Open Access

Deletion in a regulatory region is associated with underexpression of miR-148b‑3p in patients with prostate cancer

  • Authors:
    • Fernando Bergez‑Hernández
    • Fred Luque‑Ortega
    • Noemí García‑Magallanes
    • Marco Alvarez‑Arrazola
    • Eliakym Arámbula‑Meraz
  • View Affiliations

  • Published online on: January 30, 2024     https://doi.org/10.3892/br.2024.1740
  • Article Number: 52
  • Copyright: © Bergez‑Hernández 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

Prostate cancer (PCa) is the leading cause of cancer‑related death in men. This pathology is complex and heterogeneous; therefore, elucidating the molecular mechanisms that lead to its origin and progression is imperative. MicroRNAs (miRNAs or miRs) are part of the epigenetic machinery that regulates the expression of human genes, therefore, mutations in the genes that encode them can lead to a dysregulation in their expression, which directly impacts their target genes, which could be oncogenes or tumor suppressor genes. In PCa several dysregulated expression levels of miRNAs are associated with perturbed cellular processes. A differential expression of miRNAs such as miR-145‑5p and miR-148‑3p has been observed in PCa, possibly due to mutations in regions near the miRNAs. However, the molecular mechanisms that lead to the dysregulation of these miRNAs still need to be clarified. Therefore, the present study aimed to analyze the expression of miRNAs and their relationship with mutations in patients with and without PCa. In total, 71 patients were analyzed: 41 of whom had PCa (CAP group) and 30 with benign pathology (BPD group). Underexpression was observed in miR-145‑5p and miR-148b‑3p in PCa patients (P=0.03 and P=0.001, respectively). In miR-145‑5p, no mutations related to its expression were identified. For miR-148b‑3p, a set of mutations were identified in the chr12:54337042/54337043 region, which were grouped into the mutation named DelsAAG. Although this mutation's abnormal allele is related to PCa (P=0.017), a statistically significant difference was observed in the expression of miR-148b‑3p between carriers and non‑carriers of the mutated allele, identifying a mechanism likely to be involved in the miR-148b‑3p dysregulation.

Introduction

Several studies have suggested that the microRNAs (miRNAs or miRs) miR-145-5p and miR-148b-3p are linked to the development of prostate cancer (PCa) as they have been observed to be dysregulated during this disease, and their expression levels differ from patients with benign prostate diseases (1-4). Furthermore, mutations in the genes that code for these miRNAs and in nearby regions have been reported, suggesting a possible explanation for their dysregulation. miR-145-5p has been classified as a tumor suppressor, specifically in PCa; this miRNA has been linked to various mechanisms that promote the development of this pathology (5,6). Some factors have been associated with the underexpression of this miRNA; for example, single nucleotide polymorphisms (SNPs) have been suggested as possible causes of miRNA aberrant expression. In PCa, the rs4705342 polymorphism stands out; in the Asian population, it has been observed that men with the TC/CC genotypes have a significantly lower risk of having PCa than those with the TT genotype. Functionality analyses demonstrated that the T allele increases protein-binding affinity and thus reduces promoter activity, resulting in decreased transcription of the miR-145-5p (7). In recent years, this polymorphism has been studied in the European population. It has been observed that, unlike the Asian population, the C allele appears to be a risk factor in Serbian men (8). These findings highlight the importance of analyzing mutations to identify their involvement in regulating this miRNA and PCa susceptibility.

miR-148b-3p has been observed to be overexpressed in PCa, both in early and metastatic stages (9). Regarding the regulation of this miRNA through single nucleotide polymorphisms, Chen et al (10) observed in 2014 that the rs11170877 (A>G) polymorphism, specifically the G allele and the rs12231393 (T>C) allele C, were associated as a protective factor for gastric cancer in Chinese patients. The rs11170877 variant is located in the exon of COPZ1, the gene that contains or hosts miR-148b. These two polymorphisms form a haplotype due to linkage disequilibrium; this haplotype showed a significant association with this cancer (10). Despite the interesting results obtained with the rs11170877 polymorphism in this type of cancer, few studies have linked this SNP to other types of cancer or even to different populations. Therefore, the present study aimed to identify mutations in miR-145-5p and miR-148b-3p and regions near them and relate them to their expression in patients with and without PCa.

Materials and methods

Patient samples

Non-probabilistic convenience sampling was conducted on prostatic tissue samples collected via transrectal ultrasound-guided 18G core needle biopsy. The biopsies were conducted by a single Board-Certified Diagnostic Radiologist with 20 years of experience in the procedure, utilizing the standard technique, including the sampling of ultrasound-suspicious areas (nodules). The samples were obtained from the tumor tissue of patients with a suspicious diagnosis of PCa based on either positive digital rectal examination or rising prostate specific antigen (PSA). The collected samples were stored in Qiazol® (Qiagen GmbH) at 20˚C for later analysis.

The diagnosis and classification of patients with benign prostatic hyperplasia (BPH) and PCa were performed by a single Board-Certified Pathologist with 7 years of experience using histopathology. The inclusion criteria were as follows: Mexican, ≥18 years, histopathological diagnosis of PCa or BPH. The exclusion criteria included patients with other types of cancer and those who had received chemotherapy or radiotherapy for any other medical reason.

The present study consisted of 71 samples: 41 from patients with PCa diagnosis (PCa group) and 30 from patients diagnosed with benign prostatic disease (BPD group). The BPD group included patients with BPH (n=7), prostatitis (n=18), both hyperplasia and prostatitis (n=4), and intraepithelial neoplasia (n=1). All patients were recruited from the Alvarez & Arrazola Radiologists Clinic in Sinaloa, Mexico, from August 2016 to December 2021. Clinicopathological characteristics including age (applicable to individuals aged ≥18 years, with an upper limit of 90 years), weight, height, body mass index, PSA and family history of cancer were collected through direct questionnaires and the clinical database. An expert pathologist provided the diagnosis and Gleason score. All patients signed informed consent forms, which were reviewed and approved by the Alvarez and Arrazola Radiologists Clinic's Ethics and Research Committee (study approval number P-3103).

