwan), which approved their use (approval no. HBKEC-20200928-1). The IRB of the Cathay General Hospital exempted the obtaining of informed consent for tissue procurement through the Cathay General Hospital Biobank after an anonymous unlinked process (approval no. CGH-P108136). The time period of Taipei Veterans General Hospital tissue samples collection was between February 2009 and December 2015, whilst the period for the Cathay General Hospital was between January 2000 and July 2020. The histological diagnosis of CRC in the present study was made by certified anatomical pathologists at Taipei Veterans General Hospital or Cathay General Hospital. The present study was performed in accordance with the Declaration of Helsinki, and all clinical characteristics of patients, including sex, onset age, primary or recurrent, initial AJCC stage and follow-up information, were obtained retrospectively.

Table I.

Clinical characteristics of non-recurrent (n=4) and recurrent (n=5) patients for next-generation sequencing analysis.

Table I.

Clinical characteristics of non-recurrent (n=4) and recurrent (n=5) patients for next-generation sequencing analysis.

CharacteristicNo recurrenceRecurrence
Sex, n (%)
  Female1 (25.0)0 (0.0)
  Male3 (75.0)5 (100.0)
Median onset age57.374.8
(range), years(45.3–74.1)(62.8–81.9)
Median time to recurrence (range), months-4.1 (1.9–7.0)
Stage, n (%)
  T3N0M04 (100.0)5 (100.0)
  AJCC II4 (100.0)5 (100.0)

[i] T, tumor; N, node; M, metastasis; AJCC, American Joint Committee on Cancer.

Table II.

Clinical characteristics of non-recurrent (n=11) and recurrent (n=4) patients for the tissue array.

Table II.

Clinical characteristics of non-recurrent (n=11) and recurrent (n=4) patients for the tissue array.

CharacteristicNo recurrenceRecurrence
Sex, n (%)
  Female6 (54.5)1 (25.0)
  Male5 (45.5)3 (75.0)
Median age of onset71.361.0
(range), years(34.6–84.5)(48.5–78.0)
Median time to-102.0
recurrence (range), months (7.3–174.1)
T stage, n (%)
  T310 (90)4 (100.0)
  T41 (9.1)0 (0.0)
N stage, n (%)
  N06 (54.5)1 (25.0)
  N12 (18.1)3 (75.0)
  N23 (27.3)0 (0.0)
AJCC stage, n (%)
  II6 (54.5)1 (25.0)
  III5 (45.5)3 (75.0)

[i] T, tumor; N, node; M, metastasis; AJCC, American Joint Committee on Cancer.

DNA purification from FFPE colonic tissues

Genomic DNA from FFPE colonic tissues was extracted and purified using the High Pure FFPET DNA Isolation Kit (cat. no. 06650767001, Roche Diagnostics). Briefly, the fixed paraffin-embedded colonic tissue sections (10-mm thick) were immersed in xylene to remove the extra paraffin and rehydrated with 100% alcohol at room temperature. After RNase treatment, the genomic DNA was quantified using a NanoDrop ND-1000 Spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc.) and the quality and integrity of the genomic DNA was verified using the Qubit dsDNA Quantification Assay Kit (cat. no. Q32851; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions.

Library construction and sequencing by WES

To generate standard exome capture libraries, the Agilent SureSelect XT Reagent kit (cat. no. G9611A; Agilent Technologies, Inc.) for the Illumina Multiplexed Paired-End sequencing library was used with the SureSelect XT Clinical Research exome V2 (cat. no. 5190-9492; Agilent Technologies, Inc.) probe set. A total of 1 mg genomic DNA was used to construct a library with the Agilent SureSelect XT Reagent kit. The amplification adapter-ligated sample was purified using Agencourt AMPure XP beads (cat. no. A63882; Beckman Coulter, Inc.) and analyzed on a TapeStation 4200 D1000 screentape (Agilent Technologies, Inc.). A 750 ng-genomic DNA library was prepared for hybridization with the capture baits, and the sample was hybridized for 24 h at 65°C, captured with Dynabeads MyOne Streptavidin T1 (cat. no. 65602; Thermo Fisher Scientific, Inc.), and purified using Agencourt AMPure XP beads. The Agilent protocol was used to add index tags by posthybridization amplification. Finally, all samples were sequenced on an Illumina NovaSeq 6000 platform (cat. no. 20012850; Illumina, Inc.) using the 150PE protocol. The sequence reads are deposited in the National Center for Biotechnology Information (NCBI) under the accession number PRJNA-1063437.

Variant analysis of sequencing data

The qualified read data were then processed through genomic alignment against the Ensembl database (version 86; http://ftp.ensembl.org/pub/release-86) and using the Burrows-Wheeler Aligner (version 0.7.17; http://github.com/lh3/bwa) to obtain basic sequence information (32,33). The Genome Analysis Toolkit (version 3.7.0) was used to analyze variants (34,35), and the Variant Effect Predictor (version 86; http://github.com/Ensembl/ensembl–vep/releases?page=7) predicted the effects SNPs on proteins (36,37). Paired-end reads were quality-checked using FastQC (version 0.32; http://github.com/s–andrews/FastQC/releases) and trimmed using Trimmomatic (version 0.11.7; http://github.com/usadellab/Trimmomatic/releases) (38). Finally, the association between the sequencing data and nonrecurrent and recurrent CRC was assessed.

CRC cell lines and their rs28382740 SNP genotypes

In the present study, four CRC cells classed as AJCC stage II, LS 123 [cat. no. CCL-255; American Type Culture Collection (ATCC)], HCT 116 (cat. no. CCL-247; ATCC), LS 174T (cat. no. CL-188; ATCC) and SW480 (cat. no. CCL-228; ATCC), and two CRC cells classed as AJCC stage III, SW620 (cat. no. CCL-227; ATCC) and LoVo (cat. no. CCL-229; ATCC), were cultured according to the protocol of the ATCC. Briefly, all cells were cultured to 80% confluence with specific culture medium [LS 123 and LS 174T: Minimum Essential Medium (cat. no. 41500034; Thermo Fisher Scientific, Inc.); HCT 116 and LoVo: Dulbecco's Modified Eagle's Medium (cat. no. 12800017; Thermo Fisher Scientific, Inc.); SW480 and SW620: Leibovitz's L-15 Medium (cat. no. 41300039; Thermo Fisher Scientific, Inc.)] in the presence of 10% fetal bovine serum (cat. no. A06806-35; NQBB International biological Corp.) and 1X antibiotic-antimycotic solution (100 units/ml of penicillin, 100 µg/ml of streptomycin and 0.25 µg/ml of amphotericin B; cat. no. 15240062; Thermo Fisher Scientific, Inc.) at 37°C and 5% CO2, except that SW480 and SW620 cells were cultured in the absence of CO2. A 170-bp fragment containing the rs28382740 SNP in the 3′-untranslated region (3′-UTR) of XIAP from these cell lines was PCR-amplified in the presence of 0.25 units of AmpliTaq Gold 360 DNA Polymerase (cat. no. 4398833; Thermo Fisher Scientific. Inc.), 2.0 mM MgCl2, 50 ng of genomic DNA and 1 µM primer pair (Table III) in a 10-µl reaction mixture. The PCR program was as follows: Denaturation at 95°C for 10 min, a 45-cycle program (95°C, 30 sec; 60°C, 30 sec; 72°C, 30 sec), and a final extension at 72°C for 7 min. Sequences of the rs28382740 SNP were then determined by Sanger sequencing.

