A microRNA‑125a variant, which affects its mature processing, increases the risk of radiation‑induced pneumonitis in patients with non‑small‑cell lung cancer

  • Authors:
    • Hong‑Yan Quan
    • Tian Yuan
    • Jian‑Feng Hao
  • View Affiliations

  • Published online on: August 20, 2018     https://doi.org/10.3892/mmr.2018.9406
  • Pages: 4079-4086
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to investigate the role of microRNA (miR)‑125a in the development of pneumonitis inpatients with non‑small‑cell lung cancer that received radiotherapy. In addition, the study aimed to determine how the miR‑125a affects its target, transforming growth factor β (TGFβ). Bioinformatics tools were used to identify a potential miR‑125a binding site in the 3'untranslated region of TGFβ, which was subsequently confirmed using a dual‑luciferase reporter system. In addition, tissue samples were collected from patients with lung cancer and genotyped as CC (n=36), CT (n=28) or TT (n=6). The expression levels of miR‑125a and TGFβ in these samples were determined, and CC genotype samples demonstrated upregulated miR‑125a expression, and downregulated TGFβ protein and mRNA expression compared with samples carrying the minor allele, T. To further investigate the association between the rs12976445 polymorphism and the risk of pneumonitis in patients with lung cancer that received radiotherapy, 534 lung cancer patients diagnosed with pneumonitis and 489lung cancer patients without pneumonitis were recruited. rs12976445 was shown to be significantly associated with the risk of pneumonitis. In conclusion, the rs12976445 polymorphism increased expression levels of TGFβ by decreasing the expression of miR‑125a, and therefore may be associated with the development of pneumonitis in patients with lung cancer that receive radiotherapy.

Introduction

Lung cancer is the leading cause of cancer-associated mortality, with 1 million patients worldwide each year succumbing to this disease (1). The majority of lung cancer cases (>80%) are non-small cell lung cancer (NSCLC), which consists of large cell carcinoma, adenocarcinoma and squamous cell carcinoma. However, ~30% of patients with NSCLC do not receive a specific classification (2,3). A standard treatment approach for patients with unresectable NSCLC is radiotherapy alone or combined with chemotherapy, as this may improve overall survival and control or reduce the lesion site, and prevent metastases (4). It has been reported that radiation-induced pneumonitis in patients with NSCLC is correlated with numerous factors (5). The most extensively investigated factors include dose-volume parameters to healthy lung tissue and selected cytokines (6); however, thus far none have been demonstrated to be accurate enough to control the radiation dose to prevent further damage. In addition, it is difficult to balance the requirement for high-dose radiation to achieve adequate tumor control with the need to limit the dose to healthy lung tissue in patients with larger tumors (7). A previous study demonstrated that inflammatory and fibrogenic cytokines are critical in the development of pneumonitis as a complication of radiotherapy (8). Inflammation may be regulated by transforming growth factor β (TGFβ), a pleiotropic cytokine that stimulates connective tissue collagen formation and reduces degradation, thus resulting in fibrosis (9). TGFβ has been revealed to be markedly associated with damage to the lung architecture (8). Previous studies have identified TGFβ as a marker of patients at risk of developing symptomatic pneumonitis (10,11).

In the majority of cells, differentiation, proliferation and other functions are controlled by the TGFβ1 gene. Numerous cellular processes in the developing embryo and the adult organism, including homeostasis, apoptosis, differentiation and growth are associated with the TGFβ1 signaling pathway (12). As an important modulator of the inflammatory response, TGFβ has been extensively investigated in the irradiation-induced development of tissue fibrosis (13). Plasma levels of TGFβ1 have been investigated as a predictor for lung injury induced by radiotherapy. Human and animal studies have indicated that TGFβ acts as primary regulator in the process of lung injury induced by radiation (7,14,15). Following administration of anti-TGFβ antibodies, TGFβ activation was reduced and the inflammatory response decreased several weeks after radiotherapy, further demonstrating that targeting the TGFβ signaling pathway may be a potential strategy to prevent lung injury induced by radiotherapy (16).