RNA and DNA extraction

Tissue samples were used for RNA extraction. The prostate tissue in Qiazol was macerated using sterile pistils, until leaving a homogeneous solution. Total RNA, including miRNAs, was isolated using the miRNEasy kit (Qiagen) according to the manufacturer's protocol. DNA extraction was performed using organic solvents and salt precipitation from the remnants of Qizol® obtained from RNA extraction using the previously described methodology of Chomczynski (1993) (11). The concentration of isolated RNA and DNA was measured with a GENESYS 10S UV-Vis Spectrophotometer (Thermo Fisher Scientific, Inc.) at 260 nm. It also allows for the evaluation of RNA and DNA integrity, detection of contaminants, the analysis of low or atypical concentrations, and warning of invalid results. The concentration and integrity analysis were performed in triplicate.

Analysis of polymorphisms in miRNAs

To design primers for miRNA amplification and sequencing, the accession numbers NC_00005 and NC_000012 (https://www.ncbi.nlm.nih.gov/nuccore/NC_000005.10 and https://www.ncbi.nlm.nih.gov/nuccore/NC_000012, respectively) were used for miR-145 and miR-148b, respectively. CLC Sequence Viewer program (https://resources.qiagenbioinformatics.com/manuals/clcsequenceviewer/current/index.php?manual=Introduction_CLC_Sequence_Viewer.html) was used to select a region of ~1,000 base pairs spanning the gene sequence of each selected miRNA to cover the gene and possible regulatory sequences.

Primer-BLAST Tool from NCBI (https://www.ncbi.nlm.nih.gov/tools/primer-blast/) was employed to design primers used the sequence selected, considering the following characteristics: 40-60% G + C, non-self-complementary and no more than 3˚C (melting temperature difference) between both primers. The primers selected for miR-148b-3p were forward 5'-CAGGCTTTAGAAGCCCCTGA-3' and reverse, 5'-GCGCTTAAATGCCGCTTCA-3' with a 986 bp amplicon. And the primers used for miR-145-5p were forward, 5'-TCTCCAGTAGGTCGTGGACT-3' and reverse, 5'-CACAAGAGGGCGTTCTGAGT-3', with a 1,000 bp amplicon. Sealed 10 µl aliquots of DNA were subsequently sent to the Korean company MACROGEN for capillary electrophoresis sequencing. Once the data was obtained, Chromas V2.6.6 (https://technelysium.com.au/wp/chromas/) was used to clean the sequence, eliminating the ends in which the sequence was not reliable using the default parameters. The reference sequence GRCh38.p14 was obtained using the CLC sequence viewer 8.0 program. Subsequently, the SnackVar v2.4.3(12) and NovoSNP v.3.0.1(13) tools were used to identify mutations. Both programs are automatically using the default parameters detect SNVs and indel type mutations by comparing them with a reference sequence. Once the mutations were identified, only those that coincided in both programs and those that had a quality score >40 were included in this analysis.

Reverse transcription (RT) PCR and relative miRNA expression

RT was performed from 10 ng of total RNA with the TaqMan Advanced miRNA cDNA Synthesis kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) in a T100 Thermal Cycler (Bio-Rad Laboratories, Inc.). To obtain cDNA, the TaqMan Advanced miRNA cDNA Synthesis kit was used, which consists of four steps as follows: i) Polyadenylation reaction: First, a reaction mix was prepared where, for a final volume of 5 µl, 0.5 µl of 10x Poly(A) buffer, 0.5 µl of ATP, 0.3 µl of Poly A enzyme and 2 µl of miRNAs were added. Vortexing was briefly performed. The T100 Thermal Cycler (Bio-Rad Laboratories, Inc.) was programmed as follows: 45 min at 37˚C and 10 min at 65˚C. The next reaction was immediately carried out. ii) Ligation Reaction: The following reaction mixture was prepared for a final volume of 15 µl: 3 µl of 5X DNA ligase buffer, 4.5 µl of 50% PEG 8000, 0.6 µl of 25X ligation adapter, 1.5 µl RNA ligase and 0.4 µl of RNase-free water; and then, the 5 µl reaction mixture obtained in the polyadenylation was added and vortexed briefly. It was subsequently incubated at 16˚C for 60 min in the same thermal cycler. The reverse transcription reaction was carried out immediately afterwards. iii) Reverse transcription (RT) reaction: The following reaction mixture was prepared for a final volume of 30 µl: 6 µl of 5X RT buffer, 1.2 µl of dNTP mix (25 mM), 1.5 µl of 20X Universal RT primer, 3 µl of 10X RT enzyme mix and 3.3 µl of RNase-free water. It was mixed by pipetting, then this reaction mixture was added to the 15 µl of reaction obtained from the ligation reaction and vortexed briefly. The thermal cycler was programmed as follows: 15 min at 42˚C and 5 min at 85˚C. For each of the miRNAs, standard curves of five points of serial dilutions 1:10 were made to obtain the efficiencies from the slope. Quantitative estimations for miR-145-5p (assay ID: 477916_mir), miR-148b-3p (assay ID: 477824_mir), and miR-191-5p (assay ID: 477952_mir) were performed using TaqMan Advance miRNA Assays by Thermo Fisher Scientific, Inc. using a StepOnePlusÔ thermal cycler (Applied Biosystems). To select the normalizing miRNA, the RefFinder tool (https://blooge.cn/RefFinder/) was used, which integrates different algorithms to select normalizers, identifying miR-191-5p as the most appropriate miRNA for the present study.

Relative expression analysis

To obtain relative miRNA expression of miR-145-5p and miR-148b-3p, the previously described method by Taylor et al (2019) (14) was used based on the ∆∆Cq method (2-∆∆Cq algorithm) (15) and the miR-191-5p was used as reference gene. First, the average of the Cq of the control samples was calculated; subsequently, the average Cq obtained was subtracted individually for each test sample, obtaining a ΔCq, and then, the RQ was calculated using the formula 2∆ΔCq. Subsequently, the normalized expression was obtained for each sample using the formula RQ problem gene/RQ normalizing gene; and finally, the normalization of the expression per sample was carried out using the Log2 of each result obtained in the previous formula.

