Open Access

Role of single nucleotide polymorphisms of the HSD3B1 gene (rs6203 and rs33937873) in the prediction of prostate cancer risk

  • Authors:
    • Yasmine M. Amrousy
    • Hesham Haffez
    • Doaa M. Abdou
    • Hanaa B. Atya
  • View Affiliations

  • Published online on: July 4, 2022     https://doi.org/10.3892/mmr.2022.12787
  • Article Number: 271
  • Copyright: © Amrousy 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

3‑β‑hydroxysteroid dehydrogenase 1 (HSD3B1) is shown to affect dihydrotestosterone level in prostatic tissue which is a risk factor for prostate cancer (PC). The present study aimed to determine whether rs33937873 (G313A) and rs6203 (C338T) single nucleotide polymorphisms (SNP) in HSD3B1 gene was a potential risk factor for PC susceptibility and can predict the recurrence of PC in Egyptian patients. A total of 186 Egyptian patients were selected with incident primary PC and compared with 180 age healthy controls. The frequencies and the main effect of rs33937873 and rs6203 in HSD3B1 were compared and investigated between the patients and control using genotyping technique and statistical analysis. The mutant GA genotype of G313A in rs33937873 SNP was considered as an independent risk for PC in the multivariate regression analysis [odds ratio (OR)=2.7, 95% confidence intervals (CI): 1.2‑5.5, P=0.01] together with positive history of hypertension (HTN) (OR=6.2, 95% CI: 3.2‑12.1, P=0.0001) and begin prostatic hyperplasia (BPH; OR=8.9, 95% CI: 4.5‑17.5, P=0.0001). Conversely, in rs6203 (C338T), C allele is considered as major risk allele in the development of PC (OR=1.8, 95% CI: 1.3‑2.4, P=0.0003). The univariate logistic regression analyses indicated that CC genotype of rs6203 was a PC risk factor (OR=1.9, 95% CI: 1.3‑2.9, P=0.002). In addition, the frequency of the A‑C haplotype established by rs33937873‑rs6203 was also significantly higher for PC (P=0.013). The predication of PC recurrence was associated only with positive family history (OR=7.7, 95% CI: 2.3‑25.9, P=0.001) and not for The G313A and C338T SNPs. These results suggested that the two HSD3B1 polymorphisms rs33937873 and rs6203 may modify the risk of PC, particularly among patients with HTN and history of BPH, suggesting them as prominent future markers for prediction of PC risk.

Introduction

Prostate cancer (PC) is one of the commonest cancer types affecting men. It is the sixth cause of death worldwide with 359,000 cases in 2018 (1). The discovery of the prostate specific antigen (PSA) together with direct rectal examinations allowed the earlier detection of PC (2). Steroids have been reported as a modulating factor that changes the biochemical characteristics of different tissues such as iris/ciliary body, aqueous outflow pathway and sclera in the rabbit eye (3) and prostate tissue in human (35). Testosterone and dihydrotestosterone (DHT) are the major classes of sterols and sources for androgens in males. They pose a risk to PC patients with higher levels of ‘free’ testosterone and a growth hormone in their blood (6). Steroidogenesis enzymes also have been related to modulation in hormonal level and associated with related diseases (710). Therefore, the hormonal biosynthesis pathway and their receptors can be altered by genetic variations of the related genes altering and contributing to individual susceptibility to PC (11,12). Androgen deprivation therapy (ADT) is one of the standard care treatments in advanced and metastatic cases, whether through testosterone reduction or antagonism of their mechanism of action (13). However, most patients respond well to the ADT, while some patients still show recurrence and failure to therapy and proceed to castration resistant PC (CRPC) (14). This resistance was referred to as either synthesis of the intratumorally androgen from steroid adrenal precursor or from synthesis of de novo cholesterol (15).