MicroRNAs (miRNAs) are endogenously encoded single-stranded RNAs ~22 nucleotides in length, which are essential in various pathological conditions (17). miRNAs bind specifically to target messenger RNAs (mRNAs) and induce mRNA degradation or translational repression, resulting in posttranscriptional gene suppression (18). Various biological processes, including immunity, inflammation, cellular development, apoptosis, metabolism, differentiation and proliferation are regulated by miRNAs (1719). However, a single nucleotide polymorphism (SNP; rs12976445) in pre-microRNA-125a may compromise the mature processing of the miRNA, reducing miR-125a production (20). Therefore, it has been hypothesized that rs12976445 may be associated with the risk of developing pneumonitis in patients with NSCLC treated with radiotherapy, by affecting the production of miR-125a, and consequently its target, TGFβ expression.

Materials and methods

Ethical considerations

The present study was conducted according to the Declaration of Helsinki guidelines and the study protocol was approved by the Human Ethics Committee of Shaanxi Friendship Hospital (Xi'an, China). Written informed consent was obtained from all participants prior to the study.

Patient eligibility and study design

The present study was performed on patients with NSCLC (n=1,023) who were admitted to the Oncology Department, Shaanxi Friendship Hospital. All patients received chest computed tomography (CT), pulmonary function tests, biochemical analysis, a complete blood count and a physical examination, and a complete medical history was taken (Table I). The patients were recorded and treated in accordance with the Radiation Therapy Oncology Group and the European Organization for the Research and Treatment of Cancer guidelines. All patients were followed up; assessments included chest CT, chest X-ray and a physical examination. Assessments were performed at three-month intervals for a period of two years following the completion of radiotherapy, and subsequently at six-month intervals. Patients were divided into two groups based on the presence or absence of pneumonitis (pneumonitis positive, n=534; pneumonitis negative, n=489). The criteria for enrollment in the present study included: i) A diagnosis of NSCLC; ii) no history of surgery and chemotherapy; iii) good heart, liver and kidney function; iv) no distant metastasis; v) no history of myocardial infarction, cerebral infarction or other critical illness in the previous six months; vi) a life expectancy of at least six months; and vii) a prescribed radiation dose of 50–70 Gy. Patients with any of the following were excluded: i) A history of surgery or chemotherapy for a thoracic tumor; ii) a history of severe pulmonary dysfunction or pulmonary fibrosis; iii) a history of total or partial pulmonary lobectomy; iv) poor general health; v) intolerance to radiation or incomplete radiotherapy; vi) a diagnosis of asthma, serious chronic bronchitis, emphysema or severe pulmonary infection; and vii) a diagnosis of another serious disease, including myocardial infarction and cerebral infarction within the previous six months. Lung tissue samples (70) were obtained from 1,023 NSCLC patients: [CC=36, 18 pneumonitis (+), 18, pneumonitis (−); CT=28, 14 pneumonitis (+), 14 pneumonitis (−); TT=6, 3 pneumonitis (+), 3 pneumonitis (−)] and genotyping for rs12976445 was conducted on blood samples obtained from all patients.

Table I.

Demographic and clinicopathological features of the participants of the present study.

Table I.

Demographic and clinicopathological features of the participants of the present study.

ParameterPneumonitis (+)Pneumonitis (−)
Number534489
Sex (M:F)534 (276:258)489 (254:235)
Average age (years)52.63±8.6351.37±9.33
Hypertension (%)4.287.87
Current smokers (%)12.142.86
Hyperlipidemia (%)2.049.36
Diabetes mellitus (%)2.341.27
Coronary heart disease (%)7.686.12
Body mass index (kg/m2)20.25±1.4121.03±0.97

[i] M, male; F, female; Data expressed as mean ± standard deviation.

Genotyping

The QIAamp DNA Blood Mini kit (Qiagen, Inc., Valencia, CA, USA) was used to extract genomic DNA from blood (200 µl) samples, according to the manufacturer's protocol. Sanger sequencing using an ABI PRISM® 3100 Genetic Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc., Waltham, MA, USA) was performed to determine the genotype of rs12976445.

Cell culture and transfection

A549 human lung carcinoma cells were obtained from the American Type Culture Collection (Manassas, VA, USA), and were cultured in Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 100 mg/ml streptomycin, 100 U/ml penicillin and 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a 5% CO2 atmosphere. Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) was used to perform transient cell transfections of small RNAs or plasmids. miR-125a mimic/antagomir (5′-CTATGTTTGAATGAGGCTTCAG-3′ and 5′-CGCGTCGCCGCGTGTTTAAACG-3′; Sangon Biotech Co., Ltd., China) or TGFβ small interfering (si)RNA (5′-CCCAGAACCAGGAGAAGAA-3′ and 5′-UUCUUCUCCUGGUUCUGGG-3′; Sangon Biotech Co., Ltd., China) were transfected at a final concentration of 100 nM. Briefly, the concentration of pcDNA3 was 16 µg/ml, the number of cells 2.0×105/ml and the duration and temperature of the transfection was 6 h at 37°C. A scramble control oligo sequence with no known target in the human genome (5′-TGCACAATTTGATGCCGGTTTAGTAT-3′ and 5′-TTAAAATGCAGATGCTGAACTGGGAA-3′; Invitrogen; Thermo Fisher Scientific, Inc.) served as a control.