Statistical analysis

Once the information was collected, the data was organized into tables and figures. Measures of central tendency (mean, median, mode), measures of dispersion (standard deviation), and confidence intervals were estimated for the quantitative variables. For the qualitative variables, the frequencies and percentages were estimated, as well as their respective confidence intervals. To identify differences between means of the different quantitative variables in patients with and without PCa, the unpaired Student's t-test and Mann-Whitney U statistics were performed for parametric and non-parametric variables, respectively. To find a relationship between qualitative variables, the Chi-square test was used. For the correlation analyses, the Pearson's correlation coefficient and the Spearman's test were used, for parametric and non-parametric variables, respectively. All the analyses were carried out using the statistical package SPSS v.20 (IBM Corp.). To determine if the distribution of the genotypes of each mutation was in Hardy-Weinberg (EHW) equilibrium, the SNPStats software (http://www.snpstats.net/start.htm) was used. Similarly, the haplotype frequencies and linkage analysis were calculated using the same software. P<0.05 was considered to indicate a statistically significant difference.

Results

miRNAs expression analysis

Relative expression of miR-145-5p and miR-148b-3p was quantified in all samples of BPD and PCa patients (Fig. 1). When analyzing miR-145-5p expression, an expression factor of 0.71±0.15 was identified in PCa patients compared with BPD patients, showing a significant difference (P=0.033), and miR-148b-3p was underexpressed in PCa patients with an expression factor of 0.44±0.28 with a statistically significant difference between groups (P=0.001).

Identification of mutations

A total of 18 mutations were identified in the analyzed sequence for miR-148b-3p in 71 study samples (Table I). Different types of mutations were observed, which were single nucleotide variations (SNVs), deletions (Del), an insertion (Ins), and a duplication (Dup). Among the mutations obtained, 12 had already been registered, while the remaining six had not. In position chr12:54337042/54337043, there were four different deletions, two of which were previously registered (rs1465330647 and rs1187801243), and two which have not been reported in any database (Fig. 2). The rs1465330647 mutation is the deletion of AAAGAA, the rs1187801243 mutation is the loss of AAGAA, and the other two unrecorded mutations comprise the deletion of AAAGA in one case and AAGAAG in the other. As they are very similar to each other and have a low frequency separately, they were grouped into a single variable called DelsAAG. To analyze the relationship of these mutations with the expression of miR-148b-3p, those with the highest frequency of the mutated allele were selected: DupCT (rs59761210), SNV G/T (rs1381668656), SNV A/C (NA), DelG (rs368666376), SNV G/C (rs56818309) and DelsAAG (Table I). Mutation analysis for miR-145-5p was only performed on 44 of the 71 study samples since the remaining samples could not be sequenced. Of the 44 samples, 21 were from the PCa and 23 from the BPD groups. Mutation analysis for miR-145-5p was performed, where 62 mutations were identified, of which 24 had previously been reported. Of the 62 mutations, 59 were single nucleotide variations (SNV), two deletions (Del), and one duplication (Dup). Most of the mutations had a low frequency of the mutated allele; for this reason, only the rs353291 mutation, consisting of a T/C SNV with a frequency of 0.18 of the mutated alleles in the total population, was chosen.

Table I

Mutations identified for miR-148b-3p.

Table I

Mutations identified for miR-148b-3p.

MutationPositionIDMutated allele frequency
SNV G/T54336876NA0.01
SNV G/A54337007NA0.01
DupCT54337015rs597612100.15a
SNV C/G54337017rs5553061720.01
SNV G/T54337018rs13816686560.11a
SNV A/C54337021NA0.10a
SNV A/C54337042rs19538830520.02
SNV A/C54337043rs9184053350.01
DelAAAGA54337042NA0.01
DelAAAGAA54337042rs14653306470.08
DelAAGAA54337043rs11878012430.02
DelAAGAAG54337043NA0.09
DelG54337045rs3686663760.12a
InsA54337045NA0.01
SNV G/A54337045rs8791463270.01
SNV A/C54337046rs10488734420.01
SNV A/C54337047rs14810500820.02
SNV G/C54336970rs568183090.27a
DelsAAGb 54337042/54337043NA0.21a

[i] aMutations selected by the frequency of the mutated allele;

[ii] bClustering of deletions (DelAAAGA, DelAAAAGAA, DelAAGAA, DelAAGAAG) (n=71). MicroRNA, miR; SNV, single nucleotide variations; dup, duplication; Del, deletion; Ins, insertion.

Analysis by inheritance model

Subsequently, an analysis of the possible inheritance models of these six mutations of miR-148-3p, was carried out according to the response variable that groups patients in PCa and BPD using the SNPStat.net software. Of the six mutations, only DelsAAG was significant for the codominant (P=0.03, AIC=95.4), dominant (P=0.01, AIC=94.7), and over dominant (P=0.02, AIC=94.9) models. To conduct subsequent analyses using genotypes and alleles, the inheritance model was selected according to the lowest value of the Akaike Information Criterion (AIC) obtained. For the rs59761210 mutation, the dominant model was selected; for the rs368666376 mutation, the recessive model, for the rs56818309 mutation, the over dominant model; and for the DelsAAG mutation, the dominant model. In the case of the rs1381668656 and SNV A/C mutations, the analysis by different inheritance models was not obtained because the mutated homozygous genotype was not observed in the study groups; hence the codominant inheritance model was selected. In the case of miR-145-5p for the rs353291 mutation, none of the inheritance models were statistically significant; therefore, the model with the lowest AIC value was chosen (over dominant model, AIC=62.2).

