Open Access

Association of FGFR2 rs2981582, SIRT1 rs12778366, STAT3 rs744166 gene polymorphisms with pituitary adenoma

  • Authors:
    • Brigita Glebauskiene
    • Alvita Vilkeviciute
    • Rasa Liutkeviciene
    • Silvija Jakstiene
    • Loresa Kriauciuniene
    • Reda Zemaitiene
    • Dalia Zaliuniene
  • View Affiliations

  • Published online on: March 10, 2017     https://doi.org/10.3892/ol.2017.5840
  • Pages: 3087-3099
  • Copyright: © Glebauskiene 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

The aim of the present study was to determine the association between sirtuin 1 (SIRT1), fibroblast growth factor receptor 2 (FGFR2) and signal transducer and activator of transcription 3 (STAT3) polymorphisms, and pituitary adenoma (PA) development, invasiveness, hormonal activity and recurrence. The present study included 143 patients with a diagnosis of PA. The reference group involved 808 healthy subjects. The genotyping of SIRT1 rs12778366, FGFR2 rs2981582 and STAT3 rs744166 was performed using the quantitative polymerase chain reaction method. The SIRT1 rs12778366 polymorphism analysis in the overall group revealed differences in the genotype distribution between patients with PA and control group subjects. The rs12778366 T/C genotype was observed to be different in non‑invasive, non‑recurrent and inactive PA subgroups compared with the control group, while the C/C genotype was observed to be different in invasive, recurrent and active PA subgroups compared with the control group. STAT3 rs744166 polymorphism analysis in the overall group revealed differences in the genotype distribution between patients with PA and the control groups. The rs744166 G/G genotype was observed to be different in invasive, non‑recurrent and active PA subgroups compared with the control group, while the rs744166 A/A genotype was observed to be different in the active PA subgroup compared with the control group, and was also different in terms of invasiveness and recurrence in PA subgroups. The present study demonstrated that SIRT1 rs12778366 is associated with pituitary adenoma development while STAT3 rs744166 is associated with PA invasiveness, hormonal activity and recurrence.

Introduction

Pituitary adenomas (PAs), located in a bone cavity termed the sella turcica, are one of the most common types of intracranial neoplasms, with reported estimated prevalence rates ranging between 14.4 and 22.5% in pooled autopsy and radiological series, respectively (1). Although the majority of PAs are benign, it is not uncommon for them to grow large and extend locally into the surrounding structures, invading the sphenoid bone inferiorly, the cavernous sinus laterally (27,8) and/or compressing the optic chiasm, if the direction of expansion is suprasellar, thus resulting in neurological complications, including headache and visual impairment (917). Certain types of PA are extremely invasive and may cause extensive destruction of the skull base (18). Investigation of tumour invasiveness is required, as this affects the management and prognosis of PA (19). The aim of the present study was to identify possible genes involved in PA tumourigenesis, which may serve as potential diagnostic and prognostic molecular markers. The present study selected 3 genes, sirtuin 1 (SIRT1), fibroblast growth factor receptor 2 (FGFR2) and signal transducer and activator of transcription 3 (STAT3), which are associated with different types of cancer, but are connected in pathogenic processes (2023).

SIRT1 is a nicotinamide adenine dinucleotide-dependent histone deacetylase (HDAC) (24), which serves an important role in maintaining the balance between cell death and survival through targeting the Ku70-B-cell lymphoma-like protein 4 pathway (25), p53 (26,27) and forkhead box O3 (28), among others. A significant increase in the level of SIRT1 in hepatocellular carcinoma (29), breast cancer (30), prostate cancer (31), ovarian cancer (32), gastric cancer (33), colon cancer (34), glioblastoma (35) and lymphoma (36) was previously suggested to be associated with the development and invasion of these tumours. Furthermore, the rs12778366 polymorphism of the SIRT1 gene was found to be associated with breast cancer (37).

FGFR2 is a member of the FGFR family of tyrosine kinase receptors and participates in the process of tumourigenesis by inducing mitogenic and survival signals, and promoting invasiveness and angiogenesis (38). If cancer cells overexpress an FGFR with altered ligand-binding specificity, FGFs, secreted from neighbouring cells, stimulate the cancer cells, creating a paracrine loop (38). FGFR2 was previously revealed to be overexpressed in bladder (39) and lung cancer (40). Additionally, the importance of the FGFR2 rs2981582 gene polymorphism was investigated in breast (4150) and prostate cancer (51).

STAT3 is activated in tumour cells and numerous immune cells of the tumour microenvironment, and is associated with tumour cell proliferation, invasion and angiogenesis (5254). The effect of STAT3 was previously studied in the tumour development of colorectal adenocarcinoma (55), hepatocellular carcinoma (56), multiple myeloma (57), glioblastoma (58), prostate cancer (59), and head and neck cancer (60). The STAT3 rs744166 polymorphism was also evaluated in gastric (61,62), colon (63) and lung cancer (64).

These findings support the hypothesised role of SIRT1, FGFR2 and STAT3 as tumour promoters. However, an association between SIRT1, FGFR2 and STAT3 polymorphisms, and PA development, invasiveness, PA activity and recurrence has not yet been reported. The aim of the present study was to determine these associations.

Materials and methods

Patients and selection

Permission to undertake the present study was obtained from the Biomedical Research Ethics Committee of Lithuanian Health Sciences University (Kaunas, Lithuania). The study was conducted in the Departments of Ophthalmology and Neurosurgery, Lithuanian Health Sciences University Hospital (Kaunas, Lithuania).

The participants comprised of 143 patients with a diagnosis of PA. The reference group involved 808 healthy subjects. The reference group was created by taking into consideration the distribution of age and gender in the PA group. Therefore, the median patient age of the control group and the PA group did not differ significantly (P<0.05). Demographic data of the study subjects are presented in Table I.

Table I.

Demographic characteristics of patients with PA and reference group subjects.

Table I.

Demographic characteristics of patients with PA and reference group subjects.

GroupnMin/max/median age, yearsFemales, n (%)
PA14319/87/52.588 (65.67)
Control80820/90/58510 (63.12)
P-value0.793a0.882

a P-value for comparison of the median age between the PA and control groups. PA, pituitary adenoma; min, minimum; max, maximum.

The inclusion criteria were as follows: Determined and confirmed PA via magnetic resonance imaging (MRI); general good condition of the patient; consent of the patient to take part in the study; age ≥18 years; and no other brain tumours or tumours with other localizations.

All PAs were analysed based on MRI findings. The pre-operative MRI investigations were performed with 1.5T MRI scanners (Siemens MAGNETOM Avanto: Siemens AG, Munich, Germany; 1.5 T Philips ACHIEVA: Philips Healthcare, DA Best, The Netherlands) using a head coil and a standard pituitary scanning protocol, obtaining T1-weighted (T1W) sagittal and coronal and T2W/turbo spin echo coronal pre-contrast images, and T1W coronal and sagittal gadolinium-enhanced MR images with the intravenous agent gadodiamide (Omniscan; GE Healthcare Life Sciences, Chalfont, UK). The retrospective analysis of MRI data was conducted by an experienced radiologist. The suprasellar extension and sphenoid sinus invasion by PAs were classified according to Wilson-Hardy classification (Hardy classification, modified by Wilson) (19). The degree of suprasellar and parasellar extension was graded as stages A-E. The degree of sellar floor erosion was graded between I and IV. Grade III, localized sellar destruction, and grade IV, diffuse destruction, were considered to be invasive PAs. The Knosp classification system (4) was used to quantify invasion of the cavernous sinus, in which only grades 3 and 4 define true invasion of the tumour into the cavernous sinus: Grade 0, no cavernous sinus involvement; grades 1 and 2, the tumour pushes into the medial wall of the cavernous sinus, but does not go beyond a hypothetical line extending between the centres of the two segments of the internal carotid artery (grade 1) or it goes beyond such a line, but without passing a line tangential to the lateral margins of the artery itself (grade 2); grade 3, the tumour extends laterally to the internal carotid artery within the cavernous sinus; and grade 4, total encasement of the intracavernous carotid artery.