A number of studies have focused on different targets for steroid synthesis pathways such as P450 cytochromes of CYP17 and CYP3A4, 5-alpha-reductase type-2 (SRD5A2) (16,17) and 3β-Hydroxysteroid dehydrogenase (3BHSD) genes (18). The 3β-hydroxysteroid dehydrogenases (3β-HSD)/Δ4,5-isomerase is the most important enzyme responsible for catalyzing the 3β-HSD dehydrogenation and Δ4,5-isomerization of the Δ5-steroid precursors into their corresponding Δ4-ketosteroids (19). The activity of this enzyme is important for the synthesis of a number of steroidal hormones including testosterone. 3β-HSD has two key isoenzymes designated as type 1 and type 2 (20). Although these two isoenzymes are encoded by two different genes, they are 93.5% homologous and located on chromosome 1p13.1 (21). The type 1 gene (HSD3B1) is the most important isoenzyme and a rate-limiting enzyme required for dihydrotestosterone synthesis (18), exclusively expressed in the prostate tissue (20). By contrast, the type 2 gene (HSD3B2) is predominantly expressed as 3β-HSD in the adrenal gland, ovary and testis. HSD3B1 converts DHT to 3-α-diol which is metabolized further by uridine diphosphate-glucuronosyltransferases (22). Excess biosynthesis of testosterone is known to upregulate MMP-2 and/or MMP-9 in a number of tissues including prostate (23,24). A number of studies propose a mechanism of association between androgen level, MMPs and PC progression (2528). This is confirmed by the association between significantly higher levels of MMP-2 and −9 levels in serum of PC patients compared with control subjects (25,26). MMPs also serve a pivotal role in determining the influence of the extracellular matrix and its structure remodeling with cell phenotype, cell adhesion molecules, a number of cytokines, chemokines and growth factors. Hence, this results in increasing tumor growth, invasion and metastasis in various pathological conditions such as cancer (26,29,30).

Several common forms of polymorphisms are correlated to allele frequencies in HSD3B1 that affect synthesis, activity and stability of DHT such as rs33937873, rs6203, rs33913717, rs6205 and rs1047303 (31,32). rs6203 has been shown to be implicated in several pathogenesis including myopia (33), gastric cancer (34), sex hormone metabolism (35), hypertensive disorders of pregnancy (36) and hypertension (HTN) with left ventricular structure abnormality (37). As to rs6203 and rs33937873, there is no information about their role, effects or relationships with prevalence of PC even in a small-scale study population. Therefore, in the present study, these two SNPs were selected based on their presence in coding region of HSD3B1 gene which may affect the gene product. The genetic variation in HSD3B1 can lead to an elevation in plasma aldosterone with subsequent elevation in HTN and risk of PC (38). The present study investigated the prevalence of rs6203, which is the C/T silent substitution at codon 338 in exon 4 of HSD3B1 (33) and codon 313 of rs33937873 on HSD3B1 gene in Egyptian PC patients. The present study demonstrated the association between the single nucleotide polymorphisms of rs6203 and rs33937873 in HSD3B1 gene and the risk of PC in Egyptian patients. Additionally, the present study investigated the effect of each SNP, alone or in combination, shedding the light on their haplotype effect, disease susceptibility and any associated clinical parameters.

Patients and methods

Patient samples

A total of 366 Egyptian men were incorporated in the study, categorized into 186 clinically diagnosed PC patients with a mean age of 69.7±0.7 years (range, 54–84 years) and 180 healthy controls with a mean age of 62.2±0.9 years (range, 58–79 years). Patients were recruited from the Urology outpatient clinic of Cairo University Hospital and Badr Hospital, Helwan University between June and December 2021. All participants were acknowledged with the study design and risks with written informed consents taken. The present study protocol was performed according to the ethics guidelines and regulations of the Helsinki Declaration. All experimental protocols were approved by the Scientific Research Ethics Committee of the Faculty of Pharmacy, Helwan University (approval number 03H2021). The clinical guidelines for the National Comprehensive Cancer Network (NCCN) were used for proper diagnosis of PC patients (39). The required informed written consents were obtained according to the regulations of the Institutional Ethical Committee (Faculty of Medicine, Cairo University) which govern the nature of the study. The complete history for the patients including (age, family history, history of benign prostatic hyperplasia (BPH), disease onset and treatment) was recorded with essential laboratory assessment including PSA, prostate size, MRI prostate volume and Gleason grading system. A structured questionnaire was administered to collect information on history of illness, occupation, smoking status, and demographic and anthropometric characteristics of the enrolled subjects. Inclusion criteria for control healthy patients included no evidence of prostate cancer or tumor history before or during the study, and patients were randomly selected for matching by geographic region and the expected age distribution of cases. Inclusion criteria for patients with prostate cancer were a clinical diagnosis of primary adenocarcinoma of the prostate by histopathology and a serum prostate-specific antigen level >4 ng/ml (normal range, 2.5-4.0 ng/ml). Exclusion criteria included: i) Receipt of medical therapy known to affect PSA levels (such as betamethasone or testosterone replacement therapy to increase PSA level and aspirin, ibuprofen, naproxen, atorvastatin, simvastatin and thiazide diuretics to decrease PSA level) (40); ii) previous invasive treatment for benign prostatic hyperplasia, with indwelling urethral catheters (40); iii) voided volume on initial uroflowmetry of <150 ml (40); iv) previous prostate surgery, including transurethral resection of the prostate (41); v) any other cancer or metastatic cancer that has been present during the last 3 years (42); vi) a relationship with another participant at the 3rd degree or closer (43); and vii) missing data pertaining to the essential variables (43).