Genotypic and allele frequencies and their association with PCa

The genotype and allele frequencies were calculated for each of the selected mutations, and both genotypes and alleles were associated with PCa. The allele and genotypic frequencies of the rs59761210, rs1381668656, rs368666376, rs56818309 and SNV A/C mutations are in Hardy-Weinberg equilibrium (P=0.052, P=0.59, P=0.98, P=0.36 and P=0.44, respectively); in the case of the DelsAAG mutation, this was not observed in Hardy-Weinberg equilibrium (P=0.001). The DelsAAG mutation was significantly related to the presence of PCa (P=0.014), wherein a dominant model, both mutated homozygotes and heterozygotes, were related to the pathology, unlike normal homozygotes. Likewise, the analysis by alleles revealed that patients who carry the DelsAAG mutation are 4.16 (P=0.017) times more likely to develop PCa; the other mutations were not observed to be related to the pathology (Table II). The rs353291 mutation demonstrated a frequency of the heterozygous genotype of 33.3% in patients with PCa, and 13% in the BPD group. The genotype analysis found no statistically significant relationship between the genotypes and the presence of PCa. The mutated allele presents a frequency of 0.21 in the PCa group and 0.15 in the BPD group; no significant relationship between the mutated allele and the pathology was observed (P=0.11) (Table III).

Table II

Allele and genotypic frequencies of miR-148b-3p mutations.

Table II

Allele and genotypic frequencies of miR-148b-3p mutations.

MutationInheritance modelGenotypeGroup BPDGroup PCaRM (95% CI)P-value
dupCT rs59761210DominantCT/CT21 (70%)32 (78%)0.66 (0.22-1.92)0.44
  CT/CTCT7 (23%)7 (17%)  
  CTCT/CTCT2 (7%)2 (5%)  
  CTCT0.180.130.66 (0.22-1.92)0.44
  G/G22 (73%)33 (81%)  
SNP G/T rs1381668656CodominantG/T8 (27%)8 (19%)0.67 (0.22-2.04)0.48
  T/T--  
  T0.130.10.72 (0.33-1.56)0.41
  G/G22 (74%)33 (80%)  
DelG rs368666376Recessive G/G-7 (23%)8 (20%)0.00 (0.00-NA)0.19
   G-/G-1 (3%)0 (0%)  
  G-0.130.10.41 (0.31-0.55)0.24
  G/G13 (44%)23 (56%)  
SNV G/C rs56818309OverdominantG/C16 (53%)16 (39%)0.56 (0.22-1.45)0.23
  C/C1 (3%)2 (5%)  
  C0.30.240.56 (0.22-1.45)0.23
  A/A23 (77%)36 (88%)  
SNV A/C 54337021CodominantA/C7 (23%)5 (12%)0.46 (0.13-1.61)0.22
  C/C--  
  C0.120.060.49 (0.15-1.63)0.24
  AAG/AAG26 (87%)25 (61%)  
DelsAAGDominant AAG/AAG-1 (3%)9 (22%)4.16 (1.22-14.17)0.014
   AAG-/AAG-3 (10%)7 (17%)  
   AAG-0.120.284.16 (1.22-14.17)0.017

[i] MicroRNA, miR; BPD, benign prostatic disease; PCa, prostate cancer; RM, odds ratio.

Table III

Allele and genotypic frequencies of the rs353291 mutation.

Table III

Allele and genotypic frequencies of the rs353291 mutation.

MutationInheritance modelGenotypeGroup BPDGroup PCaRM (95% CI)P-value
SNV T/C rs353291 T/T18 (78.3%)13 (61.9%)  
     3.33 (0.73-15.17)0.11
 SobredominantT/C3 (13%)7 (33.3%)  
  C/C2 (8.7%)1 (4.8%)3.33 (0.73-15.17)0.11
  C0.150.21  

[i] BPD, benign prostatic disease; PCa, prostate cancer; RM, odds ratio.

Linkage disequilibrium and analysis by haplotypes

Linkage disequilibrium analysis was performed to identify possible haplotypes, where the set of mutations of rs368666376, rs56818309, and DelsAAG was found to be in linkage disequilibrium. In total, three haplotypes (haplotypes I, II and III) were obtained from the set of mutations, with ‘haplotype I’ having the normal alleles. An analysis was performed in which each haplotype was linked to the presence of PCa. However, no statistically significant association was found. In the case of miR-145-5p, since only one mutation was studied, this analysis was not performed.

Relationship of mutations with miRNAs expression

The gene expression of miR-148b-3p between the genotypes and alleles of each mutation was compared and was observed that there is a statistically significant difference between the expression of the DelsAAG mutation genotypes (P=0.004), specifically between the normal homozygous genotypes with a mean expression of -0.35 and heterozygous with a mean of -2.10 (P=0.003) (Table IV). Similarly, this analysis compared alleles, where it was identified that the patients carrying the mutated allele of the DelsAAG mutation had a mean of -1.53. In the case of non-carriers, a mean of -0.36 was found, with a statistically significant difference (P=0.005). The remaining mutations were not observed with significant differences in the analyses by genotypes or by alleles (Table V). To identify a relationship between the rs353291 mutation and the expression of miR-145-5p, mean difference analyses were performed between genotypes and alleles. It was observed that the patients with the heterozygous genotype had a mean miR-145-5p expression of -0.38, however, no statistically significant difference was observed when compared with the other genotypes. The allele analysis revealed that patients carrying the mutated allele had a mean expression of -0.38 and non-carrier patients had a mean expression of -0.17; however, no statistically significant difference was observed between groups (P=0.47).

Table IV

Analysis of the mean difference in the expression of miR-148b-3p between the genotypes of the mutations in the study population.

Table IV

Analysis of the mean difference in the expression of miR-148b-3p between the genotypes of the mutations in the study population.