DNA extraction and genotyping

The DNA extraction and analysis of the gene polymorphisms of SIRT1 rs12778366, FGFR2 rs2981582 and STAT3 rs744166 were performed at the Laboratory of Ophthalmology at the Institute of Neuroscience of the Lithuanian University of Health Sciences (Kaunas, Lithuania). DNA was extracted from 200 µl venous blood (white blood cells) using a DNA purification kit based on the magnetic beads method (MagJET Genomic DNA kit; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to manufacturer's instructions.

The genotyping of SIRT1 rs12778366, FGFR2 rs2981582 and STAT3 rs744166 was performed using the quantitative polymerase chain reaction (qPCR) method with a Rotor-Gene Q Real-Time PCR Quantification system (Qiagen, Inc., Valencia, CA, USA). All 3 single-nucleotide polymorphisms were determined using TaqMan® Genotyping assays (Applied Biosystems; Thermo Fisher Scientific, Inc.), C_1340370_10 (rs12778366), C_2917302_10 (rs2981582) and C_3140282_10 (rs744166), according to the manufacturer's protocols.

The Allelic Discrimination program (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used during the qPCR. The assay was then continued following the manufacturer protocols. The Allelic Discrimination program was completed, and the genotyping results were received. The program determined the individual genotypes according to the fluorescence intensity rate from different detectors: Molecular marker labeled with VIC fluorescent dye was chosen for the X axis and a molecular marker labeled with FAM fluorescent dye was selected for the Y axis. These dy-labeled probes were included in the TaqMan® Genotyping assays.

Statistical analysis

Statistical analysis was performed using SPSS 20.0 software (IBM SPSS, Armonk, NY, USA). The data are presented as absolute numbers with percentages in brackets, and as median with minimum/maximum values. The frequencies of genotypes are presented as percentages.

Hardy-Weinberg analysis was performed to compare the observed and expected frequencies of rs12778366, rs2981582 and rs744166 using the χ2 test in all groups. The distribution of rs12778366, rs2981582 and rs744166 single-nucleotide polymorphisms (SNPs) in the PA and control groups was compared using the χ2 test or Fisher's exact test. Binomial logistic regression analysis was performed to estimate the impact of genotypes on PA development. Odds ratios (ORs) and 95% confidence intervals (CIs) are presented. Only statistically significant variables are presented in the tables. The selection of the most suitable genetic model was based on the Akaike Information Criterion (AIC), whereby the best genetic models were those with the lowest AIC values (65). P<0.05 was considered to indicate a statistically significant difference.

Results

Genotype distribution in the PA patients and the control group

The genotyping of SIRT1 rs12778366, FGFR2 rs2981582 and STAT3 rs744166 was performed in the PA group and the control group subjects (Table II).

Table II.

Frequency of single nucleotide polymorphisms in patients with PA and the control group.

Table II.

Frequency of single nucleotide polymorphisms in patients with PA and the control group.

Gene markerControl group, n (%)P-value HWEPA group, n (%)P-value HWEχ2P-value
SIRT1 rs12778366
  Genotype
  T/T647 (80.1)<0.01116 (81.1)<0.00191.139<0.001
  T/C141 (17.5)a 0 (0.0)a
  C/C20 (2.5)b 27 (18.9)b
  Total808 (100.0) 143 (100)
  Allele
  T1,435 (88.8) 232 (81.1)
  C181 (11.2) 54 (18.9)
FGFR2 rs2981582
  Genotype
  G/G336 (41.6)<0.00156 (39.2)<0.0013.5020.174
  G/A429 (53.1) 84 (58.7)
  A/A43 (5.3) 3 (2.1)
  Total808 (100.0) 143 (100.0)
  Allele
  G1,101 (68.1) 196 (68.53)
  A515 (31.9) 90 (31.47)
STAT3 rs744166
  Genotype
  G/G154 (19.1)c<0.00113 (9.1)c0.3548.8250.012
  G/A363 (44.9) 68 (47.6)
  A/A291 (36.0) 62 (43.4)
  Total808 (100.0) 143 (100.0)
  Allele
  G671 (41.5) 94 (32.9)
  A945 (58.5) 192 (67.1)

a SIRT1 rs1277836 T/C genotype was significantly less frequent (P<0.001) in the PA group compared with the control group.

b SIRT1 rs1277836 C/C genotype was significantly more frequent (P<0.001) in the PA group compared with the control group.

c STAT3 rs744166 G/G genotype was significantly less frequent (P=0.003) in the PA group compared with the control group. PA, pituitary adenoma; SIRT1, sirtuin 1, FGFR2, fibroblast growth factor receptor 2; STAT3, signal transducer and activator of transcription 3; HWE, Hardy-Weinberg equilibrium.

The distribution of analysed SIRT1 genotypes and allele frequencies in the control and PA groups did not match the Hardy-Weinberg equilibrium. The SIRT1 rs12778366 polymorphism analysis in the overall group revealed differences in the genotype distribution between patients with PA and control group subjects (P<0.001). The genotype T/C was significantly less frequent in the PA group compared with the healthy controls (0 vs. 17.5%; P<0.001) and the genotype C/C was significantly more frequent in the PA group compared with the healthy control group (18.9 vs. 2.5%, respectively; P<0.001) (Table II).

The distribution of analysed FGFR2 genotypes and allele frequencies in the control and PA groups did not match the Hardy-Weinberg equilibrium. Statistical analysis did not reveal significant genotype (G/G, G/A and A/A) distribution differences between the control and PA groups: 41.6 vs. 39.2%, 53.1 vs. 58.7%, and 5.3 vs. 2.1%, respectively (P=0.174) (Table II).

The distribution of the analysed STAT3 rs744166 genotypes and allele frequencies did not match the Hardy-Weinberg equilibrium in the control group, but it did in the group of patients with PA. STAT3 rs744166 polymorphism analysis in the overall group revealed differences in the genotype distribution between the patients with PA and the control group (P=0.012). The genotype G/G was less frequent in the PA group compared with the healthy controls (9.1 vs. 19.1%, respectively; P=0.003) (Table II).

Genotype distribution in the PA patients and the control group by gender

All 3 SNPs were analysed in the PA and control groups according to gender (Table III). SIRT1 rs12778366 polymorphism analysis did not revealed any statistically significant differences between females and males with PA in genotype (T/T, T/C and C/C) distribution (80.7, 0 and 19.3% vs. 81.8, 0 and 18.2%, respectively; Table III). Comparing SIRT1 rs12778366 genotype distribution in healthy females and females with PA, significant differences were revealed. The T/C genotype was less frequently present in females with PA compared with the healthy control females (0 vs. 17.5%, respectively; P<0.001) and C/C was more frequent in PA females compared with healthy females (19.3 vs. 2.7%, respectively; P<0.001). The T/T genotype did not exhibit any significant differences when healthy females and females with PA were compared. When analysing genotype distribution in males, T/C genotype distribution showed statistically significant difference between males with PA and healthy males (0 vs. 17.4%, respectively; P<0.001) and the C/C genotype was more frequent in males with PA compared with the control group (18.2 vs. 2.0%, respectively; P<0.001) (Table III).

Table III.

Frequency of single nucleotide polymorphisms in patients with PA and control group according to gender.

Table III.

Frequency of single nucleotide polymorphisms in patients with PA and control group according to gender.