Table III.

Primers for the amplification, sequencing and cloning of DNA fragments containing rs28382740 single nucleotide polymorphism of X-linked inhibitor of the apoptosis protein.

Table III.

Primers for the amplification, sequencing and cloning of DNA fragments containing rs28382740 single nucleotide polymorphism of X-linked inhibitor of the apoptosis protein.

Use of primerDirectionSequence (5′-3′)Product length, bp
Sequencing of DNA fragments within the 3′-UTRForward TGACAAGTGTCCCATGTGCT170
Reverse TTGGTAGCAAATGCTAATGGAA
Cloning 3′-UTR fragment Forward-1aACTAGTTCTAACTCTATAGTAGGCATG604
Forward-2bACTAGTTCTAACTCTATGGTAGGCATG
ReverseGTTTAAACATCATTAAATACGCTTTCAA

a Used to amplify genotype A;

b Used to amplify genotype G. The rs28382740 SNP is indicated by underlined fonts; SpeI (ACTAGT) and PmeI (GTTTAAAC) are indicated by italics and underlined fonts. 3′-UTR, 3′-untranslated region.

Hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) and interpretation

Tissues were immersed in 4% formaldehyde for 1 day at room temperature, processed in Tissue-Tek VIP 5 (Sakura Finetek USA, Inc.) for dehydration, and embedded in Paraplast Plus (cat. no. 39602004; Leica Biosystems, Inc.) to form a donor block. Tissue arrays were then constructed by removing a core of tissue from a donor block and transferring this core to a predetermined position on a recipient block. Sections of 5 mm were cut from the blocks of tissue arrays, stained with H&E and immunostained using an avidin-biotin-immunoperoxidase method. Briefly, H&E staining was performed using a Tissue-Tek DRS 2000 Automated Slide Stainer (Sakura Finetek USA, Inc.) following a general and serial protocol at room temperature: Deparaffinization (two consecutive xylene changes of 5 min each, followed by a 7 min change), rehydration [a sequential alcohol changes (100% alcohol, 60 sec; 100% alcohol, 90 sec; 95% alcohol, 60 sec; 75% alcohol, 60 sec) and running water, 3 min] and staining with hematoxylin for 5 min, followed by dipping the slides 5 times in 1% acid alcohol (1% HCl in 70% alcohol). Before mounting the tissue sections, the sections were rinsed, stained with eosin for 3 min at room temperature, dehydrated with graded alcohol, and washed in xylene.

IHC assays were performed on a BenchMark GX IHC/ISH slide automated system (Ventana Medical Systems, Inc.). The automated IHC program included deparaffinization with EZ Prep solution (cat. no. 950-102; Ventana Medical Systems, Inc.) at 75°C for 8 min, antigen retrieval with Cell Conditioning 1 solution (cat. no. 950-124; Ventana Medical Systems, Inc.) at 95°C for 64 min, and incubation with primary anti-XIAP antibody (dilution 1:100; cat. no. SC-55550; Santa Cruz Biotechnology, Inc.) at 37°C for 1 h. Finally, positive signals were developed following an incubation with secondary antibody and chromogen (OptiView DAB IHC Detection Kit; cat. no. 760-700; Roche Diagnostics). The Reaction Buffer (cat. no. 950-300; Ventana Medical Systems, Inc.) was used for all wash steps at room temperature. All sections were further processed by counterstaining with hematoxylin II (cat. no. 790-2208; Ventana Medical Systems, Inc.) for 8 min at room temperature and Bluing Reagent (cat. no. 760-2037; Ventana Medical Systems, Inc.) for 4 min at room temperature, and then visualized using light microscopy (Olympus BX41 Microscope; Olympus Corp.).

Sections were evaluated at a high magnification by a research pathologist blinded to tissue type to determine the proportion of cells expressing XIAP, and the tissue sections were scored using a semiquantitative method based on a four-layer system: Score 0 for negative expression; score 1 for weakly positive expression; score 2 for positive expression; and score 3 for strongly positive expression (39).

Knockdown of XIAP in SW480 cells and cell proliferation assessment

For XIAP knockdown in SW480 cells, the plasmid (pLKO_005) carrying a non-targeting control lentivirus-mediated small hairpin (sh)RNA (cat. no. TRCN0000231719; shLuc; 5′-GCGGTTGCCAAGAGGTTCCAT-3′) or a specific lentivirus-mediated shRNA targeting XIAP (shXIAP; cat. no. TRCN0000231579; 5′-ACACGTACTTGTGCGAATTAT-3′) was purchased from the National RNAi Core Facility of Academia Sinica, Taiwan. The first-generation lentiviral vectors were used to package lentiviruses. Infection of each lentivirus into SW480 cells and selection of stable clones with shLuc (shLuc-SW480) or with shXIAP (shXIAP-SW480) by puromycin and efficacy validation of XIAP knockdown were performed according to a previous protocol (TRC protocol: Lentivirus infection V3) (40). After washing cells with PBS, bright-field images of live cells were taken using an ECHO Revolve microscope (ECHO RVL-100-M; BICO Group AB). To determine cell proliferation, images were analyzed using QuPath (version 0.3.0; http://qupath.github.io) and Adobe Photoshop 2022 (version 23.0.1; Adobe Systems, Inc.) to obtain the daily coverage area of cells (41,42).