MutationGenotypeNAverageStandard errorP-value
dupCT rs59761210CT/CT53-0.880.23 
 CT/CTCT14-0.880.300.17
 CTCT/CTCT40.410.71 
SNP G/T rs1381668656G/G55-0.540.23 
 G/T16-0.730.330.69
 T/T0-- 
DelG rs368666376G/G56-0.710.21 
  G/G-14-0.660.490.9
  G-/G-1-- 
SNV G/C rs56818309G/G36-1.030.31 
 G/C32-0.340.230.18
 C/C3-0.270.29 
SNV A/C 54337021A/A59-0.690.21 
 A/C12-0.660.440.95
 C/C0-- 
DelsAAGAAG/AAG51-0.360.19 
  AAG/AAG-10-2.100.750.004 (0.03a)
  AAG-/AAG-10-0.950.33 

[i] aMultiple comparisons using Tukey's test, difference between normal homozygous and heterozygous genotypes.

Table V

Analysis of the mean difference in the expression of miR-148b-3p between the alleles of the mutations in the study population.

Table V

Analysis of the mean difference in the expression of miR-148b-3p between the alleles of the mutations in the study population.

MutationAlleleNAverageStandard errorP-value
dupCT rs59761210Carrier18-0.130.280.046
 Normal53-0.880.23 
SNP G/T rs1381668656Carrier33-0.530.220.53
 Normal109-0.730.16 
DelG rs368666376Carrier10.03-0.65
 Normal70-0.70.19 
SNV G/C rs56818309Carrier32-0.340.230.09
 Normal39-0.980.28 
SNV A/C 54337021Carrier12-0.660.440.95
 Normal130-0.70.14 
DelsAAGCarrier20-1.530.420.005
 Normal51-0.360.19 

Discussion

miRNAs are small non-coding RNAs and are part of the epigenetic machinery that can regulate the expression of genes; therefore, increasingly altered mechanisms are identified due to the malfunction of these miRNAs, for example, it has been observed that several miRNAs dysregulated in PCa, are related to the regulatory T cell marker FOXP3, suggesting a relationship between immune cells, miRNAs and PCa (16). Therefore, the identification of dysregulated miRNAs could have implications both in understanding the pathology and in identifying possible targets for the detection, prognosis and treatment of various cancers (17,18).

The dysregulation of the expression of a miRNA can be due to mutations in its coding genes or in their regulatory regions; for example, SNPs in miR-146a and in miR-100 have been identified that act as a factor of bad and an improved prognosis in PCa. In addition, it has been mentioned that these polymorphisms could alter miRNA function (19). Furthermore, it has been observed that mutations can affect the biogenesis of the mature miRNA while it is in the ‘seed’ region of the passenger strand, thus decreasing its expression (20,21). Finally, mutations in the promoter regions can also cause changes in the expression of miRNAs, which have been observed in the miR-143/145 cluster associated with PCa (22).

In the case of miR-145-5p, several mutations were identified; among them, the one with the highest frequency was the rs353291 mutation. This mutation is located in an intronic region of the CARMN gene, 450 bp downstream of miR-145-5p. According to the NCBI database, this SNV has a mutated allele frequency of C=0.40 and normal allele frequency of T=0.60 in the global population. In the present study, the frequencies in the general population were C=0.18 and T=0.82, which are remarkably similar to those reported in the African and Afro-American population (C=0.23 and T=0.77) and to those reported in Latin Americans (C=0.30 and T=0.70). The Asian population is the most distinct from the frequencies observed in the present study (C=0.41 and T=0.59) (23).

The present results indicated that the rs353291 mutation is unrelated to CaP or miR-145-5p underexpression. At present, there is only one study that analyzes the relationship of this miR-145-5p SNV with cancer in an Australian population, which observed that the mutated allele was significantly related to breast cancer; additionally, it was suggested that this SNV could be part of the gene regulation mechanisms of miR-145-5p. It should be noticed, however, that they did not analyze miRNA expression (24). The dysregulation of this miRNA in CaP is probably caused by molecular mechanisms other than mutations close to the miRNA, such as DNA methylation. This is consistent with previous research; Zaman et al (2010) (25), reported that the promoter of this miRNA was hypermethylated in PCa cell lines. Similarly, previous studies have shown that methylation plays a role in regulating miR-145-5p expression, which contributes to tumor growth and response to treatment (26,27). Furthermore, Xue et al (2015) (28), observed that miR-145-5p targets the DNMT3b methyltransferase, one of its mechanisms as a tumor suppressor miRNA. In addition, they observed that DNMT3b leads to hypermethylation of the miRNA promoter, with feedback regulation between the two. To the best of the authors' knowledge, the only study analyzing the regulation of this miRNA by epigenetic mechanisms in tissue from patients with PCa is that of Suh et al (2011) (29) in the United States population, who observed a relationship between the underexpression of this miRNA and the hypermethylation in its promoter region. This suggested that epigenetic mechanisms in the regulation of miR-145-5p may be the most common cause of its gene expression dysregulation.

At present, the mechanisms that lead to the dysregulation of miR-148b-3p in PCa have not been studied. The present study identified a set of deletions with a start site at chr12:54337042/54337043, which were unified and called DelsAAG. This mutation was related to the presence of PCa; likewise, it was associated with the underexpression of miR-148b-3p. These deletions are located in a particular region, specifically in the first intron of the COPZ1 gene, in the sequence comprising chr12:54337020–54337044, there is a set of 25 adenines in tandem, behaving as a pure or perfect microsatellite, and as previously reported, this type of repeated sites are hot spots for mutations, as they are susceptible to the generation of deletions and insertions, being highly polymorphic, having mutation rates ranging from 103 and 106 for each cell generation, thus this could explain the existence of the mutations in the analyzed site (30-32). According to the GeneCards database (33), this mutation is in a region that functions as a proximal intragenic enhancer (ID: GH12J054336), which could be a key cis-acting regulator promoting the expression of miR-148b-3p (34). Similarly, according to the ENSEMBL database, this mutation is found within a binding site for the transcription factors E2F1, E2F2 and E2F3 (chr:54337040-54337055) (35); previously these transcription factors have been identified to regulate the expression of other miRNAs, such as miRNAs 17 and 20, by binding to their promoter regions and promoting their expression (36,37). This is consistent with previous studies, as ~50% of the variants or mutations in transcription factor binding regions (RUFT) are in intronic regions (38). Previously, some RUFT mutations in Pac have been reported to modify their affinity and be involved in pathology development. For example, Huang et al (2014) (39), reported that an SNV increases the binding affinity of HOXB13, resulting in the overexpression of RFX6, leading to proliferation, migration and invasion of prostate tumor cells. Likewise, the rs7077275 polymorphism increases the binding of CTFC, which leads to the overexpression of CTBP2, decreasing apoptosis, and increasing tumor growth in PCa (40). Eickhardt et al (41) reported in 2016 that deletions are more disruptive variations than SNVs, leading to a more significant modification in the affinity of the transcription factors for the affected sequence. The aforementioned description indicated that the DelsAAG mutation could affect the binding of transcription factors, resulting in the absence or reduction of miR-148b-3p transcription.