Plasmid construction, target prediction and luciferase assay

By scanning target gene prediction databases (www.targetscan.org), the putative target gene of the miR-125a were pooled from 3 databases. Experimental validation on miR-125a and TGFβ was performed. The full-length cDNA of the human TGFβ gene was amplified by polymerase chain reaction (PCR) and was subcloned into a pcDNA3 vector (Invitrogen; Thermo Fisher Scientific, Inc.) using the following cloning primer: Forward, 5-ATGCCGCCCTCCGGGCTGCGGCTGCTG-3′ and reverse, 5′-TCAGCTGCACTTGCAGGAGCGCA-3′ (Sangon Biotech Co., Ltd., Shanghai, China). The following primers were used to build the TGFβ 3′untranslated region (UTR) reporter plasmids (psiCHECK; Promega Corporation, Madison, WI, USA): Forward, 5′-GGTCCCGCCCCGCCCCGCCCCGCCCCG-3′ and reverse, 5′-GGCCTGAACTACTATCTTTTA-3′. The predicted binding sequence (www.mirdb.org) was mutated using QuikChange XL Site-Directed Mutagenesis kit (Stratagene; Agilent Technologies, Inc., Santa Clara, CA, USA). A549 cells were seeded onto 6-well plates at a density of 5×105. Following transfection for 48 h, cells were lysed to determine reporter gene activity with the Dual-Luciferase® Reporter assay system (Promega Corporation, Madison, WI, USA), by measuring the proportion of Renilla plasmid vs. firefly plasmid (10:1). All experiments were performed in triplicate.

Western blot analysis

Cells were collected and lysed in radio immunoprecipitation assay buffer [1 mM phenylmethylsulfonyl fluoride, 0.1% sodium dodecyl sulfate (SDS), 1% sodium deoxycholate, 1% NP-40, 10% glycerol, 1 mM dithiothreitol, 150 mM NaCl and 50 mM Tris-HCl (pH 8.0)]. Cell lysates were cleared of cell debris by centrifugation at 20,800 × g for 15 min at 4°C. The proteins were extracted using a BCA Protein Assay kit and the concentration was determined by ultraviolet absorption (21), prior to being separated by 12% SDS-polyacrylamide gel electrophoresis with 35 µg protein loaded on each lane and transferred onto a polyvinylidene difluoride membrane (GE Healthcare Life Sciences, Chalfont, UK). Membranes were blocked with 5% non-fat milk in Tris-buffered saline with 0.1% Tween-20 at room temperature for 2 h. The membranes were subsequently probed with primary antibodies against TGFβ (anti-mouse; 3711S; 1:10,000; Cell Signaling Technology, Inc., Danvers, MA, USA) and β-actin (anti-mouse; 4967S; 1:10,000; Cell Signaling Technology, Inc.). Following three washes with TBST, membranes were incubated with secondary antibody (Goat anti mouse-IgG-HRP, 1:10,000, Cell Signaling Technology, Inc.). Signals were developed using an Enhanced Chemiluminescence kit (Thermo Fisher Scientific, Inc.). Immuno reactive bands were quantified individually by estimating the number of pixels in ImageJ software version 1.4.1 (National Institutes of Health, Bethesda, MD, USA).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from cells using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) in accordance with the manufacturer's protocol, later tested by the following 50 µl PCR reaction system: 37.5 µl ddH2O, 10 mm dNTP 1 µl, 10×5 µl PCR buffer, 25 mm Mgcl 2 3 µl, upstream primer 1 µl, downstream primers 1 µl, template cDNA 1 µl (100°C water bath for 1 min, 0.5 µl Tag enzymes added over ice then 50 µl liquid paraffin added followed by centrifugation at 11,180 × g at 4°C for 1 min. PCR conditions were: 94°C 1 min, 58°C 50 sec, 72°C 90 sec, 40 cycles and then 72°C for 10 min, followed by storage at −20°C. RNAs were then reverse transcribed using the PrimeScript™ RT-PCR kit (Takara Biotechnology Co., Ltd., Dalian, China). qPCR was performed on the resultant cDNA using the SYBR® Green Master mix (Takara Biotechnology Co., Ltd.) to determine the expression of miR-125a. PCR-master mix (45 µl) containing 2 µl of enzyme-mix from the Qiagen OneStep RT-PCR kit (including the RT and the hot-start Taq polymerase), 1X RT-PCR buffer (included in kit) 400 µM of each dNTP, 20U of Riblock RNase Inhibitor (Fermentas; Thermo Fisher Scientific, Inc.) and 200 nM of each forward and reverse primer was added to 5 µl of previously treated sample RNA. The initial steps of the cycle program consisted of 30 min at 50°C to produce enough specific initial DNA and 15 min at 95°C for the activation of the Taq polymerase. Overall, 40 cycles of 15 sec at 95°C, 30 sec at 58°C and 1 min at 72°C were performed. Following a final elongation step at 72°C for 10 min, the amplification product was cooled at 8°C. The following primers were used (Sangon Biotech Co., Ltd.): Forward, 5′-CGATGTCGTATATGCGTCGTATG-3′ and reverse, 5′-CGTAGTCGTCGTATGCTAGCGT-3′ for miR-125a; forward, 5′-CGTAGCTAGTCGTATGCTGA-3′ and reverse, 5′-CGATGTCGTAGGTCTAGCTGATGC-3′ for TGFβ. U6 forward, 5′-ATGACACGCAAATTCCCTCGAGGCGTGAAGCGTTCCATA-3′ and reverse, 5′-TATGGAACGCTTCACGCCTCGAGGGAATTTGCGTGTCAT-3′ small nuclear RNA served as an internal control for miRNA and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) forward, 5′-GCACCGTCAAGGCTGAGAAC-3′ and reverse, 5′-TGGTGAACGCCAGTGGA-3′ for mRNA. Data was normalized to U6 and GAPDH using the 2−ΔΔCq method (22).