Biochemical analysis

Blood samples (total volume of ~10 ml from each patient) were used for the determination of PSA level (PSA-ELISA Kit; catalog no. MBS590045; MyBioSource, Inc.) and DNA analysis in both groups.

Genotyping assay

Whole blood samples were subjected for genomic DNA extraction using spin-column technique (GeneJET Genomic DNA Purification kit, Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. SNPs genotyping assays were ordered as follows: (rs6203: C_175679504_10 and rs33937873: C__25619111_10; Thermo Fisher Scientific, Inc.). Genotyping of the two SNPs were performed using TaqMan master mix (Applied Biosystems; Thermo Fisher Scientific, Inc.). Analysis of data was performed by investigating allele using built-in integrated software for investigating allele frequency in Rotor gene-Q machine (Qiagen GmbH).

Statistical analysis

The χ2 test was used to assess the association between the investigated polymorphisms and PC. The odds ratio (OR) and confidence intervals (CI) were performed using SPSS 21.00 software (IBM Corp.). Logistic regression analysis was used for the prediction of risk factors using generalized linear models. Hardy-Weinberg equilibrium and the linkage disequilibrium were both calculated using a goodness-of-fit χ2 test. The HaploView program (version 4.2; Broad Institute) was applied to estimate the haplotypes (44) using the expectation maximization algorithm. Comparisons were performed using two-tailed unpaired t-test and one-way ANOVA (with Tukey's post hoc test) using GraphPad Prism software (version 5.0; GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Demographic data

All clinical and demographic data of PC patients and control are shown in Table I. It was observed that age was not significantly different between groups (P>0.05). The genotypes of both SNPS were in Hardy-Weinberg equilibrium; however, the two investigated polymorphisms were in linkage equilibrium (D'=0.816 & R2=0.026). Moreover, demographic data showed that ~50% of PC patients had DM, HTN and BPH in association.

Table I.

Clinical and demographic data of patients and controls in the study.

Table I.

Clinical and demographic data of patients and controls in the study.

ParametersProstate cancer patients (n=186)Healthy controls (n=180)
Mean age ± SEM, years69.7±0.762.2±0.9
Mean PSA on diagnosis ± SEM, ng/ml14.9±0.62.7±0.2
Family history (prostate cancer), n
  Yes14
  No172
Mean duration ± SEM, years7.7±0.3
TMN staging, n
  T118
  T2a28
  T2b20
  T391
  T430
Recurrence, n
  Yes52
  No134
Mean Gleason score ± SEM6.9±0.16
Benign prostatic hyperplasia, n
  Yes94
  No92
Mean ADT therapy duration ± SEM, months7.8±0.15
Radiotherapy, n
  Yes48
  No138
Diabetes mellitus, n
  Yes7414
  No112166
Hypertension, n
  Yes9636
  No90144

[i] PSA, prostate-specific antigen; TNM, Tumor-Node-Metastasis; ADT, androgen deprivation therapy.