Cloning of 3′-UTR regions of XIAP into a pMIR-REPORT vector and luciferase assay

The pMIR-REPORT miRNA Expression Reporter Vector System (cat. no. AM5795; Thermo Fisher Scientific. Inc.) was used to analyze the potential target of miR-24 in the 3′-UTR of XIAP, including the significant rs28382740 SNP. Fragments of 608 bp in length with different genotypes at the rs28382740 SNP within this long noncoding region were amplified from genomic DNA of HCT116 (type A on the rs28382740 SNP) in the presence of 1.25 units of GoTaq DNA Polymerase (cat. no. M3001; Promega Corp.), 2.5 mM MgCl2, 50 ng of genomic DNA and 0.8 µM appropriate primer pair (Table III) in a 10-µl reaction mixture using the following PCR program: Denaturation at 95°C for 10 min, a 45-cycle program (95°C, 30 sec; 60°C, 30 sec; 72°C, 50 sec) and a final extension at 72°C for 7 min. Amplified fragments were cloned into the SpeI and PmeI restriction sites of the pMIR-REPORT vector with T4 DNA ligase (cat. no. T4L0500; Bioman Scientific Co., Ltd.) for 16 h at 4°C. Subsequently, changes in the luciferase activity of two pMIR-REPORT vectors (pMIR-REPORT-A and pMIR-REPORT-G) in HCT116 cells were assessed by cotransfection of pCMV-MIR vectors (cat. no. PCMVMIR; OriGene Technologies, Inc.) with miR-24-1 precursor (cat. no. SC400296; OriGene Technologies, Inc.) or miR-24-2 precursor (cat. no. SC400297; OriGene Technologies, Inc.), and a pMIR-REPORT β-galactosidase (β-gal) control plasmid (cat. no. AM5795; Thermo Fisher Scientific. Inc.). Briefly, 7.5×103 HCT116 cells/well were seeded on to a 96-well plate and co-transfected with 60 ng pMIR-REPORT vector, 80 ng empty vector (pCMV-MIR) and 10 ng pMIR-REPORT β-gal control plasmid the next day or when cells reached 30–50% confluency, using 450 nl jetPRIME® reagent (cat. no. 101000046; Polyplus-transfection SA), according to the manufacturer's instructions. Following an initial 24-h cultivation after transfection, the medium was replaced with fresh complete medium, and culture continued for an additional 24 h before detecting luciferase activity using the Luc-Screen Extended-Glow Luciferase Reporter Gene Assay System (cat. no. T1035; Thermo Fisher Scientific, Inc.) and galactosidase with the Galacto-Light Plus β-Galactosidase Reporter Gene Assay System (cat. no. T1007; Invitrogen; Thermo Fisher Scientific, Inc.). Finally, the luciferase activity [measured in relative light units (RLUs)] was normalized to the activity of β-gal as RLU/β-gal. Transfection efficiency was calculated as the number of pCMV-MIR green fluorescent protein-positive cells as a % of the number of cells with Hoechst 33342-stained nuclei (cat. no. 910-3015; ChemoMetec A/S). The ECHO Revolve microscope (RVL-100-M; BICO Group AB) was also used to capture fluorescent images of transfected cells, and positive cells were quantified using QuPath (version 0.3.0; http://qupath.github.io) (41).

Statistical analysis

The frequencies of the G allele in the rs28382740 SNP of the XIAP 3′-UTR and CRC recurrence in the 15 patients with CRC were compared using Fisher's exact test using SPSS Statistics for Windows (version 20; IBM Corp.). Risk analysis was estimated by calculating the odds ratio (OR) and the 95% confidence interval (CI). The relative XIAP expressions of two groups (shLuc-SW480 and shXIAP-SW480) were compared using the unpaired Student's t-test, and the relative luciferase activity and cell proliferation of different groups were compared using one-way ANOVA, followed by Tukey's post hoc test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Significant SNPs in patients with recurrent CRC

For long follow-up times, archived FFPE specimens and appropriate samples were available to assess genes involved in recurrent CRC. Therefore, patients with CRC were recruited who were followed ≥5 years to confirm their recurrence status. Using WES, 27 SNPs with common variant sequences and distribution in 21 genes were revealed in the group of patients with recurrent CRC (Table SI). Due to the significant molecular functions of XIAP in cell proliferation, apoptosis, invasion and metastasis, this apoptosis-related protein was further studied for CRC progression. As presented in Table IV, the most genotype of the rs28382740 SNP in the XIAP 3′-UTR was type A in nonrecurrent cases and type G in recurrent cases. In addition, three early-stage CRC cell lines (LS123, HCT116 and LS174T) demonstrated genotype A, and two CRC cell lines with recurrence potential (SW480 cell line and its metastasis-derived SW620 cell line) were sequenced as genotype G in the rs28382740 SNP.

Table IV.

rs28382740 single nucleotide polymorphism in patients with colorectal cancer (n=15).

Table IV.

rs28382740 single nucleotide polymorphism in patients with colorectal cancer (n=15).

Prognosis

rs28382740 SNP alleleTotal patients with CRC, nNo recurrenceRecurrenceP-value
G allele52 (40.0)3 (60.0)0.077
A allele109 (90.0)1 (10.0)

[i] Values are expressed as n (%). SNP, single nucleotide polymorphism; CRC, colorectal cancer.

The expression of XIAP in the shXIAP-SW480 cells was significantly reduced in comparison with that in shLuc-SW480 cells (Fig. 1A). Furthermore, the proliferation of shXIAP-SW480 cells was notably reduced compared with that of shLuc-SW480 cells (Fig. 1B), and the relative quantitative results, which represented the level of proliferation by cell coverage, also demonstrated that shXIAP-SW480 cells had significantly lower levels of proliferation than that of shLuc-SW480 cells from day 2 (P<0.001 for Day 2 and P<0.0001 for Day 3; Fig. 1C).

XIAP levels and genotypes of patients with CRC in the tissue array

Among the 15 patients with CRC whose tissues were assembled into the tissue array, 4 were diagnosed with recurrent CRC (median time to recurrence, 102.0 months; range, 7.3–174.1 months), whilst 11 were not demonstrated to have any signs of recurrence during the follow-up period (median follow-up time, 64.4 months; range, 26.9–124.9 months). To assess the clinical relevance of XIAP in these patients with CRC, paired colon tissues (non-CRC and CRC cores) were evaluated for XIAP levels using IHC. The results demonstrated that markedly higher levels of XIAP were detected in the CRC tissue with a more aggressive phenotype, whereas the corresponding non-CRC tissue showed negative immunostaining (Fig. 2A). The XIAP levels were then semi-quantitatively scored based on the intensity and % of epithelial cells in colon tissues. Fig. 2B presents representative images of the four scores (scores 0–3). Briefly, tissues with an intensity score 32 were classified into the high XIAP-expressed group, and tissues with an intensity score <2 were considered the low XIAP-expressed group. In the paired tissues of 4 recurrent patients, 50% (2/4) of non-CRC tissues and 75% (3/4) of CRC tissues expressed high XIAP levels. In comparison, among the 11 nonrecurrent patients, 54.5% (6/11) of the CRC tissues had high XIAP levels, but only 9.1% (1/11) of non-CRC tissues had high XIAP levels. However, this difference in patients with no recurrence was not statistically significant (P=0.063; Table V). Further observation of the subcellular localization of the XIAP protein revealed markedly increased nuclear XIAP intensity in patients with CRC recurrence (Fig. 3). In addition, the genotypes of the rs28382740 SNP in the XIAP 3′-UTR of patients with CRC in the tissue array were analyzed (the A/G heterotype of the rs28382740 SNP; Fig. 4). The frequency of G-allele carriers was 14.3% (1/7) in patients with AJCC stage II and 50.0% (4/8) in patients with AJCC stage III. Among the G-allele carriers, 60% (3/5) were diagnosed with CRC recurrence, whilst only 10% (1/10) of patients without a G allele had recurrent CRC (P=0.077; Table IV). Furthermore, the G allele at the rs28382740 SNP notably increased the risk of recurrent CRC at AJCC stages II and III, with an OR of 13.5 (95% CI, 0.88–207.62). Conversely, according to the refSNP cluster ID number (e.g. rs28382740) (43), the Asian population has a markedly higher proportion of the G allele in the rs28382740 SNP (36.6–48.0%) than the European and African populations (15.7–19.2%; Table VI).

Table V.

Expression level of X-linked inhibitor of the apoptosis protein in non-recurrent (n=11) and recurrent (n=4) patients with colorectal cancer.

Table V.

Expression level of X-linked inhibitor of the apoptosis protein in non-recurrent (n=11) and recurrent (n=4) patients with colorectal cancer.