Statistical analysis

The relative expression of miRNA in each target was expressed (mean ± standard deviation/standard error of the mean), and the difference expression of miRNA between different groups was compared with non-paired test or single factor variance analysis. The Hardy-Weinberg equilibrium was tested using the Pearson's Chi-square test (goodness-of-fit) for the SNP. Genotype distribution differences between cohorts were examined using the Pearson's χ2 test and logistic regression analysis. All statistical analyses were two-sided and were performed in SPSS software version 17.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference. The experiments were repeated three times.

Results

TGFβ is a target of miR-125a

Based on reports that TGFβ was functionally involved in the development of pneumonitis in patients with lung cancer that received radiotherapy, and that miR-125a negatively regulates TGFβ, the present study aimed to investigate the molecular mechanism underlying pneumonitis, a primary adverse effect of radio therapy in the treatment of lung cancer, including the downstream mediators and signaling pathways of miR-125a. Online miRNA target prediction tools (http://www.targetscan.org) were used to search the target gene of miR-125a; these identified TGFβ as a candidate target gene in lung cancer cells with the ‘seed sequence’ (390–397 bp) in the 3′UTR of TGFβ (Fig. 1).

To verify TGFβ as a direct target gene of miR-125a, a dual-luciferase activity reporter assay was performed on miR-125a-overexpressing lung cancer cells transfected with wild-type TGFβ 3′UTR constructs or mutant TGFβ 3′UTR constructs. As presented in Fig. 2, compared with the scramble controls, cells transfected with wild-type TGFβ 3′UTR constructs exhibited reduced relative luciferase activity (P<0.05), whereas cells transfected with mutant TGFβ 3′UTR constructs demonstrated comparable luciferase activity (P>0.05), indicating that TGFβ was a direct target of miR-125a and that the binding site is located at 390–397 bp of the 3′UTR of TGFβ.

Effects of rs12976445 on miR-125a expression

Due to the negative regulatory association between miR-125a and TGFβ, the effect of the rs12976445 polymorphism on the interaction between miR-125a and TGFβ was investigated. C-509T is located 509 bp upstream of the exon1 codon of the gene. Tissue samples were collected from patients with lung cancer and genotyped as CC (n=36), CT (n=18) or TT (n=6). The miR-125a expression levels of the three genotypes were then analyzed. As presented in Fig. 3, the expression levels of miR-125a were significantly greater in the CC genotype group compared with the genotype groups carrying the minor allele, the CT and TT groups (P<0.05). These two groups exhibited comparable miR-125a expression levels, suggesting that the presence of the minor allele, T compromised the expression of miR-125a.