Control group, n (%)PA group, n (%)


Gene markerFemalesMalesP-value HWEP-valueFemalesMalesP-value HWEP-value
SIRT1 rs12778366
  Genotype
  T/T407 (79.8)240 (80.5)0.8110.80171 (80.7)45 (81.8)0.8660.866
  T/C89 (17.5)a52 (17.4)b 1.0000 (0.0)a0 (0.0)b 1.00
  C/C14 (2.7)c6 (2.0)d 0.51817 (19.3)c10 (18.2)d 0.866
  Total510 (100.0)298 (100.0) 88 (100.0)55 (100.0)
  Allele
  T903 (88.5)532 (89.3) 142 (80.7)90 (81.8)
  C117 (11.5)64 (10.7) 34 (19.3)20 (18.2)
FGFR2 rs2981582
  Genotype
  G/G218 (42.7)118 (39.6)0.6750.38139 (44.3)17 (30.9)0.1970.117
  G/A265 (52.0)164 (55.0) 0.39848 (54.5)36 (65.5) 0.224
  A/A27 (5.3)16 (5.4) 1.01 (1.1)2 (3.6) 0.559
  Total510 (100)298 (100) 88 (100)55 (100)
  Allele
  G701 (68.7)400 (67.1) 126 (71.6)70 (63.6)
  A319 (31.3)196 (32.9) 50 (28.4)40 (36.4)
STAT3 rs744166
  Genotype
  G/G104 (20.4)e50 (16.8)0.3780.2077 (8.0)e6 (10.9)0.8150.563
  G/A229 (44.9)134 (45.0) 0.98643 (48.9)25 (45.5) 0.733
  A/A177 (34.7)114 (38.3) 0.31238 (43.2)24 (43.6) 1.00
  Total510 (100)298 (100) 88 (100)55 (100)
  Allele
  G437 (42.84)234 (39.3) 57 (32.4)37 (33.6)
  A583 (57.2)362 (60.7) 119 (67.6)73 (66.4)

a SIRT1 rs12778366 T/C genotype is significantly less frequent (P<0.001) in PA females compared with control females.

b SIRT1 rs12778366 T/C genotype is significantly less frequent (P<0.001) in PA males compared with control males.

c SIRT1 rs12778366 C/C genotype is significantly more frequent (P<0.001) in PA females compared with control females.

d SIRT1 rs12778366 C/C genotype is significantly more frequent (P<0.001) in PA males compared with control males.

e STAT3 rs744166 G/G genotype is significantly less frequent (P=0.004) in PA females compared with control females. PA, pituitary adenoma; SIRT1, sirtuin 1; FGFR2, fibroblast growth factor receptor 2; STAT3, signal transducer and activator of transcription 3; HWE, Hardy-Weinberg equilibrium.

FGFR2 rs2981582 polymorphism analysis by gender was performed, but it did not reveal any genotype distribution differences between females and males.

STAT3 rs744166 polymorphism analysis did not reveal any significant differences between females and males with PA in the genotype (G/G, G/A and A/A) distribution (8.0, 48.9 and 43.2% vs. 10.9, 45.5 and 43.6%, respectively; Table III) either. When comparing STAT3 genotype distribution between healthy females and females with PA, there were significant differences. The STAT3 rs744166 G/G genotype was less frequently present in PA females compared with healthy control females (8.0 vs. 20.4%, respectively; P=0.004). The STAT3 rs744166 G/A and A/A genotype distribution did not exhibit any significant differences when healthy females and females with PA were compared. STAT3 rs744166 analysis between male groups did not reveal any statistically significant differences (Table III).

Binomial logistic regression analysis of the patients with PA and the control group

Binomial logistic regression analysis of the patients with PA and the control group was performed (Table IV). SIRT1 rs12778366 analysis revealed that there were significant variables in the co-dominant (OR=7.530; 95% CI: 4.087–13.873; P<0.001), recessive (OR=9.171; 95% CI: 4.982–16.881; P<0.001) and additive (OR=1.584: 95 % CI: 1.187–2.115; P=0.002) models of the patients with PA and the control group (Table IV).

Table IV.

Binomial logistic regression analysis in patients with pituitary adenoma and the control group.

Table IV.

Binomial logistic regression analysis in patients with pituitary adenoma and the control group.

GeneModelGenotypeOR (95% CI)P-valueAIC
SIRT1 rs12778366Co-dominantT/T1.000 720.516
T/C0 (0.000)0.995
C/C7.530 (4.087–13.087)<0.001
RecessiveT/T+T/C1.000 780.895
C/C9.171 (4.982–16.982)<0.001
Additive1.584 (1.187–2.187)0.002780.214
STAT3 rs744166Co-dominantA/A1.000 801.223
G/G0.879 (0.603–1.603)0.504
G/G0.396 (0.211–0.211)0.004
RecessiveA/A+G/A1.000 799.670
G/G0.425 (0.234–0.234)0.005
Additive0.702 (0.541–0.541)0.008801.881

[i] OR, odds ratio; CI, confidence interval; AIC, Akaike Information Criterion; SIRT1, sirtuin 1; STAT3, signal transducer and activator of transcription 3.

FGFR2 rs2981582 analysis did not reveal any significant variables.

STAT3 rs744166 analysis revealed that there were significant variables in the co-dominant (OR=0.396; 95% CI: 0.211–0.743; P=0.004), recessive (OR=0.425; 95% CI: 0.234–0.771; P=0.005) and additive (OR=0.702; 95% CI: 0.541–0.911; P=0.008) models of the patients with PA and the control group (Table IV).

Binomial logistic regression analysis in the patients with PA and the control group according to gender was performed (Table V). In the SIRT1 rs12778366 analysis there were statistically significant variables in the co-dominant (P<0.001) and recessive (P<0.001) models of males. The co-dominant (P<0.001), recessive (P<0.001) and additive (P=0.013) variables were also significant in females.

Table V.

Binomial logistic regression analysis in patients with pituitary adenoma and control subjects according to gender.

Table V.

Binomial logistic regression analysis in patients with pituitary adenoma and control subjects according to gender.

GeneGenderModelGenotypeOR (95% CI)P-valueAIC
SIRT1 rs12778366MaleCo-dominantT/T1.000 275.783
T/C0.000 (0.000)0.997
C/C8.889 (3.076–25.076)<0.001
RecessiveT/T+T/C1.000 290.082
C/C10.815 (3.748–31.748)<0.001
FemaleCo-dominantT/T1.000 450.358
T/C0.000 (0.000)0.996
C/C6.961 (3.285–14.285)<0.001
RecessiveT/T+T/C1.000 474.428
C/C8.483 (4.008–17.008)<0.001
Additive1.580 (1.100–2.100)0.013497.979
STAT3 rs744166FemaleCo-dominantA/A1.000 496.249
G/A0.875 (0.542–1.542)0.583
G/G0.314 (0.135–0.135)0.007
RecessiveA/A+G/A1.000 494.549
G/G0.337 (0.151–0.151)0.008
Additive0.654 (0.469–0.469)0.012497.077

[i] OR, odds ratio; CI, confidence interval; AIC, Akaike Information Criterion; SIRT1, sirtuin 1; STAT3, signal transducer and activator of transcription 3.

Binomial logistic regression analysis of FGFR2 rs2981582 in the patients with PA and in the control group according to gender was performed, but no significant variables were observed.

However, binomial logistic regression analysis of STAT3 rs744166 in the patients with PA and in the control group according to gender showed statistically significant variables only in the co-dominant (P=0.007), recessive (P=0.008) and additive (P=0.012) models of females (Table V).

Genotype distribution in the control group and the PA patients by different PA subgroups

Analysis of SIRT1 rs1277836, FGFR2 rs2981582 and STAT3 rs744166 polymorphisms was performed by different PA subgroups (Tables VIVIII).

Table VI.

Frequency of SNPs in patients with PA and in control group according to PA invasiveness.

Table VI.

Frequency of SNPs in patients with PA and in control group according to PA invasiveness.