A, No recurrence

Expression level

Sample typeHighLowP-value
Non-CRC tissue1 (9.1)10 (90.9)0.063
CRC tissue6 (54.5)5 (45.5)

B, Recurrence

Expression level

Sample typeHighLowP-value

Non-CRC tissue2 (50.0)2 (50.0)1.000
CRC tissue3 (75.0)1 (25.0)

[i] Values are expressed as n (%). CRC, colorectal cancer.

Table VI.

Allele frequency of rs28382740 in different populations.

Table VI.

Allele frequency of rs28382740 in different populations.

Allele frequencya, %

PopulationSample sizeAG
European20,11480.819.2
African3,39484.315.7
African American3,28084.315.7
Latin American61081.818.2
Asian16858.341.7
East Asian11263.436.6
South Asian9852.048.0

a Frequency data were compiled from https://www.ncbi.nlm.nih.gov/snp/rs28382740.

rs28382740 SNP in miR binding sites

SNPs in miR binding sites are known to be potential cancer biomarkers with clinical significance (44). The rs28382740 SNP in the XIAP 3′-UTR was demonstrated to have putative binding sites for two different miR-24s (miR24-1-5p and miR24-2-5p) (45). Therefore, the clinical significance of this miR binding to the rs28382740 SNP in CRC was assessed. The fragment of the XIAP 3′-UTR containing any genotype of the rs28382740 SNP was cloned and inserted into separate luciferase reporters (pMIR-REPORT-A and pMIR-REPORT-G) and transfected into HCT116 cells (Fig. 5A). Following an appropriate antibiotic selection, green fluorescence was expressed in the transfected cells (Fig. S1), and the transfection efficiencies of pCMV-MIR-miR24-1 and pCMV-MIR-miR24-2 were 20.1 and 20.5%, respectively (Fig. 5B). The luciferase activity was determined following co-transfection with a different effector plasmid (pCMV-MIR-miR-24-1 or pCMV-MIR-miR24-2). Both miR-24-1-5p and miR-24-2-5p significantly downregulated the relative luciferase activity of HCT116 cells with different rs28382740 SNP genotypes compared with cells with the empty vector pCMV-MIR (Fig. 5C). Furthermore, in HCT116 cells with rs28382740 SNP genotype A, miR-24-1-5p significantly reduced the relative luciferase activity compared with miR-24-2-5p (Fig. 5C), whilst this reduction was undetectable in HCT116 cells with rs28382740 SNP genotype G (Fig. 5C).

Discussion

CRC is a common malignant disease of the gastrointestinal system, and recurrence results in poor clinical outcomes following surgery and postsurgical treatment (46). Despite established clinical strategies, including surgery, adjuvant chemotherapy and targeted therapy, the recurrence rate of CRC has not yet decreased (47). Currently, CRC recurrence is associated with multiple risk factors, such as molecular subtypes, clinical stages and epigenetic alterations (29,48,49). Targeted therapy and precision medicine for recurrent CRC may improve the clinical outcomes of these vulnerable patients. Therefore, whether the prognosis will improve when patients with CRC experience a significant reduction in cancer recurrence after surgery warrants exploration. Using postoperative specimens to assess recurrence is the most advantageous and convenient clinical strategy, and FFPE tissue, which contains most of the pathological information of a patient, is a feasible source to assess molecular and clinical follow-up data (50,51).

From archived tissue samples, the present study demonstrated that the XIAP gene with genotype G at the rs28382740 SNP was primarily detected in patients with recurrent CRC. A total of >50% of the non-CRC and CRC tissues of patients with recurrent CRC, and CRC tissues of patients with nonrecurrent CRC, expressed higher levels of XIAP. Nevertheless, only the non-CRC tissues of patients with nonrecurrent CRC expressed lower levels of XIAP. These results indicate that the genotypes and levels of XIAP may be associated with patient prognosis. Furthermore, the results of the present study were partially consistent with the conclusion of Xiang et al (39), who reported that the status of XIAP expression could be an independent prognostic marker in CRC. Moreover, it has been reported that XIAP is the strongest member of the family of inhibitors of apoptosis proteins (52). Therapeutic benefits in diseases such as cancers caused by inappropriate inhibition of cell death, may result from reinduction or triggering of apoptosis (53). Therefore, XIAP, as a potent inhibitor of cell death, may be involved in chemotherapy resistance and tumor aggressiveness in several cancers (45,52,54,55). High levels of XIAP may be a potential therapeutic target (18). For example, inhibition of XIAP has been reported to increase carboplatin sensitivity in ovarian cancer (56). In contrast, the present study demonstrated that patients with CRC and advanced-stage disease had increased XIAP expression in the nucleus. This finding was similar to that of Delbue et al (57) who revealed that an elevated expression of nuclear XIAP may be associated with drug resistance and poor prognosis in breast cancer. Taken together, the results of the present study imply that increases in XIAP expression, not only in whole cells or the nucleus, appear to be an adverse prognostic factor for clinical outcomes in many cancers (57,58). XIAP may be targeted in CRC with a poor prognosis, but its concrete role needs to be further explored.

XIAP has been known to inhibit the growth of several cancers through different pathways (22,57,59). This inhibition was also demonstrated by the results of the present study, which indicated that parental SW480 cells grew faster than XIAP-knockdown SW480 cells in CRC. The inhibition could be inferred to slow CRC cell growth by reducing XIAP expression. Due to the molecular significance of XIAP in the apoptosis and growth of cells, it has become a potential therapeutic target in tumors and inflammatory diseases (60,61).

As XIAP is critical for CRC progression and miRs are known to develop or mitigate cancer by modulating target expression, it is important to understand the interaction between XIAP and specific miRs. Prabhu et al (62) reported that changes in gene expression were caused by the interaction of miRs and SNPs, and this effect was related to the sequence of the miR-mRNA binding site within the target gene. For example, in CRC, high miR-503 and high miR-183 have been positively associated with tumor progression (63) and poor prognosis (28). Conversely, patients with CRC and high miR-23b in plasma have been reported to exhibit an improved survival rate (64). Taken together, miRs in CRC have been studied for years, but whether they are oncogenic or tumor suppressor miRs remains to be determined (65). Understanding the regulation of several miRs with clinical significance for CRC is warranted.