This miRNA is directly involved in molecular mechanisms related to the development of PCa. Tomeva et al (2022) (42), observed that miR-148b-3p was underexpressed in tumors that occur at the androgen receptor (AR). Feng et al (43), in 2019 demonstrated that this miRNA was underexpressed in tumor tissue of patients with PCa. In addition, they observed that the 3'-untranlated region of the KLF4 gene contains the conserved miR-148b-3p binding site, making it a potential target for this miRNA binding, which was verified through their in vitro experiments (43). KLF4 is a transcription factor that can regulate the expression of various genes and that has been previously observed to be overexpressed in PCa (44). This transcription factor has an important implication in the prostate, since Siu et al (2016) (45), demonstrated a reciprocal relationship between KLF4 and the AR, where AR can induce the expression of this transcription factor. KLF4 can then bind to the AR promoter generating its transcription. In turn, AR activity is commonly altered in PCa, which promotes the evolution and progression of this cancer (46,47). Therefore, it was proposed by the auhtors that the mutation near miR-148b-3p, which includes a set of deletions in a binding site for transcription factors, promotes miRNA underexpression, which means that it cannot regulate the expression of KLF4 increasing its expression and, as a result, leading to the overexpression of the AR, enhancing the proliferation and progression of PCa (Fig. 3).

In conclusion, mutations in regions near the gene that codes for a miRNA may be related to its expression and be part of the regulation mechanisms in a pathogenic event such as PCa. Thus, the present study becomes the first to propose a method for regulating the expression of miR-148b-3p in PCa, involving deletions in a transcription factor binding site.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Autonomous University of Sinaloa (grant no. PROFAPI PRO_A3_001) and the Polytechnic University of Sinaloa internal resource.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

FBH and FLO performed the statistical analysis, critically reviewed literature findings and drafting the manuscript. NGM, MAA and EAM collected the sample, conceived and designed the study and revised the manuscript. FBH and NGM confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved (approval no. P-3103) by the Alvarez and Arrazola Radiologists Clinic's Ethics and Research Committee (Mazatlán, Mexico). Written informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Xiao J, Gong AY, Eischeid AN, Chen D, Deng C, Young CY and Chen XM: miR-141 modulates androgen receptor transcriptional activity in human prostate cancer cells through targeting the small heterodimer partner protein. Prostate. 72:1514–1522. 2012.PubMed/NCBI View Article : Google Scholar

2 

Sun Q, Zhao X, Liu X, Wang Y, Huang J, Jiang B, Chen Q and Yu J: miR-146a functions as a tumor suppressor in prostate cancer by targeting Rac1. Prostate. 74:1613–1621. 2014.PubMed/NCBI View Article : Google Scholar

3 

Bergez-Hernández F, Arámbula-Meraz E, Alvarez-Arrazola M, Irigoyen-Arredondo M, Luque-Ortega F, Martínez-Camberos A, Cedano-Prieto D, Contreras-Gutiérrez J, Martínez-Valenzuela C and García-Magallanes N: Expression Analysis of miRNAs and their potential role as biomarkers for prostate cancer detection. Am J Mens Health. 16(15579883221120989)2022.PubMed/NCBI View Article : Google Scholar

4 

Arámbula-Meraz E, Bergez-Hernández F, Leal-León E, Romo-Martínez E, Picos-Cárdenas V, Luque-Ortega F, Romero-Quintana J, Alvarez-Arrazola M and García-Magallanes N: Expression of miR-148b-3p is correlated with overexpression of biomarkers in prostate cancer. Genet Mol Biol. 43(e20180330)2020.PubMed/NCBI View Article : Google Scholar

5 

Xu WX, Liu Z, Deng F, Wang DD, Li XW, Tian T, Zhang J and Tang JH: MiR-145: A potential biomarker of cancer migration and invasion. Am J Transl Res. 11:6739–6753. 2019.PubMed/NCBI

6 

Jin W, Fei X, Wang X, Song Y and Chen F: Detection and prognosis of prostate cancer using blood-based biomarkers. Mediators Inflamm. 2020(8730608)2020.PubMed/NCBI View Article : Google Scholar

7 

Chu H, Zhong D, Tang J, Li J, Xue Y, Tong N, Qin C, Yin C, Zhang Z and Wang M: A functional variant in miR-143 promoter contributes to prostate cancer risk. Arch Toxicol. 90:403–414. 2016.PubMed/NCBI View Article : Google Scholar

8 

Kotarac N, Dobrijevic Z, Matijasevic S, Savic-Pavicevic D and Brajuskovic G: Analysis of association of potentially functional genetic variants within genes encoding miR-34b/c, miR-378 and miR-143/145 with prostate cancer in Serbian population. EXCLI J. 18:515–529. 2019.PubMed/NCBI View Article : Google Scholar

9 

Watahiki A and Wang Y, Morris J, Dennis K, O'Dwyer HM, Gleave M, Gleave M, Gout PW and Wang Y: MicroRNAs associated with metastatic prostate cancer. PLoS One. 6(e24950)2011.PubMed/NCBI View Article : Google Scholar

10 

Chen X, Wang G, Lu X, Gao P, Song Y, Sun J, Li A, Xu Y, Xu H and Wang Z: Polymorphisms and haplotypes of the miR-148/152 family are associated with the risk and clinicopathological features of gastric cancer in a Northern Chinese population. Mutagenesis. 29:401–407. 2014.PubMed/NCBI View Article : Google Scholar