Gene markerControl group, n (%)P-value HWENon invasive PA group, n (%)P-value HWEInvasive PA group, n (%)P-value HWE
SIRT1 rs12778366
  Genotype
  T/T647 (80.1)<0.00147 (81.0)<0.00169 (81.2)<0.001
  T/C141 (17.5)a,b 0 (0.0)a 0 (0.0)b
  C/C20 (2.5)c,d 11 (19.0)c 16 (18.8)d
  Total808 (100.0) 58 (100.0) 85 (100.0)
  Allele
  T1,435 (88.8) 94 (81.0) 138 (81.2)
  C181 (11.2) 22 (19.0) 32 (18.8)
FGFR2 rs2981582
  Genotype
  G/G336 (41.6)e<0.00116 (27.6)e,f<0.00140 (47.1)f0.043
  G/A429 (53.1)g 42 (72.4)g,h 42 (49.4)h
  A/A43 (5.3) 0 (0.0) 3 (3.5)
  Total808 (100.0) 58 (100.0) 85 (100.0)
  Allele
  G1,101 (68.1) 74 (63.8) 122 (71.8)
  A515 (31.9) 42 (36.2) 48 (28.2)
STAT3 rs744166
  Genotype
  G/G154 (19.1)i<0.0019 (15.5)j0.3134 (4.7)i,j0.031
  G/A363 (44.9) 23 (39.7) 45 (52.9)
  A/A291 (36.0) 26 (44.8) 36 (42.4)
  Total808 (100.0) 58 (100.0) 85 (100.0)
  Allele
  G671 (41.5) 41 (35.3) 53 (31.2)
  A945 (58.5) 75 (64.7) 117 (68.8)

a SIRT1 rs12778366 T/C genotype is significantly less frequent (P=0.021) in non-invasive PA compared with the control group.

b SIRT1 rs12778366 T/C genotype is significantly less frequent (P<0.001) in invasive PA compared with the control group.

c SIRT1 rs12778366 C/C genotype is significantly more frequent (P=0.041) in non-invasive PA compared with the control group.

d SIRT1 rs12778366 C/C genotype is significantly less frequent (P<0.001) in invasive PA compared with the control group.

e FGFR2 rs2981582 G/G genotype is significantly less frequent (P=0.038) in non-invasive PA compared with the control group.

f FGFR2 rs2981582 G/G genotype is significantly more frequent (P=0.024) in invasive PA compared with the non-invasive PA group.

g FGFR2 rs2981582 G/A genotype is significantly more frequent (P=0.004) in non-invasive PA compared with the control group.

h FGFR2 rs2981582 G/A genotype is significantly less frequent (P=0.009) in invasive PA compared with the non-invasive PA group.

i STAT3 rs744166 G/G genotype is significantly less frequent (P<0.001) in invasive PA compared with the control group.

j STAT3 rs744166 G/G genotype is significantly less frequent (P=0.038) in invasive PA compared with the non-invasive PA group. PA, pituitary adenoma; SIRT1, sirtuin 1, FGFR2, fibroblast growth factor receptor 2; STAT3, signal transducer and activator of transcription 3; HWE, Hardy-Weinberg equilibrium.

Table VIII.

Frequency of single nucleotide polymorphisms in patients with PA and in the control group according to PA activity.

Table VIII.

Frequency of single nucleotide polymorphisms in patients with PA and in the control group according to PA activity.

Gene markerControl group, n (%)P-value HWEInactive PA group, n (%)P-value HWEActive PA group, n (%)P-value HWE
SIRT1 rs12778366
  Genotype
  T/T647 (80.1)<0.00148 (76.2)   0.00768 (85.0)0.044
  T/C141 (17.5)a,b 0 (0.0)a 0 (0.0)b
  C/C20 (2.5)c,d 15 (23.8)d 12 (15.0)c
  Total808 (100.0) 63 (100.0) 80 (100.0)
  Allele
  T1,435 (88.8) 96 (76.2) 136 (85.0)
  C181 (11.2) 30 (23.8) 24 (15.0)
FGFR2 rs2981582
  Genotype
  G/G336 (41.6)<0.00125 (39.7)<0.00131 (38.8)0.005
  G/A429 (53.1) 38 (60.3) 46 (57.5)
  A/A43 (5.3) 0 (0.0) 3 (3.8)
  Total808 (100.0) 63 (100.0) 80 (100.0)
  Allele
  G1,101 (68.1) 88 (69.8) 108 (67.5)
  A515 (31.9) 38 (30.2) 52 (32.5)
STAT3 rs744166
  Genotype
  G/G154 (19.1)e<0.0016 (9.5)0.1937 (8.8)e0.915
  G/A363 (44.9) 34 (54.0) 34 (42.5)
  A/A291 (36.0)f 23 (36.5) 39 (48.8)f
  Total808 (100.0) 63 (100.0) 80 (100.0)
  Allele
  G671 (41.5) 46 (36.5) 48 (30.0)
  A945 (58.5) 80 (63.5) 112 (70.0)

a SIRT1 rs12778366 T/C genotype is significantly less frequent (P<0.001) in inactive PA compared with the control group

b SIRT1 rs12778366 T/C genotype is significantly less frequent (P<0.001) in active PA compared with the control group.

c SIRT1 rs12778366 C/C genotype is significantly less frequent (P<0.001) in active PA compared with the control group.

d SIRT1 rs12778366 C/C genotype is significantly more frequent (P<0.001) in inactive PA compared with the control group.

e FGFR2 rs2981582 G/G genotype is significantly less frequent (P=0.022) in active PA compared with the control group.

f FGFR2 rs2981582 A/A genotype is significantly more frequent (P=0.029) in active PA compared with the control group. PA, pituitary adenoma; SIRT1, sirtuin 1, FGFR2, fibroblast growth factor receptor 2; STAT3, signal transducer and activator of transcription 3; HWE, Hardy-Weinberg equilibrium.

The SIRT1 rs12778366 T/C genotype was less frequently observed in non-invasive, non-recurrent and inactive PA subgroups compared with healthy controls (0 vs. 17.5%, P=0.021; 0 vs. 17.5%, P<0.001; 0 vs. 17.5%, P<0.001, respectively). However, no differences were observed between non-invasive and invasive, non-recurrent and recurrent, and inactive and active PA subgroups (Tables VIVIII).

Additional analysis revealed that the C/C genotype was more frequent in invasive, recurrent and active PA subgroups compared with the healthy controls (18.8 vs. 2.5%, P=0.041; 19.4 vs. 2.5%, P=0.047; 15.0 vs. 2.5%, P<0.001, respectively;Tables VI, VII and VIII).

Table VII.

Frequency of single nucleotide polymorphisms in patients with PA and in the control group according to PA recurrences.

Table VII.

Frequency of single nucleotide polymorphisms in patients with PA and in the control group according to PA recurrences.

Gene markerControl group, n (%)P-value HWENon-recurrent PA group, n (%)P-value HWERecurrent PA group, n (%)P-value HWE
SIRT1 rs12778366
  Genotype
  T/T647 (80.1)<0.00191 (81.3)<0.00125 (80.6)<0.001
  T/C141 (17.5)a, b 0 (0.0)a 0 (0.0)b
  C/C20 (2.5)c,d 21 (18.8)c 6 (19.4)d
  Total808 (100) 112 (100.0) 31 (100.0)
  Allele
  T1,435 (88.8) 182 (81.3) 50 (80.6)
  C181 (11.2) 42 (18.8) 12 (19.4)
FGFR2 rs2981582
  Genotype
  G/G336 (41.6)<0.00144 (39.3)<0.00112 (38.7)0.067
  G/A429 (53.1) 66 (58.9) 18 (58.1)
  A/A43 (5.3) 2 (1.8) 1 (3.2)
  Total808 (100.0) 112 (100.0) 31 (100.0)
  Allele
  G1,101 (68.1) 154 (68.8) 42 (67.7)
  A515 (31.9) 70 (31.3) 20 (32.3)
STAT3 rs744166
  Genotype
  G/G154 (19.1)e<0.0017 (6.3)e,f0.0836 (19.4)f0.305
  G/A363 (44.9) 56 (50.0) 12 (38.7)
  A/A291 (36.0) 49 (43.8) 13 (41.9)
  Total808 (100.0) 112 (100.0) 31 (100.0)
  Allele
  G671 (41.5) 70 (31.3) 24 (38.7)
  A945 (58.5) 154 (68.8) 38 (61.3)

a SIRT1 rs12778366 T/C genotype is significantly less frequent (P<0.001) in non-recurrent PA compared with the control group.