Taking XIAP as an example in several cancers, miR-215 and miR-122-5p have been reported to restrain XIAP levels to negatively regulate tumor growth for cancers in the gastrointestinal system (52,66). In addition, miR-214-3p and miR-618 have been reported to act as tumor suppressors against retinoblastoma (67) and degenerative thyroid cancer (68), respectively, by interacting with the XIAP 3′-UTR. The present study revealed the rs28382740 SNP at the XIAP 3′-UTR using WES, indicated it might overlap with the miR-24 binding region and speculated that XIAP was indeed one of the genes in the list that miR-24 could bind to through the algorithm of miRDB (http://www.mirdb.org/). This interaction may decrease the expression level of XIAP in CRC cells. Furthermore, other studies have reported that the interaction of miRs and their target SNPs produces intracellular effects with clinical importance (69,70). miR-24 has been widely studied in several human cancers with different roles (71). Further information on miR-24-1-5p and miR-24-2-5p was obtained from miRDB (Table SI). Among the top five high-scoring targets, there are three other CRC-related genes [Caldesmon 1 (CALD1), serine and arginine rich splicing factor 11 (SRSF11), and SRSF protein kinase 2 (SRPK2)] besides XIAP, which scored the third highest one (7275). Inhibiting oncomiR or inducing tumor suppressors through miR-based therapies may be effective in treating cancer (76). Therefore, miR-24-1-5p and miR-24-2-5p may contribute to regulating their potential targets, especially those known to be possible therapeutic targets for CRC, including XIAP, CALD1 and two genes associated with serine- and arginine-rich splicing factor (SRSF11 and SRPK2) (74,75,77,78). Taken together, further research is needed to explore changes in these genes in association with the genotype (A or G) of rs28382740 SNP in CRC cells. As reviewed by Mukherjee et al (79), miR-24, along with its passenger strands, miR-24-1-5p and miR-24-2-5p, may be involved in the pathobiology of several diseases and have potential for their diagnosis and prognosis. Zhang et al (80) highlighted that upregulated miR-24-1-5p may provide preventive and therapeutic strategies for CRC through intracellular molecular mechanisms. These findings illustrate that miR-24 has molecular functions for disease prevention and treatment.

In the present study, the molecular regulation of miR-24 on the downregulation of XIAP expression was genotype-dependent. The results demonstrating that miR-24-1-5p reduced the luciferase activity of SW480 cells with rs28382740 SNP genotype A indicate that patients with CRC and rs28382740 SNP genotype A may have lower levels of XIAP expression in the presence of a certain level of miR-24-1-5p. These results from clinical specimens and CRC cell lines demonstrate that the expression levels and genotypes of XIAP in CRC cells may be associated with CRC prognosis. However, the high proportion of the genotype G allele at rs28382740 SNP in the Asian population must have clinical significance. This implies that it is also important to determine the rs28382740 SNP genotype in patients in the Asian population with AJCC stage II and III CRC as patients with CRC and rs28382740 SNP genotype G have higher levels of XIAP expression and are not suitable for receiving miR-24 (miR-24-1-5p and miR-24-2-5p). The findings of the present study may benefit the future development of molecular diagnosis and personalized therapies for recurrent CRC.

Genes in the IAP family are frequently expressed at elevated levels in tumor maintenance and progression (13). Targeting IAP proteins could be an option for antitumor therapeutic intervention (81). That is, preventing apoptosis may inhibit tumor growth and recurrence and lead to an improved prognosis (82). In other words, XIAP, as a key molecule for cell death, exerts its oncogenic potential by inhibiting apoptosis to promote cell proliferation and could be a molecular target for anti-CRC (44). Clinical trials of several miR replacement therapies may demonstrate their potential for cancer treatment (26). However, there are limitations to the present study that should be noted. First, although the present study used 24 clinical samples, including 15 paired CRC tissue samples for validation, the number of clinical samples assessed should be expanded to approximate clinical complexity more closely. Second, the multigene panel is frequently considered to be associated with CRC progression (83,84). Further study of multiple genes related or unrelated to XIAP is necessary to understand the molecular biology of CRC recurrence. Third, the targets of miRs are not unique genes, so further research should assess the molecular effects of other genes regulated by miR-24 (miR-24-1-5p or miR-24-2-5p) on CRC cells. Further investigations should clarify the relationship between XIAP and CRC recurrence.

In conclusion, the results of the present study indicate that miR-24-1-5p and miR-24-2-5p could directly target the rs28382740 SNP in the 3′-UTR of XIAP to exert an inhibitory effect on XIAP protein expression, especially for miR-24-1-5p. Understanding the impact of the rs28382740 SNP on CRC recurrence may be beneficial to reduce cancer recurrence and enhance treatment. The findings infer that miR-24-1-5p downregulates XIAP expression in CRC cells due to the rs28382740 SNP genotype, which may inhibit CRC tumor growth and thereby prevent CRC recurrence, especially in patients with AJCC stages II and III.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

The present work was supported by grants from the Chi Mei Medical Center and Taipei Medical University (grant nos. 110CM-TMU-08 and 111CM-TMU-14, respectively).

Availability of data and materials

The original sequencing data generated in the present study may be found in the National Center for Biotechnology Information database under accession number PRJNA1063437 (https://www.ncbi.nlm.nih.gov/bioproject/PRJNA1063437). All other data generated in the present study may be requested from the corresponding author.

Authors' contributions

YFT and CCC conceived the project, oversaw the study and secured funding. YFT, SHY, CSH and CCC analyzed and interpreted the patient data. CJH and CYL performed the histological and pathological assessments. CJH, SHY and KYL collected and analyzed the data. CJH performed most of the experiments. KYL and CLL provided technical support in cloning and the research reagents. CLL obtained and interpretated cloning data, ensured accuracy of cell coverages, and carefully approved the relevant revision. YFT, CJH, CYL and CCC drafted the manuscript. YFT, CJH and CCC confirmed the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Ethics approval was obtained from the Ethical Committees of Taipei Veterans General Hospital (Taipei, Taiwan; approval no. 2017-07-030AC) and Cathay General Hospital Biobank (Taipei, Taiwan; approval no. HBKEC-20200928-1). The Institutional Review Board of Cathay General Hospital exempted the present study from obtaining informed consent for tissue procurement through the Biobank after an anonymous unlinked process (approval no. CGH-P108136). The study was performed in accordance with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

IAP

inhibitor of the apoptosis protein

XIAP

X-linked IAP

SNP

single nucleotide polymorphism

miR

microRNA

FFPE

formalin-fixed paraffin-embedded

IRB

Institutional Review Board

3′-UTR

3′-untranslated region

H&E

hematoxylin and eosin

IHC

immunohistochemistry

β-gal

β-galactosidase

OR

odds ratio

CI

confidence interval

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Balboa-Barreiro V, Pertega-Diaz S, Garcia-Rodriguez T, González-Martín C, Pardeiro-Pértega R, Yáñez-González-Dopeso L and Seoane-Pillado T: Colorectal cancer recurrence and its impact on survival after curative surgery: An analysis based on multistate models. Dig Liver Dis. 56:1229–1236. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Zare-Bandamiri M, Fararouei M, Zohourinia S, Daneshi N and Dianatinasab M: Risk factors predicting colorectal cancer recurrence following initial treatment: A 5-year cohort study. Asian Pac J Cancer Prev. 18:2465–2470. 2017.PubMed/NCBI

4 

Ogunwobi OO, Mahmood F and Akingboye A: Biomarkers in colorectal cancer: Current research and future prospects. Int J Mol Sci. 21:53112020. View Article : Google Scholar : PubMed/NCBI