To investigate the association between the rs12976445 polymorphism and the risk of pneumonitis in patients with lung cancer that received radio therapy, 534 lung cancer patients with diagnosed pneumonitis and 489 lung cancer patients without pneumonitis were enrolled in the present study. The distributions of the rs12976445 polymorphism were in Hardy-Weinberg equilibrium among the case group and controls. As the expression levels of miR-125a were similar in the TT and CT groups, and significantly reduced compared with the CC group, a dominant model of the minor allele was indicated. Since the frequency of TT was relatively low in the population assessed, the CT and TT groups were combined. Significant differences were observed regarding genotype distribution of rs12976445 between patients with and without pneumonitis [odds ratio (OR)=1.43; 95% confidence interval (CI)=1.13–1.94; P=0.03], as presented in Table II. This is despite the fact that the polymorphism is not located in the binding site of miR-125a in the 3′UTR of TGFβ, and the effect of the polymorphism on the interaction between miR-125a and TGFβ may therefore be attributed to an alteration in the secondary structure caused by the presence of the minor allele, or linkage disequilibrium with another variant that may directly cause the disruption between miR-125a and TGFβ. Alternatively, the polymorphism may compromise the transcription of TGFβ.

Table II.

miR-125a genotype distribution in lung cancer patients with and without pneumonitis.

Table II.

miR-125a genotype distribution in lung cancer patients with and without pneumonitis.

GenotypePneumonitis (+) (n=534) (%)Pneumonitis (−) (n=489) (%)Adjusted OR (95% CI)
CC363 (67.97)361 (73.82)
CT160 (29.96)122 (24.95)
TT11 (2.07)6 (1.23)
CT/TT171 (32.03)128 (26.18)1.43 (1.13–1.94)
P=0.03

[i] OR, odds ratio; CI, confidence interval.

RT-qPCR and western blot analyses were performed to investigate the mRNA and protein expression levels of TGFβ in each rs12976445 polymorphism genotype group. As presented in Fig. 4, the mRNA and protein expression levels of TGFβ were reduced in the CC genotype group, compared with the minor-allele-carrying CT/TT genotype groups (P<0.05), which had decreased expression levels of miR-125a. These findings further support the hypothesis of the negative association between miR-125a and TGFβ.

To investigate the interaction between miR-125a and TGFβ, the mRNA and protein expression levels of TGFβ in A549 cells transfected with TGFβ siRNA, an miR-125a mimic, and anti-miR-125a were analyzed and compared with the scramble controls. TGFβ protein expression levels were reduced in cells transfected with an miR-125a mimic or TGFβ siRNA, compared with the scramble control (Fig. 5A). TGFβ protein expression levels were greater in cells transfected with anti-miR-125a, compared with the scramble control (Fig. 5B). A similar pattern was observed in TGFβ mRNA expression levels (P<0.05; Fig. 5C and D). The above findings further validate the negative association between miR-125a and its target TGFβ.

Discussion

A common complication of lung radio therapy, radiation-induced lung injury, including radiation fibrosis and pneumonitis, is associated with a poor prognosis. Radio therapy-induced pneumonitis may be predicted by pulmonary function parameters in the clinic (23). The infiltration of inflammatory cells into alveolar spaces and the pulmonary interstitium is a characteristic of pneumonitis at the cellular level. In the present study, bioinformatics tools were used to identify potential binding sites of miR-125a in the 3′UTR of TGFβ, and this was subsequently confirmed using a dual-luciferase reporter system. In addition, tissue samples were collected from patients with lung cancer and genotyped as CC (n=36), CT (n=28) or TT (n=6). The expression levels of miR-125a and TGFβ were determined in these samples, which exhibited upregulated miR-125a and downregulated TGFβ protein and mRNA expression levels in cells genotyped as CC compared with cells carrying the minor allele, T.