b SIRT1 rs12778366 T/C genotype is significantly less frequent (bP=0.005) in recurrent PA compared with the control group.

c SIRT1 rs12778366 C/C genotype is significantly more frequent (P<0.001) in non-recurrent PA compared with the control group.

d SIRT1 rs12778366 C/C genotype is significantly more frequent (P=0.047) in recurrent PA compared with the control group.

e STAT3 rs744166 G/G genotype is significantly less frequent (P<0.001) in non-recurrent PA compared with the control group.

f FGFR2 rs2981582 G/G genotype is statistically more frequent (P=0.036) in recurrent PA compared with non-recurrent PA. PA, pituitary adenoma; SIRT1, sirtuin 1, FGFR2, fibroblast growth factor receptor 2; STAT3, signal transducer and activator of transcription 3; HWE, Hardy-Weinberg equilibrium.

The FGFR2 rs2981582 G/G genotype was less frequently observed in the non-invasive PA subgroup compared with the healthy controls (27.6 vs. 41.6%, respectively; P=0.038), but the G/A genotype was more frequently observed in the non-invasive PA subgroup compared with the control group (72.4 vs. 53.1%, respectively; P=0.004) and the invasive PA subgroup (72.4 vs. 49.4% respectively; P=0.009) (Table VI).

Statistical analysis was performed to evaluate the FGFR2 rs2981582 association with PA activity and recurrence (Tables VII and VIII). This analysis did not reveal any association between SNP and active or non-active PA, and PA without recurrence or with recurrence.

The STAT3 rs744166 G/G genotype was less frequently observed in invasive, non-recurrent and active PA subgroups compared with healthy controls (4.7 vs. 19.1%, P<0.001; 6.2 vs. 19.1%, P<0.001; 8.8 vs. 19.1%, P=0.022, respectively).

The STAT3 rs744166 A/A genotype was more frequent in the active PA subgroup compared with the control group (48.8 vs. 36.0%, respectively; P=0.029). There were differences between non-invasive and invasive, non-recurrent and recurrent PA subgroups as well, with the exception of comparing inactive and active PA. The STAT3 rs744166 G/G genotype was more frequent in non-invasive PA compared with invasive PA (15.5 vs. 4.7%, respectively; P=0.038) and in recurrent PA group comparing to non-recurrent PA (19.4 vs. 6.2%, respectively; P=0.036) (Tables VIVIII).

Binomial logistic regression analysis of the control group and the PA patients by different PA subgroups

Binomial logistic regression analysis in the non-invasive PA, invasive PA and control groups was performed (Table IX). Analysing the SIRT1 polymorphism in non-invasive PA group and control group this analysis showed that the co-dominant (P<0.001), recessive (P<0.001) and additive (P=0.025) variables were significant. Binomial logistic regression analysis in the patients with invasive PA and the control group revealed significance of the same co-dominant (P<0.001), recessive (P<0.001) and additive (P=0.010) variables (Table IX).

Table IX.

Binomial logistic regression analysis in non-invasive and invasive PA, and the control group.

Table IX.

Binomial logistic regression analysis in non-invasive and invasive PA, and the control group.

GenePA subgroupModelGenotypeOR (95% CI)P-valueAIC
SIRT1 rs12778366Non-invasiveCo-dominantT/T1.000 390.145
T/C0.000 (0.000)0.996
C/C7.571 (3.426–16.426)<0.001
RecessiveT/T+T/C1.000 406.092
C/C9.221 (4.175–20.175)<0.001
Additive1.649 (1.065–2.065)0.025425.148
InvasiveCo-dominantT/T1.000 509.448
T/C0.000 (0.000)0.996
C/C7.501 (3.715–15.715)<0.001
RecessiveT/T+T/C1.000 533.418
C/C9.136 (4.528–18.528)<0.001
Additive1.616 (1.120–2.120)   0.010459.515
FGFR2 rs2981582Non-invasiveCo-dominantG/G1.000 419.170
G/A2.056 (1.136–3.136)0.017
A/A0.000 (0.000)0.998
DominantG/G1.000 425.028
G/A+A/A1.869 (1.033–3.033)0.039
Over-dominantG/G+A/A1.000 420.961
G/A2.319 (1.283–4.283)0.005
STAT3 rs744166InvasiveCo-dominantA/A1.000 553.313
G/A1.002 (0.630–1.630)0.993
G/G0.210 (0.073–0.073)0.004
RecessiveA/A+G/A1.000 551.313
G/G0.210 (0.076–0.076)0.003
Additive0.651 (0.467–0.467)0.011558.771

[i] PA, pituitary adenoma; SIRT1, sirtuin 1; FGFR2, fibroblast growth factor receptor 2; STAT3, signal transducer and activator of transcription 3; OR, odds ratio; CI, confidence interval; AIC, Akaike Information Criterion.

Binomial logistic regression analysis of FGFR2 rs2981582 in the non-invasive PA and control groups showed that the co-dominant (P=0.017), dominant (P=0.039) and over-dominant (P=0.005) variables were significant, but this analysis in the patients with invasive PA and the control group did not reveal any significance of these models.

Binomial logistic regression analysis of STAT3 rs744166 was also performed (Table IX). The analysis showed that the co-dominant (P=0.004), recessive (P=0.003) and additive (P=0.011) variables were statistically significant only in the invasive PA and control groups (Table IX).

Binomial logistic regression analysis in the inactive PA and control groups, and in the active PA and control groups, was performed for all 3 SNPs (Table X).

Table X.

Binomial logistic regression analysis in inactive and active PA, and control groups.

Table X.

Binomial logistic regression analysis in inactive and active PA, and control groups.

GenePA subgroupModelGenotypeOR (95% CI)P-valueAIC
SIRT1 rs12778366InactiveCo-dominantT/T1.000 402.990
T/C0.000 (0.000)0.996
C/C10.109 (4.868–20.868)<0.001
RecessiveT/T+T/C1.000 419.306
C/C12.312 (5.933–25.933)<0.001
Additive2.045 (1.388–3.388)<0.001444.842
ActiveCo-dominantT/T1.000 497.635
T/C0.000 (0.000)0.996
C/C5.709 (2.675–12.675)<0.001
RecessiveT/T+T/C1.000 521.245
C/C6.953 (3.260–14.260)<0.001
STAT3 rs744166ActiveCo-dominantA/A1.000 535.474
G/A0.669 (0.430–1.430)0.148
G/G0.339 (0.148-.0776)0.010
DominantA/A1.000 536.765
G/A+G/G0.592 (0.373–0.373)0.026
RecessiveA/A+G/A1.000 535.576
G/G0.407 (0.184–0.184)0.027
Additive0.622 (0.442–0.442)0.007533.914

[i] PA, pituitary adenoma; SIRT1, sirtuin 1; STAT3, signal transducer and activator of transcription 3; OR, odds ratio; CI, confidence interval; AIC, Akaike Information Criterion.

Inactive PA group analysis of SIRT1 rs12778366 showed that the co-dominant (P<0.001), recessive (P<0.001) and additive (P<0.001) variables were significant. The analysis of the active PA group revealed significance in the co-dominant (P<0.001) and recessive (P<0.001) models (Table X).

Binomial logistic regression analysis of FGFR2 rs2981582 was performed in the inactive PA, active PA and control groups, but this analysis did not reveal significance in these models.