5 

Guraya SY: Pattern, stage, and time of recurrent colorectal cancer after curative surgery. Clin Colorectal Cancer. 18:e223–e228. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Jia S, Zhang R, Li Z and Li J: Clinical and biological significance of circulating tumor cells, circulating tumor DNA, and exosomes as biomarkers in colorectal cancer. Oncotarget. 8:55632–55645. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Hao YX, Fu Q, Guo YY, Ye M, Zhao HX, Wang Q, Peng XM, Li QW, Wang RL and Xiao WH: Effectiveness of circulating tumor DNA for detection of KRAS gene mutations in colorectal cancer patients: A meta-analysis. Onco Targets Ther. 10:945–953. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Tiedje V, Ting S, Herold T, Synoracki S, Latteyer S, Moeller LC, Zwanziger D, Stuschke M, Fuehrer D and Schmid KW: NGS based identification of mutational hotspots for targeted therapy in anaplastic thyroid carcinoma. Oncotarget. 8:42613–42620. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Huang Q, Li S, Cheng P, Deng M, He X, Wang Z, Yang CH, Zhao XY and Huang J: High expression of anti-apoptotic protein Bcl-2 is a good prognostic factor in colorectal cancer: Result of a meta-analysis. World J Gastroenterol. 23:5018–5033. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Su P, Yang Y, Wang G, Chen X and Ju Y: Curcumin attenuates resistance to irinotecan via induction of apoptosis of cancer stem cells in chemoresistant colon cancer cells. Int J Oncol. 53:1343–1353. 2018.PubMed/NCBI

11 

Sun D, Tao W, Zhang F, Shen W, Tan J, Li L, Meng Q, Chen Y, Yang Y and Cheng H: Trifolirhizin induces autophagy-dependent apoptosis in colon cancer via AMPK/mTOR signaling. Signal Transduct Target Ther. 5:1742020. View Article : Google Scholar : PubMed/NCBI

12 

Zhou Y, Zhang X, Zhang J, Fang J, Ge Z and Li X: LRG1 promotes proliferation and inhibits apoptosis in colorectal cancer cells via RUNX1 activation. PLoS One. 12:e01751222017. View Article : Google Scholar : PubMed/NCBI

13 

Miura K, Fujibuchi W, Ishida K, Naitoh T, Ogawa H, Ando T, Yazaki N, Watanabe K, Haneda S, Shibata C and Sasaki I: Inhibitor of apoptosis protein family as diagnostic markers and therapeutic targets of colorectal cancer. Surg Today. 41:175–182. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Cetraro P, Plaza-Diaz J, MacKenzie A and Abadia-Molina F: A review of the current impact of inhibitors of apoptosis proteins and their repression in cancer. Cancers (Basel). 14:16712022. View Article : Google Scholar : PubMed/NCBI

15 

Hector S and Prehn JH: Apoptosis signaling proteins as prognostic biomarkers in colorectal cancer: A review. Biochim Biophys Acta. 1795:117–129. 2009.PubMed/NCBI

16 

Cheung CHA, Chang YC, Lin TY, Cheng SM and Leung E: Anti-apoptotic proteins in the autophagic world: An update on functions of XIAP, Survivin, and BRUCE. J Biomed Sci. 27:312020. View Article : Google Scholar : PubMed/NCBI

17 

Cao C, Mu Y, Hallahan DE and Lu B: XIAP and survivin as therapeutic targets for radiation sensitization in preclinical models of lung cancer. Oncogene. 23:7047–7052. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Devi Daimary U, Girisa S, Parama D, Verma E, Kumar A and Kunnumakkara AB: Embelin: A novel XIAP inhibitor for the prevention and treatment of chronic diseases. J Biochem Mol Toxicol. 36:e229502022. View Article : Google Scholar : PubMed/NCBI

19 

Devi GR: XIAP as target for therapeutic apoptosis in prostate cancer. Drug News Perspect. 17:127–134. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Tong QS, Zheng LD, Wang L, Zeng FQ, Chen FM, Dong JH and Lu GC: Downregulation of XIAP expression induces apoptosis and enhances chemotherapeutic sensitivity in human gastric cancer cells. Cancer Gene Ther. 12:509–514. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Huang Q and Liu F: Ceramide analogue 5cc overcomes TRAIL-resistance by enhancing JNK activation and repressing XIAP expression in metastatic colon cancer cells. Chemotherapy. 68:210–218. 2023. View Article : Google Scholar : PubMed/NCBI

22 

Seo HG, Kim HB, Yoon JY, Kweon TH, Park YS, Kang J, Jung J, Son S, Yi EC, Lee TH, et al: Mutual regulation between OGT and XIAP to control colon cancer cell growth and invasion. Cell Death Dis. 11:8152020. View Article : Google Scholar : PubMed/NCBI

23 

LaCasse EC: Pulling the plug on a cancer cell by eliminating XIAP with AEG35156. Cancer Lett. 332:215–224. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Oost TK, Sun C, Armstrong RC, Al-Assaad AS, Betz SF, Deckwerth TL, Ding H, Elmore SW, Meadows RP, Olejniczak ET, et al: Discovery of potent antagonists of the antiapoptotic protein XIAP for the treatment of cancer. J Med Chem. 47:4417–4426. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Opo F, Rahman MM, Ahammad F, Ahmed I, Bhuiyan MA and Asiri AM: Structure based pharmacophore modeling, virtual screening, molecular docking and ADMET approaches for identification of natural anti-cancer agents targeting XIAP protein. Sci Rep. 11:40492021. View Article : Google Scholar : PubMed/NCBI

26 

Hosseinahli N, Aghapour M, Duijf PHG and Baradaran B: Treating cancer with microRNA replacement therapy: A literature review. J Cell Physiol. 233:5574–5588. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Alves Dos Santos K, Clemente Dos Santos IC, Santos Silva C, Gomes Ribeiro H, de Farias Domingos I and Nogueira Silbiger V: Circulating exosomal mirnas as biomarkers for the diagnosis and prognosis of colorectal cancer. Int J Mol Sci. 22:3462020. View Article : Google Scholar : PubMed/NCBI

28 

Yuan D, Li K, Zhu K, Yan R and Dang C: Plasma miR-183 predicts recurrence and prognosis in patients with colorectal cancer. Cancer Biol Ther. 16:268–275. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Moridikia A, Mirzaei H, Sahebkar A and Salimian J: MicroRNAs: Potential candidates for diagnosis and treatment of colorectal cancer. J Cell Physiol. 233:901–913. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Wang V and Wu W: MicroRNA-based therapeutics for cancer. BioDrugs. 23:15–23. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Mollaei H, Safaralizadeh R and Rostami Z: MicroRNA replacement therapy in cancer. J Cell Physiol. 234:12369–12384. 2019. View Article : Google Scholar : PubMed/NCBI

32 

He X, Cheng G, Xiao F, Zhang L, Jin G, Zhao X, Liu Y, Liang J, Li Y, Liu Z, et al: miR-4477b gene as a novel pathogenic mutation occurring during the transformation of colorectal adenoma into colorectal cancer. J Gastrointest Oncol. 12:69–78. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Li H and Durbin R: Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 26:589–595. 2010. View Article : Google Scholar : PubMed/NCBI

34 

McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M and DePristo MA: The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20:1297–1303. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Razali NN, Raja Ali RA, Muhammad Nawawi KN, Yahaya A and Mokhtar NM: Targeted sequencing of Cytokine-Induced PI3K-Related genes in ulcerative colitis, colorectal cancer and Colitis-Associated cancer. Int J Mol Sci. 23:114722022. View Article : Google Scholar : PubMed/NCBI