It has previously been demonstrated that cytokines are involved in the development of pneumonitis (24). TGFβ is involved in the pathogenesis of radiation-induced pneumonitis; therefore, the outcomes of patients with radiation-induced pneumonitis may be predicted by TGFβ expression. As a ubiquitous profibrotic and immunomodulatory cytokine, TGFβ is essential in tissue responses to irradiation. It has been observed that rising plasma levels of TGFβ1 during thoracic radio therapy is associated with the subsequent occurrence of radiation-induced pneumonitis (2527). As a result, TGFβ has been adopted as a sensitive plasma marker of radiation-induced pneumonitis following thoracic irradiation. A previous animal study demonstrated that anti-TGFβ antibodies decreased the levels of TGFβ during thoracic irradiation, resulting in attenuation of radiation-induced pneumonitis (28). In the present study, miR-125a was identified as a regulator of TGFβ in A549 cells using in silico analysis and a dual-luciferase reporter system.

Previous studies have demonstrated that the C-509T SNP is associated with TGFβ gene promoter activity enhancement, and the T allele of C-509T is associated with increased serum levels of TGFβ. The serum level of TGFβ in the CC genotype is decreased compared with the CT or TT genotypes (29). In addition, a previous study revealed that greater circulating TGFβ levels during radiation therapy are correlated with poor prognosis of locally advanced NSCLC (30). Therefore, compared with those with TGFβ C-509T CT or TT genotypes, patients with CC genotype may have improved survival due to reduced circulating TGFβ levels during radio therapy. Furthermore, various studies have implicated TGFβ in radiation-induced lung injury; in particular, a persistently elevated or rising level of TGFβ during the course of thoracic radiation therapy has been associated with symptomatic radiation pneumonitis (14,27). Since miR-125a was identified as a regulator of TGFβ and rs12976445 interferes with the mature processing of this miRNA, reducing the miRNA expression levels, the present study examined the effect of rs12976445 on the expression levels of miR-125a and TGFβ. The expression levels of miR-125a were markedly greater in the CC genotype group compared with those carrying the minor allele, the CT and TT groups, which exhibited comparable miR-125a expression levels, indicating that the T allele is dominant.

The MIR125A gene, which encodes miR-125a, is situated in a gene cluster on chromo some 19q13.41. miR-125a has been reported to be involved in the pathogenesis of human diseases, including verrucous carcinoma (31), ovarian cancer (32), gastric cancer (33), breast cancer (34) and systemic lupus erythematosus (35). It has previously been demonstrated that an SNP (rs12976445) in the precursor of miR-125a, located in the stem loop of the molecule, may undermine the mature processing of the miRNA, leading to a decrease in the production of miR-125a (21). In the present study, 534 lung cancer patients with diagnosed pneumonitis and 489 lung cancer patients without pneumonitis were recruited, and significant differences were noted regarding genotype distribution of rs12976445 between those with and without pneumonitis (OR=1.43; 95% CI=1.13–1.94; P=0.03).

In conclusion, the results of the present study demonstrated that patients with radiation-induced pneumonitis have decreased expression levels of miR-125a in lung tissue. Presence of the minor allele of the rs12976445 polymorphism increased expression levels of TGFβ by decreasing the expression levels of miR-125a, and therefore may be associated with the development of pneumonitis in patients with lung cancer receiving radio therapy. These results suggested that rs12976445 may be a potential biomarker to predict the risk of pneumonitis following radio therapy.

Acknowledgements

Not applicable.

Funding

No funding was received.

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

H-YQ planned the study, collected and interpreted the data, assembled the literature and wrote the manuscript. TY collected and interpreted the data, and wrote the manuscript. J-FH interpreted the data, assembled the literature and approved the final manuscript.

Ethics approval and consent to participate

The present study was conducted according to the Declaration of Helsinki guidelines and the study protocol was approved by the Human Ethics Committee of Shaanxi Friendship Hospital.

Patient consent for publication

Written informed consent was obtained from all participants prior to the study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Midanik LT, Klatsky AL and Armstrong MA: Changes in drinking behavior: Demographic, psychosocial, and biomedical factors. Int J Addict. 25:599–619. 1990. View Article : Google Scholar : PubMed/NCBI

2 

Zhang WC, Shyh-Chang N, Yang H, Rai A, Umashankar S, Ma S, Soh BS, Sun LL, Tai BC, Nga ME, et al: Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis. Cell. 148:259–272. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Guan X, Yin M, Wei Q, Zhao H, Liu Z, Wang LE, Yuan X, O'Reilly MS, Komaki R and Liao Z: Genotypes and haplotypes of the VEGF gene and survival in locally advanced non-small cell lung cancer patients treated with chemoradiotherapy. BMC Cancer. 10:4312010. View Article : Google Scholar : PubMed/NCBI