Genotyping analysis

The genotypes distribution and allele frequencies of (rs6203 and rs33937873) among the control subjects and PC patients are shown in Table II and Fig. 1, respectively. Significant differences were observed in the genotype's distribution pattern of rs33937873 between the patients and controls (P=0.0008). Furthermore, the difference in the allele frequencies in rs33937873 was significant (P=0.001). It was quite noticeable that the AA genotype was rare in both healthy subjects and patients (zero subjects). The odds ratio between mutant GA genotypes and wild-type GG genotype was 2.8 (95% CI: 1.5-5.2; P=0.001) in prostate patients compared with controls. OR of A and G alleles was 2.5 (95% CI: 1.4-4.7; P=0.001) in prostate patients compared with controls, suggesting that individuals carrying the A allele are 2.5 times more subjected for developing PC compared with non-carriers.

Table II.

Distribution of (rs33937873 and rs6203) genotypes and allele frequencies in the study subjects.

Table II.

Distribution of (rs33937873 and rs6203) genotypes and allele frequencies in the study subjects.

HSD3B1 gene variantsGenotypesControl (n=180), n (%)Patients (n=186), n (%)P-value, χ2 value, dfOR (95% CI), P-value
rs33937873GG164 (91.1)146 (78.5)0.0008, χ2=11.2, df=12.8 (1.5-5.2), aP=0.001
Silent mutationGA16 (8.9)40 (21.5)
(Pro 313 Pro)AA0 (0.0)0 (0.0)
G allele344 (95.6)332 (89.2)0.001, χ2=10.3, df=12.5 (1.4-4.7), aP=0.001
A allele16 (4.4)40 (10.8)
rs6203CC66 (36.7)98 (52.7)0.0006, χ2=14.7, df=2
CT90 (50.0)80 (43.0)
TT24 (13.3)8 (4.3)
Silent mutationCC66 (36.7)98 (52.7)0.002, χ2=9.4, df=11.9 (1.3-2.9), aP=0.002
(Leu 338 Leu)(TT+CT)114 (63.3)88 (47.3)
T allele138 (38.3)222 (61.7)0.0003, χ2=13.2, df=11.8 (1.3-2.4), aP=0.0003
C allele96 (25.8)276 (74.2)

a P-value data obtained from χ2 test. df, degree of freedom; OR, odds ratio.

In the rs6203, The CC genotype elevated the risk of PC incidence (OR=1.9; 95% CI=1.3-2.9; P=0.002) compared with control subjects. OR of C and T alleles was 1.8 (95% CI: 1.3-2.4, P=0.0003) in prostate patients compared with control subjects (Table II).

Haplotype analysis of studied HSD3B1 SNPs

A total of four haplotypes were generated for the two selected SNPs (rs33937873 and rs6203) of HSD3B1 among patients and control; GC was the most frequent, while the AT was the least frequent haplotypes among the studied groups. Higher frequency of AC and lower frequency of GT were significantly associated with prostate cases when compared with control group, as shown in Table III. The prediction of disease risk was weakly correlated with the susceptibility variants. The predictive performance of genetic risk models increases by merging multiple common low-risk loci. Therefore, the haplotype effect between two SNPs on predisposition of PC in Egyptian patients was studied and found that the polymorphism in both genes had an amplified influence on the risk of PC than single locus.

Table III.

Distribution of haplotype analysis in the study cohort.

Table III.

Distribution of haplotype analysis in the study cohort.

HaplotypeControlPatientsP-value
GT0.3750.2580.013
GC0.5810.6340.258
AT0.0090.0020.998
AC0.0360.1080.013
Regression analysis for prediction of PC susceptibility

To investigate the effect of these gene polymorphisms on PC, regression analysis was conducted for prediction of PC susceptibility with examined family history, DM, HTN and BPH as shown in Table IV.

Table IV.

Regression analysis for prediction of PC susceptibility.

Table IV.

Regression analysis for prediction of PC susceptibility.

Regression analysis for susceptibility of PC incidence

Univariate regression analysisMultivariate regression analysis


VariableP-valueOR95% CIP-valueAdjusted OR95% CI
Age at diagnosis0.9081.0020.974-1.0300.2111.0240.985-1.064
rs339378730.0012.811.509-5.2280.0162.5661.191-5.528
GA vs. GG
rs6203 CC vs. (CT+TT)0.0021.9241.266-2.9220.1581.4790.859-2.546
Family History0.0492.8491.004-8.0810.5830.6940.188-2.56
History of DM0.00017.2683.97-13.3040.0562.1950.978-4.928
History of HT0.00018.5334.94-14.740.00016.2513.219-12.139
History of BPH0.000110.4735.815-18.8650.00018.9724.576-17.591

[i] OR, odd ratio; CI, confidence interval; DM, diabetes mellitus; HT, hypertension; BPH, benign prostatic hyperplasia.