STAT3 rs744166 analysis in the inactive PA group showed that there were no significant variables. Analysing the active PA group, the present study revealed significance in the co-dominant (P=0.010), dominant (P=0.026), recessive (P=0.027) and additive (P=0.007) models (Table X).

Additional binomial logistic regression analysis of SNPs was performed in non-recurrence, recurrence and control groups. The analysis in the non-recurrent PA and control groups showed that the co-dominant (P<0.001), recessive (P<0.001) and additive (P=0.005) variables were statistically significant (Table XI). In the analysis of PA with recurrence, the co-dominant (P<0.001) and recessive (P<0.001) variables were also significant.

Table XI.

Binomial logistic regression analysis in non-recurrent and recurrent PA and control groups.

Table XI.

Binomial logistic regression analysis in non-recurrent and recurrent PA and control groups.

GenePA subgroupModelGenotypeOR (95% CI)P-valueAIC
SIRT1 rs12778366Non-recurrentCo-dominantT/T1.000 614.042
T/C0.000 (0.000)0.995
C/C7.465 (3.896–14.896)<0.001
RecessiveT/T+T/C1.000 645.812
C/C9.092 (4.748–17.748)<0.001
Additive1.592 (1.153–2.153)0.005678.223
RecurrentCo-dominantT/T1.000 247.718
T/C0.000 (0.000)0.996
C/C7.764 (2.868–21.868)<0.001
RecessiveT/T+T/C1.000 255.407
C/C9.456 (3.495–25.495)<0.001
STAT3 rs744166Non-recurrentCo-dominantA/A1.000 673.559
G/A0.916 (0.606–1.606)0.678
G/G0.270 (0.119–0.119)0.002
RecessiveA/A+G/A1.000 671.731
G/G0.283 (0.129–0.129)0.002
Additive0.653 (0.487–0.487)0.005677.047

[i] PA, pituitary adenoma; SIRT1, sirtuin 1; STAT3, signal transducer and activator of transcription 3; OR, odds ratio; CI, confidence interval; Akaike Information Criterion.

FGFR2 rs2981582 polymorphism analysis did not show any statistical significance.

Binomial logistic regression analysis of STAT3 rs744166 in the non-recurrent and recurrent PA groups, and the control group was performed. This revealed that in the non-recurrent PA and control groups, the co-dominant (P=0.002), recessive (P=0.002) and additive (P=0.005) variables were significant (Table XI). In the analysis of PA with recurrence there were no statistically significant variables.

Discussion

The impact of SIRT1, FGFR2 and STAT3 gene polymorphisms on the development of various tumours has been analysed in numerous studies (38,4252,60,6365), but no studies have investigated the associations with PA development, invasiveness, activity and recurrence.

A study conducted by Rizk et al (37) investigated SIRT1 gene single nucleotide polymorphism rs12778366 in patients with breast cancer, revealing that the SIRT1 rs12778366 T/T genotypes were more frequent, exhibited higher SIRT1 levels than the C/C and C/T genotypes, and were associated with histological grade and lymph node status. The T allele frequency was higher in patients with breast cancer compared with that in normal subjects.

The present study was the first to assess the association between SIRT1 rs12778366 and PA. It was found that the T/C genotype was less frequent in the PA group compared with the healthy controls (0 vs. 17.5%, respectively; P<0.001) and that the C/C genotype was more frequent in the PA group compared with the healthy control group (18.9 vs. 2.5%, P<0.001).

Numerous studies have investigated the FGFR2 rs2981582 polymorphism in breast cancer patients, and have provided controversial data on the impact of this polymorphism on tumour development. Chen et al (43) revealed that the G/A and A/A genotypes of FGFR2 rs2981582 were associated with lower mammographic density and a reduced risk of breast cancer, and Butt et al (42) revealed a statistically significant association between the FGFR2 rs2981582 A/A genotype and breast cancer risk. Shan et al (66) also revealed that patients with the A/A genotype of FGFR2 rs2981582 exhibited an increased risk of breast cancer, while Ledwoń et al (47) revealed that the rs2981582 SNP showed significant association with the familial and sporadic types of breast cancer. On the basis of these findings, the present study aimed to examine whether the polymorphism in the FGFR2 promoter may affect the risk of PA development, activity, recurrence or invasiveness. No differences in genotype (G/G, G/A and A/A) distribution were observed between the control and PA groups (41.6 vs. 39.2%, 53.1 vs. 58.7%, and 5.3 vs. 2.1%, respectively; P=0.174). No significant differences were observed between genotype distribution according to gender, PA activity, invasiveness or recurrence.

Several studies have analysed the STAT3 rs744166 polymorphism in association with various types of tumour, but none have investigated the association between STAT3 rs744166 and PA. Rocha et al (61) reported that the rs744166 polymorphic G allele was associated with gastric cancer, and a significantly decreased risk of non-small cell lung cancer was observed for carriers of STAT3 rs744166 in a study by Jiang et al (64). The present study demonstrated the differences in the distribution of the STAT3 rs744166 polymorphism between patients with PA and control group subjects (P=0.012). The G/G genotype was less frequent in the PA group compared with the healthy controls (9.1 vs. 19.1%, respectively; P=0.003). Analysis in different PA subgroups showed that the STAT3 rs744166 G/G genotype was more frequent in non-invasive PA compared with invasive PA (15.5 vs. 4.7%; P=0.038) and in recurrent PA compared with the non-recurrent PA (19.4 vs. 6.2%, respectively; P=0.036).

Overall, the present study demonstrated that the SNPs SIRT1 rs12778366 and STAT3 require replication in future larger studies, particularly with increased sample sizes to confirm the association of SIRT1 and STAT3 in patients with PA.

Acknowledgements

The present study received funding from the Research Council of Lithuania (grant no. MIP-008/2014).

References

1 

Ezzat S, Asa SL, Couldwell WT, Barr CE, Dodge WE, Vance ML and McCutcheon IE: The prevalence of pituitary adenomas: A systematic review. Cancer. 101:613–619. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Colao A, Di Somma C, Pivonello R, Faggiano A, Lombardi G and Savastano S: Medical therapy for clinically non-functioning pituitary adenomas. Endocr Relat Cancer. 15:905–915. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Kasputytė R, Slatkevičienė G, Liutkevičienė R, Glebauskienė B, Bernotas G and Tamašauskas A: Changes of visual functions in patients with pituitary adenoma. Medicina (Kaunas). 49:132–137. 2013.PubMed/NCBI

4 

Knosp E, Steiner E, Kitz K and Matula C: Pituitary adenomas with invasion of the cavernous sinus space: A magnetic resonance imaging classification compared with surgical findings. Neurosurgery. 33:610–618. 1993. View Article : Google Scholar : PubMed/NCBI

5 

Ferrante E, Ferraroni M, Castrignanò T, Menicatti L, Anagni M, Reimondo G, Del Monte P, Bernasconi D, Loli P, Faustini-Fustini M, et al: Non-functioning pituitary adenoma database: A useful resource to improve the clinical management of pituitary tumours. Eur J Endocrinol. 155:823–829. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Ahmadi J, North CM, Segall HD, Zee CS and Weiss MH: Cavernous sinus invasion by pituitary adenomas. Am J Neuroradiol. 6:893–898. 1985.