4 

Yin M, Liao Z, Yuan X, Guan X, O'Reilly MS, Welsh J, Wang LE and Wei Q: Polymorphisms of the vascular endothelial growth factor gene and severe radiation pneumonitis in non-small cell lung cancer patients treated with definitive radiotherapy. Cancer Sci. 103:945–950. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Hildebrandt MA, Komaki R, Liao Z, Gu J, Chang JY, Ye Y, Lu C, Stewart DJ, Minna JD, Roth JA, et al: Genetic variants in inflammation-related genes are associated with radiation-induced toxicity following treatment for non-small cell lung cancer. PLoS One. 5:e124022010. View Article : Google Scholar : PubMed/NCBI

6 

Vlasova MA and Moshkovskii SA: Molecular interactions of acute phase serum amyloid A: Possible involvement in carcinogenesis. Biochemistry (Mosc). 71:1051–1059. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Zhao L, West BT, Hayman JA, Lyons S, Cease K and Kong FM: High radiation dose may reduce the negative effect of large gross tumor volume in patients with medically inoperable early-stage non-small cell lung cancer. Int J Radiat Oncol Biol Phys. 68:103–110. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Rübe CE, Wilfert F, Uthe D, König J, Liu L, Schuck A, Willich N, Remberger K and Rübe C: Increased expression of pro-inflammatory cytokines as a cause of lung toxicity after combined treatment with gemcitabine and thoracic irradiation. Radiother Oncol. 72:231–241. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Kong FM, Ao X, Wang L and Lawrence TS: The use of blood biomarkers to predict radiation lung toxicity: A potential strategy to individualize thoracic radiation therapy. Cancer Control. 15:140–150. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Fosslien E: Cancer morphogenesis: Role of mitochondrial failure. Ann Clin Lab Sci. 38:307–329. 2008.PubMed/NCBI

11 

Anscher MS, Kong FM, Marks LB, Bentel GC and Jirtle RL: Changes in plasma transforming growth factor beta during radiotherapy and the risk of symptomatic radiation-induced pneumonitis. Int J Radiat Oncol Biol Phys. 37:253–258. 1997. View Article : Google Scholar : PubMed/NCBI

12 

Burger A, Löffler H, Bamberg M and Rodemann HP: Molecular and cellular basis of radiation fibrosis. Int J Radiat Biol. 73:401–408. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Hakenjos L, Bamberg M and Rodemann HP: TGF-beta1-mediated alterations of rat lung fibroblast differentiation resulting in the radiation-induced fibrotic phenotype. Int J Radiat Biol. 76:503–509. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Zhao L, Wang L, Ji W, Wang X, Zhu X, Hayman JA, Kalemkerian GP, Yang W, Brenner D, Lawrence TS and Kong FM: Elevation of plasma TGF-beta1 during radiation therapy predicts radiation-induced lung toxicity in patients with non-small-cell lung cancer: A combined analysis from Beijing and Michigan. Int J Radiat Oncol Biol Phys. 74:1385–1390. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Xue J, Li X, Lu Y, Gan L, Zhou L, Wang Y, Lan J, Liu S, Sun L, Jia L, et al: Gene-modified mesenchymal stem cells protect against radiation-induced lung injury. Mol Ther. 21:456–465. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Martin M, Lefaix J and Delanian S: TGF-beta1 and radiation fibrosis: A master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys. 47:277–290. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Mendell JT: MicroRNAs: Critical regulators of development, cellular physiology and malignancy. Cell Cycle. 4:1179–1184. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Lehmann TP, Korski K, Ibbs M, Zawierucha P, Grodecka-Gazdecka S and Jagodziński PP: rs12976445 variant in the pri-miR-125a correlates with a lower level of hsa-miR-125a and ERBB2 overexpression in breast cancer patients. Oncol Lett. 5:569–573. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Beaven GH and Holida ER: Ultraviolet absorption spectra of proteins and amino acids. Adv Prot Chem. 7:319–386. 1952. View Article : Google Scholar

22 

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. View Article : Google Scholar : PubMed/NCBI