In univariate analysis, it was found that family history, DM, HTN, history of BPH, GA genotype of rs33937873 and CC genotype of rs6203 were associated with risk of PC. However, in multivariable analysis, only patients with history of BPH, HTN and GA genotype of rs33937873 were considered independent predictors of PC susceptibility as shown in Table IV. For the prediction of the PC recurrence regression analysis, this was conducted using age, family history, DM, HTN, BPH, GA genotype of rs33937873 and CC genotype of rs6203 as covariates. Only positive family history was considered a predictor of PC recurrence as represented in Table V.

Table V.

Regression analysis for prediction of PC recurrence.

Table V.

Regression analysis for prediction of PC recurrence.

VariableP-ValueOR95% CI
Age at diagnosis0.8330.9950.952-1.040
rs33937873 GA vs. GG0.6380.8250.371-1.838
rs6203 CC vs. (CT+TT)0.6470.8610.454-1.635
Family History0.0017.742.306-25.965
History of DM0.8180.9260.48-1.786
History of HT0.7041.1320.596-2.151
History of BPH0.0631.8580.966-3.573

[i] CI, confidence interval; DM, diabetes mellitus; HT, hypertension; BPH, benign prostatic hyperplasia.

To investigate the associations of the two selected SNPs and various clinical outcomes in PC patients, patients were stratified according to the type of allelic variant at the polymorphic site of HSD3B1gene. For rs33937873, the statistical analysis was applied to prostate patients carrying the mutant genotype (GA; n=40; 21.5%) who were compared with those carrying the wild-type genotype (GG group; n=146; 78.5%) as the reference group. As for rs6203, the major risk genotype CC (n=98; 52.7%) was compared with the genotype TT (n=8, 4.3%) and heterozygous genotype CT (n=80; 43%; Table VI). In rs33937873, PC patients who carry mutant GA genotypes showed significant increase in prostate volume associated with DM compared with wild ones.

Table VI.

Influence of the HSD3B1 gene polymorphism (rs33937873 & rs6203) on different biochemical and clinical parameters in prostate cancer patients.

Table VI.

Influence of the HSD3B1 gene polymorphism (rs33937873 & rs6203) on different biochemical and clinical parameters in prostate cancer patients.

rs33937873rs6203


ParametersWild genotype (GG) (n=146)Mutant genotype (GA) (n=40)P-valueWild genotype (TT) (n=8)Heterozygous mutant genotype (CT) (n=80)Homozygous mutant genotype (CC) (n=98)
PSA on diagnosis, ng/ml14.4±0.516.8±1.10.0612.0±1.0215.2±0.715.1±0.60.35
Prostate size, cc0.31±0.010.36±0.020.070.30±0.030.31±0.020.34±0.010.36
MRI prostate volume, cm333.6±0.630±1.40.02a36.3±2.631.4±0.933.7±0.80.08
BPH, n (%)72 (49.3)22 (55.0)0.62 (25.0)36 (45.0)56 (57.1)0.09
Recurrence, n (%)42 (28.8)10 (25.0)0.72 (25.0)24 (30.0)26 (26.5)0.8
DM, n (%)64 (43.8)10 (25.0)0.04a2 (25.0)29 (36.2)43 (43.9)0.4
HT, n (%)76 (52.1)20 (50.0)0.94 (50.0)45 (56.2)47 (48.0)0.5

a P<0.05. Data are presented as mean ± SEM unless otherwise stated. PSA, prostate-specific antigen; MRI, magnetic resonance imaging; BPH, benign prostatic hyperplasia; DM, diabetes mellitus; HT, hypertension.