36 

Degalez F, Jehl F, Muret K, Bernard M, Lecerf F, Lagoutte L, Désert C, Pitel F, Klopp C and Lagarrigue S: Watch Out for a Second SNP: Focus on Multi-Nucleotide variants in coding regions and rescued Stop-Gained. Front Genet. 12:6592872021. View Article : Google Scholar : PubMed/NCBI

37 

McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GR, Thormann A, Flicek P and Cunningham F: The ensembl variant effect predictor. Genome Biol. 17:1222016. View Article : Google Scholar : PubMed/NCBI

38 

Maran MIJ and Davis GD: Benefits of merging paired-end reads before pre-processing environmental metagenomics data. Mar Genomics. 61:1009142022. View Article : Google Scholar : PubMed/NCBI

39 

Xiang G, Wen X, Wang H, Chen K and Liu H: Expression of X-linked inhibitor of apoptosis protein in human colorectal cancer and its correlation with prognosis. J Surg Oncol. 100:708–712. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Lin ZH, Lu MK, Lo HC, Chang CC, Tseng AJ, Chao CH and Lin TY: ZnF3, a sulfated polysaccharide from Antrodia cinnamomea, inhibits lung cancer cells via induction of apoptosis and activation of M1-like macrophage-induced cell death. Int J Biol Macromol. 238:1241442023. View Article : Google Scholar : PubMed/NCBI

41 

Huang CJ, Pu CM, Su SY, Lo SL, Lee CH and Yen YH: Improvement of wound healing by capsaicin through suppression of the inflammatory response and amelioration of the repair process. Mol Med Rep. 28:1552023. View Article : Google Scholar : PubMed/NCBI

42 

Huang CJ, Yang SH, Lee CL, Cheng YC, Tai SY and Chien CC: Ribosomal protein S27-like in colorectal cancer: A candidate for predicting prognoses. PLoS One. 8:e670432013. View Article : Google Scholar : PubMed/NCBI

43 

Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, Smigielski EM and Sirotkin K: dbSNP: The NCBI database of genetic variation. Nucleic Acids Res. 29:308–311. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Hanifeh M and Ataei F: XIAP as a multifaceted molecule in cellular signaling. Apoptosis. 27:441–453. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Jiang R, Su G, Chen X, Chen S, Li Q, Xie B and Zhao Y: Esculetin inhibits endometrial cancer proliferation and promotes apoptosis via hnRNPA1 to downregulate BCLXL and XIAP. Cancer Lett. 521:308–321. 2021. View Article : Google Scholar : PubMed/NCBI

46 

Liu F, Dai Z, Cheng Q, Xu L, Huang L, Liu Z, Li X, Wang N, Wang G, Wang L and Wang Z: LncRNA-targeting bio-scaffold mediates triple immune effects for postoperative colorectal cancer immunotherapy. Biomaterials. 284:1214852022. View Article : Google Scholar : PubMed/NCBI

47 

Hossain MS, Karuniawati H, Jairoun AA, Urbi Z, Ooi J, John A, Lim YC, Kibria KMK, Mohiuddin AKM, Ming LC, et al: Colorectal cancer: A review of carcinogenesis, global epidemiology, current challenges, risk factors, preventive and treatment strategies. Cancers (Basel). 14:17322022. View Article : Google Scholar : PubMed/NCBI

48 

Ganz A, Gandhi SR, Schafer J, Singh T, Puleo E, Mullett G and Wilson C: PERCEPT: indoor navigation for the blind and visually impaired. Annu Int Conf IEEE Eng Med Biol Soc. 2011:856–859. 2011.PubMed/NCBI

49 

Wolpin BM, Wei EK, Ng K, Meyerhardt JA, Chan JA, Selhub J, Giovannucci EL and Fuchs CS: Prediagnostic plasma folate and the risk of death in patients with colorectal cancer. J Clin Oncol. 26:3222–3228. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Lygirou V, Fasoulakis K, Stroggilos R, Makridakis M, Latosinska A, Frantzi M, Katafigiotis I, Alamanis C, Stravodimos KG, Constantinides CA, et al: Proteomic analysis of prostate cancer FFPE samples reveals markers of disease progression and aggressiveness. Cancers (Basel). 14:37652022. View Article : Google Scholar : PubMed/NCBI

51 

Walter RF, Mairinger FD, Wohlschlaeger J, Worm K, Ting S, Vollbrecht C, Schmid KW and Hager T: FFPE tissue as a feasible source for gene expression analysis-a comparison of three reference genes and one tumor marker. Pathol Res Pract. 209:784–789. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Lu C, Hong M, Chen B, Liu K, Lv Y, Zhou X and Su G: MicroRNA-215 regulates the apoptosis of HCT116 colon cancer cells by Inhibiting X-Linked inhibitor of apoptosis protein. Cancer Biother Radiopharm. 36:728–736. 2021.PubMed/NCBI

53 

Holcik M, Gibson H and Korneluk RG: XIAP: apoptotic brake and promising therapeutic target. Apoptosis. 6:253–261. 2001. View Article : Google Scholar : PubMed/NCBI

54 

Devi GR, Finetti P, Morse MA, Lee S, de Nonneville A, Van Laere S, Troy J, Geradts J, McCall S and Bertucci F: Expression of X-Linked inhibitor of apoptosis protein (XIAP) in breast cancer is associated with shorter survival and resistance to chemotherapy. Cancers (Basel). 13:28072021. View Article : Google Scholar : PubMed/NCBI

55 

Saha G, Sarkar S, Mohanta PS, Kumar K, Chakrabarti S, Basu M and Ghosh MK: USP7 targets XIAP for cancer progression: Establishment of a p53-independent therapeutic avenue for glioma. Oncogene. 41:5061–5075. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Zhang Y, Huang F, Luo Q, Wu X, Liu Z, Chen H and Huang Y: Inhibition of XIAP increases carboplatin sensitivity in ovarian cancer. Onco Targets Ther. 11:8751–8759. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Delbue D, Mendonca BS, Robaina MC, Lemos LGT, Lucena PI, Viola JPB, Magalhães LM, Crocamo S, Oliveira CAB, Teixeira FR, et al: Expression of nuclear XIAP associates with cell growth and drug resistance and confers poor prognosis in breast cancer. Biochim Biophys Acta Mol Cell Res. 1867:1187612020. View Article : Google Scholar : PubMed/NCBI

58 

Gao X, Zhang L, Wei Y, Yang Y, Li J, Wu H and Yin Y: Prognostic value of XIAP level in patients with various cancers: A systematic review and Meta-Analysis. J Cancer. 10:1528–1537. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Liu XG, Xu J, Li F, Li MJ and Hu T: Down-regulation of miR-377 contributes to cisplatin resistance by targeting XIAP in osteosarcoma. Eur Rev Med Pharmacol Sci. 22:1249–1257. 2018.PubMed/NCBI

60 

Jost PJ and Vucic D: Regulation of cell death and immunity by XIAP. Cold Spring Harb Perspect Biol. 12:a0364262020. View Article : Google Scholar : PubMed/NCBI

61 

Vucic D: XIAP at the crossroads of cell death and inflammation. Oncotarget. 9:27319–27320. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Prabhu NB, Vinay CM, Satyamoorthy K and Rai PS: Pharmacogenomics deliberations of 2-deoxy-d-glucose in the treatment of COVID-19 disease: An in silico approach 3. Biotech. 12:2872022.