23 

Kocak Z, Borst GR, Zeng J, Zhou S, Hollis DR, Zhang J, Evans ES, Folz RJ, Wong T, Kahn D, et al: Prospective assessment of dosimetric/physiologic-based models for predicting radiation pneumonitis. Int J Radiat Oncol Biol Phys. 67:178–186. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Tsoutsou PG and Koukourakis MI: Radiation pneumonitis and fibrosis: Mechanisms underlying its pathogenesis and implications for future research. Int J Radiat Oncol Biol Phys. 66:1281–1293. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Chen Y, Williams J, Ding I, Hernady E, Liu W, Smudzin T, Finkelstein JN, Rubin P and Okunieff P: Radiation pneumonitis and early circulatory cytokine markers. Semin Radiat Oncol. 12 1 Suppl 1:S26–S33. 2002. View Article : Google Scholar

26 

Evans ES, Kocak Z, Zhou SM, Kahn DA, Huang H, Hollis DR, Light KL, Anscher MS and Marks LB: Does transforming growth factor-beta1 predict for radiation-induced pneumonitis in patients treated for lung cancer? Cytokine. 35:186–192. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Fu XL, Huang H, Bentel G, Clough R, Jirtle RL, Kong FM, Marks LB and Anscher MS: Predicting the risk of symptomatic radiation-induced lung injury using both the physical and biologic parameters V(30) and transforming growth factor beta. Int J Radiat Oncol Biol Phys. 50:899–908. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Anscher MS, Thrasher B, Rabbani Z, Teicher B and Vujaskovic Z: Antitransforming growth factor-beta antibody 1D11 ameliorates normal tissue damage caused by high-dose radiation. Int J Radiat Oncol Biol Phys. 65:876–881. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Grainger DJ, Heathcote K, Chiano M, Snieder H, Kemp PR, Metcalfe JC, Carter ND and Spector TD: Genetic control of the circulating concentration of transforming growth factor type beta1. Hum Mol Genet. 8:93–97. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Zhao L, Ji W, Zhang L, Ou G, Feng Q, Zhou Z, Lei M, Yang W and Wang L: Changes of circulating transforming growth factor-beta1 level during radiation therapy are correlated with the prognosis of locally advanced non-small cell lung cancer. J Thorac Oncol. 5:521–525. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Odar K, Boštjančič E, Gale N, Glavač D and Zidar N: Differential expression of microRNAs miR-21, miR-31, miR-203, miR-125a-5p and miR-125b and proteins PTEN and p63 in verrucous carcinoma of the head and neck. Histopathology. 61:257–265. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Cowden Dahl KD, Dahl R, Kruichak JN and Hudson LG: The epidermal growth factor receptor responsive miR-125a represses mesenchymal morphology in ovarian cancer cells. Neoplasia. 11:1208–1215. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Nishida N, Mimori K, Fabbri M, Yokobori T, Sudo T, Tanaka F, Shibata K, Ishii H, Doki Y and Mori M: MicroRNA-125a-5p is an independent prognostic factor in gastric cancer and inhibits the proliferation of human gastric cancer cells in combination with trastuzumab. Clin Cancer Res. 17:2725–2733. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Zhao X, Tang Y, Qu B, Cui H, Wang S, Wang L, Luo X, Huang X, Li J, Chen S and Shen N: MicroRNA-125a contributes to elevated inflammatory chemokine RANTES levels via targeting KLF13 in systemic lupus erythematosus. Arthritis Rheum. 62:3425–3435. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 18 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Quan HY, Yuan T and Hao JF: A microRNA‑125a variant, which affects its mature processing, increases the risk of radiation‑induced pneumonitis in patients with non‑small‑cell lung cancer. Mol Med Rep 18: 4079-4086, 2018
APA
Quan, H., Yuan, T., & Hao, J. (2018). A microRNA‑125a variant, which affects its mature processing, increases the risk of radiation‑induced pneumonitis in patients with non‑small‑cell lung cancer. Molecular Medicine Reports, 18, 4079-4086. https://doi.org/10.3892/mmr.2018.9406
MLA
Quan, H., Yuan, T., Hao, J."A microRNA‑125a variant, which affects its mature processing, increases the risk of radiation‑induced pneumonitis in patients with non‑small‑cell lung cancer". Molecular Medicine Reports 18.4 (2018): 4079-4086.
Chicago
Quan, H., Yuan, T., Hao, J."A microRNA‑125a variant, which affects its mature processing, increases the risk of radiation‑induced pneumonitis in patients with non‑small‑cell lung cancer". Molecular Medicine Reports 18, no. 4 (2018): 4079-4086. https://doi.org/10.3892/mmr.2018.9406