7 

Falbusch R and Buchfejder M: Transsphenoidal surgery of parasellar pituitary adenomas. Acta Neurochir. 92:93–99. 1988. View Article : Google Scholar : PubMed/NCBI

8 

Scheithauer BW, Kovacs KT, Laws ER Jr and Randall RV: Pathology of invasive pituitary tumors with special reference to functional classification. J Neurosurg. 65:733–744. 1986. View Article : Google Scholar : PubMed/NCBI

9 

Moon CH, Hwang SC, Kim BT, Ohn YH and Park TK: Visual prognostic value of optical coherence tomography and photopic negative response in chiasmal compression. Invest Ophthalmol Vis Sci. 52:8527–8533. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Thomas R, Shenoy K, Seshadri MS, Muliyil J, Rao A and Paul P: Visual field defects in non-functioning pituitary adenomas. Indian J Ophthalmol. 50:127–130. 2002.PubMed/NCBI

11 

Trautmann JC and Laws ER Jr: Visual status after transphenoidal surgery at the Mayo Clinic 1971–1982. Am J Ophthalmol. 96:200–208. 1983. View Article : Google Scholar : PubMed/NCBI

12 

Kaur A, Banerji D, Kumar D and Sharma K: Visual status in suprasellar pituitary tumours. Indian J Ophthalmol. 43:131–134. 1995.PubMed/NCBI

13 

Mortini P, Losa M, Barzaghi R, Boari N and Giovanelli M: Results of transsphenoidal surgery in a large series of patients with pituitary adenoma. Neurosurgery. 56:1222–1233. 2005. View Article : Google Scholar : PubMed/NCBI

14 

el-Azouzi M, Black PM, Candia G, Zervas NT and Panagopoulos KP: Transsphenoidal surgery for visual loss in patients with pituitary adenomas. Neurol Res. 12:23–25. 1990. View Article : Google Scholar : PubMed/NCBI

15 

Marazuela M, Astigarraga B, Vicente A, Estrada J, Cuerda C, Garcia-Uria J and Lucas T: Recovery of visual and endocrine function following transsphenoidal surgery of large nonfunctioning pituitary adenomas. J Endocrinol Invest. 17:703–707. 1994. View Article : Google Scholar : PubMed/NCBI

16 

Gondim JA, de Almeida JP, de Albuquerque LA, Schops M, Gomes E and Ferraz T: Headache associated with pituitary tumors. J Headache Pain. 10:15–20. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Levy MJ, Jäger H, Powell M, Matharu MS, Meeran K and Goadsby PJ: Pituitary volume and headache: Size is not everything. Arch Neurol. 61:721–725. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Stevens JM, Valentine AR and Kendall BE: Computed cranial and spinal imagingA practical introduction. Lippincott Williams & Wilkins; Philadelphia, PA: pp. 154–156. 1988

19 

Wilson CB: Neurosurgical management of large and invasive pituitary tumorsClinical management of pituitary disorders. Tindall GT and Collins WF: Raven; New York, NY: pp. 335–342. 1979

20 

Luo JC, Peng YL, Chen TS, Huo TI, Hou MC, Huang HC, Lin HC and Lee FY: Clopidogrel inhibits angiogenesis of gastric ulcer healing via down regulation of vascular endothelial growth factor receptor 2. J Formos Med Assoc. 115:764–772. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Lu J, Zhang L, Chen X, Lu Q, Yang Y, Liu J and Ma X: SIRT1 counteracted the activation of STAT3 and NF-κB to repress the gastric cancer growth. Int J Clin Exp Med. 7:5050–5058. 2014.PubMed/NCBI

22 

Nie Y, Erion DM, Yuan Z, Dietrich M, Shulman GI, Horvath TL and Gao Q: STAT3 inhibition of gluconeogenesis is downregulated by SirT1. Nat Cell Biol. 11:492–500. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Bernier M, Paul RK, Martin-Montalvo A, Scheibye-Knudsen M, Song S, He HJ, Armour SM, Hubbard BP, Bohr VA, Wang L, et al: Negative regulation of STAT3 protein-mediated cellular respiration by SIRT1 protein. J Biol Chem. 286:19270–19279. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Imai S, Armstrong CM, Kaeberlein M and Guarente L: Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 403:795–800. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B, Howitz KT, Gorospe M, de Cabo R and Sinclair DA: Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science. 305:390–392. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Vaziri H, Dessain SK, Eaton Ng E, Imai SI, Frye RA, Pandita TK, Guarente L and Weinberg RA: hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 107:149–159. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Chen WY, Wang DH, Yen RC, Luo J, Gu W and Baylin SB: Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses. Cell. 123:437–448. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, McBurney M and Guarente L: Mammalian SIRT1 represses forkhead transcription factors. Cell. 116:551–563. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Chen HC, Jeng YM, Yuan RH, Hsu HC and Chen YL: SIRT1 promotes tumorigenesis and resistance to chemotherapy in hepatocellular carcinoma and its expression predicts poor prognosis. Ann Surg Oncol. 19:2011–2019. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Sung JY, Kim R, Kim JE and Lee J: Balance between SIRT1 and DBC1 expression is lost in breast cancer. Cancer Sci. 101:1738–1744. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Huffman DM, Grizzle WE, Bamman MM, Kim JS, Eltoum IA, Elgavish A and Nagy TR: SIRT1 is significantly elevated in mouse and human prostate cancer. Cancer Res. 67:6612–6618. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Jang KY, Kim KS, Hwang SH, Kwon KS, Kim KR, Park HS, Park BH, Chung MJ, Kang MJ, Lee DG and Moon WS: Expression and prognostic significance of SIRT1 in ovarian epithelial tumours. Pathology. 41:366–371. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Cha EJ, Noh SJ, Kwon KS, Kim CY, Park BH, Park HS, Lee H, Chung MJ, Kang MJ, Lee DG, et al: Expression of DBC1 and SIRT1 is associated with poor prognosis of gastric carcinoma. Clin Cancer Res. 15:4453–4459. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Stunkel W, Peh BK, Tan YC, Nayagam VM, Wang X, Salto-Tellez M, Ni B, Entzeroth M and Wood J: Function of the SIRT1 protein deacetylase in cancer. Biotechnol J. 2:1360–1368. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL and Yu JS: Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 5:672006. View Article : Google Scholar : PubMed/NCBI

36 

Jang KY, Hwang SH, Kwon KS, Kim KR, Choi HN, Lee NR, Kwak JY, Park BH, Park HS, Chung MJ, et al: SIRT1 expression is associated with poor prognosis of diffuse large B-cell lymphoma. Am J Surg Pathol. 32:1523–1531. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Rizk SM, Shahin NN and Shaker OG: Association between SIRT1 gene polymorphisms and breast cancer in Egyptians. PLoS One. 11:e01519012016. View Article : Google Scholar : PubMed/NCBI

38 

Wesche J, Haglund K and Haugsten EM: Fibroblast growth factors and their receptors in cancer. Biochem J. 437:199–213. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Marzioni D, Lorenzi T, Mazzucchelli R, Capparuccia L, Morroni M, Fiorini R, Bracalenti C, Catalano A, David G, Castellucci M, et al: Expression of basic fibroblast growth factor, its receptors and syndecans in bladder cancer. Int J Immunopathol Pharmacol. 22:627–638. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Davies H, Hunter C, Smith R, Stephens P, Greenman C, Bignell G, Teague J, Butler A, Edkins S, Stevens C, et al: Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 65:7591–7595. 2005.PubMed/NCBI

41 

Bayraktar S, Thompson PA, Yoo SY, Do KA, Sahin AA, Arun BK, Bondy ML and Brewster AM: The relationship between eight GWAS-identified single-nucleotide polymorphisms and primary breast cancer outcomes. Oncologist. 18:493–500. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Butt S, Harlid S, Borgquist S, Ivarsson M, Landberg G, Dillner J, Carlson J and Manjer J: Genetic predisposition, parity, age at first childbirth and risk for breast cancer. BMC Res Notes. 5:4142012. View Article : Google Scholar : PubMed/NCBI

43 

Chen Y, Shi C and Guo Q: TNRC9 rs12443621 and FGFR2 rs2981582 polymorphisms and breast cancer risk. World J Surg Oncol. 14:502016. View Article : Google Scholar : PubMed/NCBI

44 

Cui F, Wu D, Wang W, He X and Wang M: Variants of FGFR2 and their associations with breast cancer risk: a HUGE systematic review and meta-analysis. Breast Cancer Res Treat. 155:313–335. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Campa D, Barrdahl M, Gaudet MM, Black A, Chanock SJ, Diver WR, Gapstur SM, Haiman C, Hankinson S, Hazra A, et al: Genetic risk variants associated with in situ breast cancer. Breast Cancer Res. 17:822015. View Article : Google Scholar : PubMed/NCBI