63 

Noguchi T, Toiyama Y, Kitajima T, Imaoka H, Hiro J, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Toden S and Kusunoki M: miRNA-503 promotes tumor progression and is associated with early recurrence and poor prognosis in human colorectal cancer. Oncology. 90:221–231. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Kou CH, Zhou T, Han XL, Zhuang HJ and Qian HX: Downregulation of mir-23b in plasma is associated with poor prognosis in patients with colorectal cancer. Oncol Lett. 12:4838–4844. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Bonfrate L, Altomare DF, Di Lena M, Travaglio E, Rotelli MT, De Luca A and Portincasa P: MicroRNA in colorectal cancer: New perspectives for diagnosis, prognosis and treatment. J Gastrointestin Liver Dis. 22:311–320. 2013.PubMed/NCBI

66 

Yin Z, Zhou Y, Ma T, Chen S, Shi N, Zou Y, Hou B and Zhang C: Down-regulated lncRNA SBF2-AS1 in M2 macrophage-derived exosomes elevates miR-122-5p to restrict XIAP, thereby limiting pancreatic cancer development. J Cell Mol Med. 24:5028–5038. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Yang L, Zhang L, Lu L and Wang Y: miR-214-3p regulates Multi-Drug resistance and apoptosis in retinoblastoma cells by targeting ABCB1 and XIAP. Onco Targets Ther. 13:803–811. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Cheng Q, Zhang X, Xu X and Lu X: MiR-618 inhibits anaplastic thyroid cancer by repressing XIAP in one ATC cell line. Ann Endocrinol (Paris). 75:187–193. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Chhichholiya Y, Suryan AK, Suman P, Munshi A and Singh S: SNPs in miRNAs and target sequences: Role in cancer and diabetes. Front Genet. 12:7935232021. View Article : Google Scholar : PubMed/NCBI

70 

Preskill C and Weidhaas JB: SNPs in microRNA binding sites as prognostic and predictive cancer biomarkers. Crit Rev Oncog. 18:327–340. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Wang S, Liu N, Tang Q, Sheng H, Long S and Wu W: MicroRNA-24 in cancer: A double side medal with opposite properties. Front Oncol. 10:5537142020. View Article : Google Scholar : PubMed/NCBI

72 

Kohler C: Histochemical localization of caldesmon isoforms in colon adenocarcinoma and lymph node metastases. Virchows Arch. 459:81–89. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Oh J, Oh JM and Cho SY: METTL3-mediated downregulation of splicing factor SRSF11 is associated with carcinogenesis and poor survival of cancer patients. Eur Rev Med Pharmacol Sci. 27:2561–2570. 2023.PubMed/NCBI

74 

Pan YJ, Huo FC, Kang MJ, Liu BW, Wu MD and Pei DS: Alternative splicing of HSPA12A pre-RNA by SRSF11 contributes to metastasis potential of colorectal cancer. Clin Transl Med. 12:e11132022. View Article : Google Scholar : PubMed/NCBI

75 

Wang J, Wu HF, Shen W, Xu DY, Ruan TY, Tao GQ and Lu PH: SRPK2 promotes the growth and migration of the colon cancer cells. Gene. 586:41–47. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Menon A, Abd-Aziz N, Khalid K, Poh CL and Naidu R: miRNA: A promising therapeutic target in cancer. Int J Mol Sci. 23:115022022. View Article : Google Scholar : PubMed/NCBI

77 

Alnuaimi AR, Nair VA, Malhab LJB, Abu-Gharbieh E, Ranade AV, Pintus G, Hamad M, Busch H, Kirfel J, Hamoudi R and Abdel-Rahman WM: Emerging role of caldesmon in cancer: A potential biomarker for colorectal cancer and other cancers. World J Gastrointest Oncol. 14:1637–1653. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Qiao L, Dai Y, Gu Q, Chan KW, Zou B, Ma J, Wang J, Lan HY and Wong BC: Down-regulation of X-linked inhibitor of apoptosis synergistically enhanced peroxisome proliferator-activated receptor gamma ligand-induced growth inhibition in colon cancer. Mol Cancer Ther. 7:2203–2211. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Mukherjee S, Shelar B and Krishna S: Versatile role of miR-24/24-1*/24-2* expression in cancer and other human diseases. Am J Transl Res. 14:20–54. 2022.PubMed/NCBI

80 

Zhang H, Guo J, Mao L, Li Q, Guo M, Mu T, Zhang Q and Bi X: Up-regulation of miR-24-1-5p is involved in the chemoprevention of colorectal cancer by black raspberry anthocyanins. Br J Nutr. 122:518–526. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Vucic D and Fairbrother WJ: The inhibitor of apoptosis proteins as therapeutic targets in cancer. Clin Cancer Res. 13:5995–6000. 2007. View Article : Google Scholar : PubMed/NCBI

82 

Nagaraju GP, Bramhachari PV, Raghu G and El-Rayes BF: Hypoxia inducible factor-1α: Its role in colorectal carcinogenesis and metastasis. Cancer Lett. 366:11–18. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Belardinilli F, Capalbo C, Malapelle U, Pisapia P, Raimondo D, Milanetti E, Yasaman M, Liccardi C, Paci P, Sibilio P, et al: Clinical multigene panel sequencing identifies distinct mutational association patterns in metastatic colorectal cancer. Front Oncol. 10:5602020. View Article : Google Scholar : PubMed/NCBI

84 

Li HD and Liang C: Multigene panel predicting survival of patients with colon cancer. J Cancer. 10:6792–6800. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 28 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian Y, Huang C, Liu C, Yang S, Hung C, Lin K, Lai C and Chang C: MicroRNA‑24 alleviates colorectal cancer progression via a rs28382740 single nucleotide polymorphism in the long noncoding region of X‑linked inhibitor of apoptosis protein. Oncol Lett 28: 591, 2024.
APA
Tian, Y., Huang, C., Liu, C., Yang, S., Hung, C., Lin, K. ... Chang, C. (2024). MicroRNA‑24 alleviates colorectal cancer progression via a rs28382740 single nucleotide polymorphism in the long noncoding region of X‑linked inhibitor of apoptosis protein. Oncology Letters, 28, 591. https://doi.org/10.3892/ol.2024.14724
MLA
Tian, Y., Huang, C., Liu, C., Yang, S., Hung, C., Lin, K., Lai, C., Chang, C."MicroRNA‑24 alleviates colorectal cancer progression via a rs28382740 single nucleotide polymorphism in the long noncoding region of X‑linked inhibitor of apoptosis protein". Oncology Letters 28.6 (2024): 591.
Chicago
Tian, Y., Huang, C., Liu, C., Yang, S., Hung, C., Lin, K., Lai, C., Chang, C."MicroRNA‑24 alleviates colorectal cancer progression via a rs28382740 single nucleotide polymorphism in the long noncoding region of X‑linked inhibitor of apoptosis protein". Oncology Letters 28, no. 6 (2024): 591. https://doi.org/10.3892/ol.2024.14724