11 

Chomczynski P: A reagent for the single-step simultaneous isolation of RNA, DNA and proteins from cell and tissue samples. Biotechniques. 15:532–4, 536-7. 1993.PubMed/NCBI

12 

Kim YG, Kim MJ, Lee JS, Lee JA, Song JY, Cho SI, Park SS and Seong MW: SnackVar: An open-source software for sanger sequencing analysis optimized for clinical use. J Mol Diagn. 23:140–148. 2021.PubMed/NCBI View Article : Google Scholar

13 

Rijk PD and Del-Favero J: novoSNP3: Variant detection and sequence annotation in resequencing projects. Methods Mol Biol. 396:331–344. 2007.PubMed/NCBI View Article : Google Scholar

14 

Taylor SC, Nadeau K, Abbasi M, Lachance C, Nguyen M and Fenrich J: The Ultimate qPCR experiment: Producing publication quality, reproducible data the first time. Trends Biotechnol. 37:761–774. 2019.PubMed/NCBI View Article : Google Scholar

15 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

16 

Akalin I, Erol B, Aslan E, Ozkanli SS, Efiloglu O, Yildirim S, Caskurlu T, Yildirim A and Karaman MI: A new promising pathway in aggressive prostate cancer: Treg/mir-let8c/lin28b. Arch Esp Urol. 75:459–466. 2022.PubMed/NCBI View Article : Google Scholar

17 

Liu S, Zhao Y, Zhao Y and Tang X: The prognostic value of miR-487a in clear cell renal cell carcinoma and its influence on cell biological behavior. Arch Esp Urol. 75:346–353. 2022.PubMed/NCBI View Article : Google Scholar

18 

Wang Z, Zhu X, Zhai H, Wang Y and Hao G: Integrated analysis of mRNA-single nucleotide polymorphism-microRNA interaction network to identify biomarkers associated with prostate cancer. Front Genet. 13(922712)2022.PubMed/NCBI View Article : Google Scholar

19 

Camargo JA, Lopes RE, Ferreira GFD, Viana NI, Guimaraes V, Leite KRM, Nahas WC, Srougi M, Antunes AA and Reis ST: The role of single nucleotide polymorphisms of miRNAs 100 and 146a as prognostic factors for prostate cancer. Int J Biol Markers. 36:50–56. 2021.PubMed/NCBI View Article : Google Scholar

20 

Xu B, Feng NH, Li PC, Tao J, Wu D, Zhang ZD, Tong N, Wang JF, Song NH, Zhang W, et al: A functional polymorphism in Pre-miR-146a gene is associated with prostate cancer risk and mature miR-146a expression in vivo. Prostate. 70:467–472. 2010.PubMed/NCBI View Article : Google Scholar

21 

Huang S, Cui H, Lou Z, Wang X, Chen L, Xie Z, Hehir M, Yao X, Ren Y, Cen D and Weng G: Association of rs3787016 in Long Non-coding RNAs POLR2E and rs2910164 in MiRNA-146a with prostate cancer: A systematic review and meta-analysis. Iran J Public Health. 47:623–632. 2018.PubMed/NCBI

22 

Zhao CG, Xu HB and Xu B: The rs4705342 gene mutation in the promoter region of the miR-143/145 cluster associated with the risk of prostate cancer in the Chinese Han population. Zhonghua Nan Ke Xue. 25:696–702. 2019.PubMed/NCBI(In Chinese).

23 

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.PubMed/NCBI View Article : Google Scholar

24 

Chacon-Cortes D, Smith RA, Haupt LM, Lea RA, Youl PH and Griffiths LR: Genetic association analysis of miRNA SNPs implicates MIR145 in breast cancer susceptibility. BMC Med Genet. 16(107)2015.PubMed/NCBI View Article : Google Scholar

25 

Zaman MS, Chen Y, Deng G, Shahryari V, Suh SO, Saini S, Majid S, Liu J, Khatri G, Tanaka Y and Dahiya R: The functional significance of microRNA-145 in prostate cancer. Br J Cancer. 103:256–264. 2010.PubMed/NCBI View Article : Google Scholar

26 

Bellissimo T, Ganci F, Gallo E, Sacconi A, Tito C, De Angelis L, Pulito C, Masciarelli S, Diso D, Anile M, et al: Thymic Epithelial Tumors phenotype relies on miR-145-5p epigenetic regulation. Mol Cancer. 16(88)2017.PubMed/NCBI View Article : Google Scholar

27 

Donzelli S, Mori F, Bellissimo T, Sacconi A, Casini B, Frixa T, Roscilli G, Aurisicchio L, Facciolo F, Pompili A, et al: Epigenetic silencing of miR-145-5p contributes to brain metastasis. Oncotarget. 6:35183–35201. 2015.PubMed/NCBI View Article : Google Scholar

28 

Xue G, Ren Z, Chen Y, Zhu J, Du Y, Pan D, Li X and Hu B: A feedback regulation between miR-145 and DNA methyltransferase 3b in prostate cancer cell and their responses to irradiation. Cancer Lett. 361:121–127. 2015.PubMed/NCBI View Article : Google Scholar

29 

Suh SO, Chen Y, Zaman MS, Hirata H, Yamamura S, Shahryari V, Liu J, Tabatabai ZL, Kakar S, Deng G, et al: MicroRNA-145 is regulated by DNA methylation and p53 gene mutation in prostate cancer. Carcinogenesis. 32:772–778. 2011.PubMed/NCBI View Article : Google Scholar

30 

Nesta AV, Tafur D and Beck CR: Hotspots of human mutation. Trends Genet. 37:717–729. 2021.PubMed/NCBI View Article : Google Scholar

31 

Gemayel R, Cho J, Boeynaems S and Verstrepen KJ: Beyond junk-variable tandem repeats as facilitators of rapid evolution of regulatory and coding sequences. Genes (Basel). 3:461–480. 2012.PubMed/NCBI View Article : Google Scholar