46 

Siddiqui S, Chattopadhyay S, Akhtar MS, Najm MZ, Deo SV, Shukla NK and Husain SA: A study on genetic variants of fibroblast growth factor receptor 2 (FGFR2) and the risk of breast cancer from North India. PLoS One. 9:e1104262014. View Article : Google Scholar : PubMed/NCBI

47 

Ledwoń JK, Hennig EE, Maryan N, Goryca K, Nowakowska D, Niwińska A and Ostrowski J: Common low-penetrance risk variants associated with breast cancer in Polish women. BMC Cancer. 13:5102013. View Article : Google Scholar : PubMed/NCBI

48 

Murillo-Zamora E, Moreno-Macías H, Ziv E, Romieu I, Lazcano-Ponce E, Ángeles-Llerenas A, Pérez-Rodríguez E, Vidal-Millán S, Fejerman L and Torres-Mejía G: Association between rs2981582 polymorphism in the FGFR2 gene and the risk of breast cancer in Mexican women. Arch Med Res. 44:459–466. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Ottini L, Silvestri V, Saieva C, Rizzolo P, Zanna I, Falchetti M, Masala G, Navazio AS, Graziano V, Bianchi S, et al: Association of low-penetrance alleles with male breast cancer risk and clinicopathological characteristics: Results from a multicenter study in Italy. Breast Cancer Res. 138:861–868. 2013. View Article : Google Scholar

50 

Jara L, Gonzalez-Hormazabal P, Cerceño K, Di Capua GA, Reyes JM, Blanco R, Bravo T, Peralta O, Gomez F, Waugh E, et al: Genetic variants in FGFR2 and MAP3K1 are associated with the risk of familial and early-onset breast cancer in a South-American population. Breast Cancer Res Treat. 137:559–569. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Miles FL, Rao JY, Eckhert C, Chang SC, Pantuck A and Zhang ZF: Associations of immunity-related single nucleotide polymorphisms with overall survival among prostate cancer patients. Int J Clin Exp Med. 8:11470–11476. 2015.PubMed/NCBI

52 

Yu H, Kortylewski M and Pardoll D: Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat Rev Immunol. 7:41–51. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Yu H, Pardoll D and Jove R: STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 9:798–809. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Niu G, Wright KL, Huang M, Song L, Haura E, Turkson J, Zhang S, Wang T, Sinibaldi D, Coppola D, et al: Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene. 21:2000–2008. 2002. View Article : Google Scholar : PubMed/NCBI

55 

Kusaba T, Nakayama T, Yamazumi K, Yakata Y, Yoshizaki A, Nagayasu T and Sekine I: Expression of p-STAT3 in human colorectal adenocarcinoma and adenoma; correlation with clinicopathological factors. J Clin Pathol. 58:833–838. 2005. View Article : Google Scholar : PubMed/NCBI

56 

He G and Karin M: NF-κB and STAT3-key players in liver inflammation and cancer. Cell Res. 21:159–168. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Pandey MK, Sung B and Aggarwal BB: Betulinic acid suppresses STAT3 activation pathway through induction of protein tyrosine phosphatase SHP-1 in human multiple myeloma cells. Int J Cancer. 127:282–1392. 2010.PubMed/NCBI

58 

Gariboldi MB, Ravizza R and Monti E: The IGFR1 inhibitor NVP-AEW541 disrupts a pro-survival and pro-angiogenic IGF-STAT3-HIF1 pathway in human glioblastoma cells. Biochem Pharmacol. 80:455–462. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Shin J, Lee HJ, Jung DB, Jung JH, Lee HJ, Lee EO, Lee SG, Shim BS, Choi SH, Ko SG, et al: Suppression of STAT3 and HIF-1 alpha mediates anti-angiogenic activity of betulinic acid in hypoxic PC-3 prostate cancer cells. PLoS One. 6:e214922011. View Article : Google Scholar : PubMed/NCBI

60 

Leeman-Neill RJ, Wheeler SE, Singh SV, Thomas SM, Seethala RR, Neill DB, Panahandeh MC, Hahm ER, Joyce SC, Sen M, et al: Guggulsterone enhances head and neck cancer therapies via inhibition of signal transducer and activator of transcription-3. Carcinogenesis. 30:1848–1856. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Rocha GA, Rocha AM, Gomes AD, Faria CL Jr, Melo FF, Batista SA, Fernandes VC, Almeida NB, Teixeira KN, Brito KS and Queiroz DM: STAT3 polymorphism and helicobacter pylori CagA strains with higher number of EPIYA-C segments independently increase the risk of gastric cancer. BMC Cancer. 15:5282015. View Article : Google Scholar : PubMed/NCBI

62 

Yuan K, Liu H, Huang L, Ren X, Liu J, Dong X, Tian W and Jia Y: rs744166 polymorphism of the STAT3 gene is associated with risk of gastric cancer in a Chinese population. Biomed Res Int. 2014:5279182014. View Article : Google Scholar : PubMed/NCBI

63 

Ryan BM, Wolff RK, Valeri N, Khan M, Robinson D, Paone A, Bowman ED, Lundgreen A, Caan B, Potter J, et al: An analysis of genetic factors related to risk of inflammatory bowel disease and colon cancer. Cancer Epidemiol. 38:583–590. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Jiang B, Zhu ZZ, Liu F, Yang LJ, Zhang WY, Yuan HH, Wang JG, Hu XH and Huang G: STAT3 gene polymorphisms and susceptibility to non-small cell lung cancer. Genet Mol Res. 10:1856–1865. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Akaike H: Information theory as an extension of the maximum likelihood principlePetrov BN and Csaki F: 2nd International Symposium on Information Theory. Akademiai Kiado; Budapest: pp. 267–281. 1973

66 

Shan J, Mahfoudh W, Dsouza SP, Hassen E, Bouaouina N, Abdelhak S, Benhadjayed A, Memmi H, Mathew RA, Aigha II, et al: Genome-wide association studies (GWAS) breast cancer susceptibility loci in Arabs: Susceptibility and prognostic implications in Tunisians. Breast Cancer Res Treat. 135:715–724. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

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
Glebauskiene B, Vilkeviciute A, Liutkeviciene R, Jakstiene S, Kriauciuniene L, Zemaitiene R and Zaliuniene D: Association of FGFR2 rs2981582, SIRT1 rs12778366, STAT3 rs744166 gene polymorphisms with pituitary adenoma. Oncol Lett 13: 3087-3099, 2017
APA
Glebauskiene, B., Vilkeviciute, A., Liutkeviciene, R., Jakstiene, S., Kriauciuniene, L., Zemaitiene, R., & Zaliuniene, D. (2017). Association of FGFR2 rs2981582, SIRT1 rs12778366, STAT3 rs744166 gene polymorphisms with pituitary adenoma. Oncology Letters, 13, 3087-3099. https://doi.org/10.3892/ol.2017.5840
MLA
Glebauskiene, B., Vilkeviciute, A., Liutkeviciene, R., Jakstiene, S., Kriauciuniene, L., Zemaitiene, R., Zaliuniene, D."Association of FGFR2 rs2981582, SIRT1 rs12778366, STAT3 rs744166 gene polymorphisms with pituitary adenoma". Oncology Letters 13.5 (2017): 3087-3099.
Chicago
Glebauskiene, B., Vilkeviciute, A., Liutkeviciene, R., Jakstiene, S., Kriauciuniene, L., Zemaitiene, R., Zaliuniene, D."Association of FGFR2 rs2981582, SIRT1 rs12778366, STAT3 rs744166 gene polymorphisms with pituitary adenoma". Oncology Letters 13, no. 5 (2017): 3087-3099. https://doi.org/10.3892/ol.2017.5840