32 

Vieira ML, Santini L, Diniz AL and Munhoz Cde F: Microsatellite markers: What they mean and why they are so useful. Genet Mol Biol. 39:312–328. 2016.PubMed/NCBI View Article : Google Scholar

33 

Fishilevich S, Nudel R, Rappaport N, Hadar R, Plaschkes I, Iny Stein T, Rosen N, Kohn A, Twik M, Safran M, et al: GeneHancer: Genome-wide integration of enhancers and target genes in GeneCards. Database (Oxford). 2017(bax028)2017.PubMed/NCBI View Article : Google Scholar

34 

Peng Y and Zhang Y: Enhancer and super-enhancer: Positive regulators in gene transcription. Animal Model Exp Med. 1:169–179. 2018.PubMed/NCBI View Article : Google Scholar

35 

Cunningham F, Allen JE, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, Austine-Orimoloye O, Azov AG, Barnes I, Bennett R, et al: Ensembl 2022. Nucleic Acids Res. 50:D988–D995. 2022.PubMed/NCBI View Article : Google Scholar

36 

Sylvestre Y, De Guire V, Querido E, Mukhopadhyay UK, Bourdeau V, Major F, Ferbeyre G and Chartrand P: An E2F/miR-20a autoregulatory feedback loop. J Biol Chem. 282:2135–2143. 2007.PubMed/NCBI View Article : Google Scholar

37 

Woods K, Thomson JM and Hammond SM: Direct regulation of an oncogenic micro-RNA cluster by E2F transcription factors. J Biol Chem. 282:2130–2134. 2007.PubMed/NCBI View Article : Google Scholar

38 

de Santiago I, Liu W, Yuan K, O'Reilly M, Chilamakuri CS, Ponder BA, Meyer KB and Markowetz F: BaalChIP: Bayesian analysis of allele-specific transcription factor binding in cancer genomes. Genome Biol. 18(39)2017.PubMed/NCBI View Article : Google Scholar

39 

Huang Q, Whitington T, Gao P, Lindberg JF, Yang Y, Sun J, Väisänen MR, Szulkin R, Annala M, Yan J, et al: A prostate cancer susceptibility allele at 6q22 increases RFX6 expression by modulating HOXB13 chromatin binding. Nat Genet. 46:126–135. 2014.PubMed/NCBI View Article : Google Scholar

40 

Zhang P, Tillmans LS, Thibodeau SN and Wang L: Single-Nucleotide polymorphisms sequencing identifies candidate functional variants at prostate cancer risk loci. Genes (Basel). 10(547)2019.PubMed/NCBI View Article : Google Scholar

41 

Eickhardt E, Als TD, Grove J, Boerglum AD and Lescai F: Estimating the functional impact of INDELs in transcription factor binding sites: A Genome-Wide Landscape. Bioinformatics, 2016 Jun. Available from: http://biorxiv.org/lookup/doi/10.1101/057604.

42 

Tomeva E, Switzeny OJ, Heitzinger C, Hippe B and Haslberger AG: Comprehensive approach to distinguish patients with solid tumors from healthy controls by combining androgen receptor mutation p. H875Y with Cell-Free DNA methylation and circulating miRNAs. Cancers (Basel). 14(462)2022.PubMed/NCBI View Article : Google Scholar

43 

Feng F, Liu H, Chen A, Xia Q, Zhao Y, Jin X and Huang J: miR-148-3p and miR-152-3p synergistically regulate prostate cancer progression via repressing KLF4. J Cell Biochem. 120:17228–17239. 2019.PubMed/NCBI View Article : Google Scholar

44 

Wei LZ, Wang YQ, Chang YL, An N, Wang X, Zhou PJ, Zhu HH, Fang YX and Gao WQ: Imbalance of a KLF4-miR-7 auto-regulatory feedback loop promotes prostate cancer cell growth by impairing microRNA processing. Am J Cancer Res. 8:226–244. 2018.PubMed/NCBI

45 

Siu MK, Suau F, Chen WY, Tsai YC, Tsai HY, Yeh HL and Liu YN: KLF4 functions as an activator of the androgen receptor through reciprocal feedback. Oncogenesis. 5(e282)2016.PubMed/NCBI View Article : Google Scholar

46 

Desai K, McManus JM and Sharifi N: Hormonal therapy for prostate cancer. Endocr Rev. 42:354–373. 2021.PubMed/NCBI View Article : Google Scholar

47 

Auchus RJ and Sharifi N: Sex Hormones and Prostate Cancer. Annu Rev Med. 71:33–45. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2024
Volume 20 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bergez‑Hernández F, Luque‑Ortega F, García‑Magallanes N, Alvarez‑Arrazola M and Arámbula‑Meraz E: Deletion in a regulatory region is associated with underexpression of miR-148b‑3p in patients with prostate cancer. Biomed Rep 20: 52, 2024
APA
Bergez‑Hernández, F., Luque‑Ortega, F., García‑Magallanes, N., Alvarez‑Arrazola, M., & Arámbula‑Meraz, E. (2024). Deletion in a regulatory region is associated with underexpression of miR-148b‑3p in patients with prostate cancer. Biomedical Reports, 20, 52. https://doi.org/10.3892/br.2024.1740
MLA
Bergez‑Hernández, F., Luque‑Ortega, F., García‑Magallanes, N., Alvarez‑Arrazola, M., Arámbula‑Meraz, E."Deletion in a regulatory region is associated with underexpression of miR-148b‑3p in patients with prostate cancer". Biomedical Reports 20.3 (2024): 52.
Chicago
Bergez‑Hernández, F., Luque‑Ortega, F., García‑Magallanes, N., Alvarez‑Arrazola, M., Arámbula‑Meraz, E."Deletion in a regulatory region is associated with underexpression of miR-148b‑3p in patients with prostate cancer". Biomedical Reports 20, no. 3 (2024): 52. https://doi.org/10.3892/br.2024.1740