Discussion

Prostate cancer is associated with resistance, poor recovery and metastasis. Hence, early diagnosis is essential for improving the outcome (45). PSA was considered as the gold marker in PC; however, the recorded drawbacks, including non-specificity in PC patients leading to misdiagnosis and failure in cancer treatment, limit its clinical applications (46). Therefore, the ideal for prostate tumor is a molecular marker that is highly specific and sensitive to avoid false positive results. The use of genotype information as an aid for selection can be a rapid and accurate way to enhance selection efficiency of PC patient in a cost-effective manner. In the present study, HSD3B1 was a major enzyme of the androgen biosynthetic pathway (47). It catalyzes the conversion of dehydroepiandrosterone to androstenedione in steroidogenic tissues such as the adrenal and prostate tissues (48). HSD3B1 is considered to serve an important role in the production of androgens that fuel PC development with carcinogenesis and resistance later in a castrate environment (49).

A total of two single nucleotide polymorphisms were selected (rs33937873 with codon 313 and rs6203 with codon 338), however, the functional impact of these two polymorphisms has not yet been fully elucidated in PC and their ethnic distribution was not studied in Egyptian PC patients. The mutant A allele and GA genotype of HSD3B1 gene (rs33937873) indicated a positive association with PC patients (individuals carrying the minor A allele are 2.5 times more susceptible for developing PC compared with non-carriers). The same situation in HSD3B1 gene (rs6203) was observed with C allele significantly increasing the risk of PC incidence and individuals carrying the C allele are 1.8 times more susceptible for developing PC compared with non-carriers. These results are consistent with a number of findings reported for other polymorphisms in HSD3B1 gene suggesting that polymorphisms in this gene is ‘probably damaging’. SNPs can be divided into two main types; non-synonymous SNP or mutation when it presents within the coding region of a gene and this leads to change in amino acid sequence of the resultant protein (50). The other type is synonymous SNPs that affect translation rates or mRNA half-life rather than change the nature of the amino acid (51). SNPs can affect the binding interaction of RNA-protein by modification of secondary structure of RNA (52,53). Additionally, SNPs can affect both gene expression level of the specific protein or its binding with transcription factors (54,55). In all previously mentioned mechanisms, these can lead to modifications in either function or structure of the translated proteins (folding) and related metabolic pathways such as increased cell proliferation, protein dimerization and activation of a number of mediators (56). In the current study, the suggested polymorphisms, either G313A or C338T, may create a new potential protein with different structure and function which induces cellular carcinogenesis, resistance and apoptosis (57). Another suggestion is that the two polymorphisms may render HSD3B1 resistant to ubiquitination and proteasomal degradation, leading to a large amount of protein (DHT) accumulation in the cell, causing prostate tissue carcinogenesis as well as resistance to androgen-deprivation therapy in PC recurrence (5760). The results suggested that these variants of the HSD3B1 steroidogenic enzyme gene could be a powerful new biomarker capable of identifying patients with aggressive disease who warrant early escalated therapy and in clinical management of the disease. Additionally, the data obtained and suggestions were matched with literature about the role of this enzyme in the degradation of DHT (61) and malfunction in accumulation of DHT in prostatic tissue (62,63).

For the correlation of the contribution of the studied SNPs to PC susceptibility, regression analysis was performed using rs33937873, rs6203 and other variables as covariates. Positive family history, DM, HTN, GA genotype of rs33937873 and CC genotype of rs6203 were associated with risk of PC in univariable analysis. On the other hand, in multivariable analysis, only patients with a history of BPH, HTN and GA genotype of rs33937873 were considered independent predictors of PC susceptibility. Family history was considered the only predictor of PC recurrence in the present study, as a well-known risk factor for developing PC (64). The literature matched this observation and shows that there is a trend of increasing risk of PC incidence in patients with two or three first degree relatives affected to have a five and 11-fold increased risk of developing PC (6568). In the US, those with a family history of PC should be advised of their significantly increased PC risk to ~9–10% in their lifetime (69). In Africa, some studies found a correlation between PC incidence in different African cultures and their family history reached between 30–70% (69,70). With respect to this biological heterogeneity of PC, the observation is important in understanding PC etiology and incidence risk factor to correctly assess the clinical state of the patient using PSA continuous screening to avoid aggressiveness and high mortality rate.

In addition, the findings of the present study reported significant association between the mutant genotype GA of rs33937873 in PC patients with DM. Although studies mention that DM and PC have an inverse relationship (7173), some other studies could not find any evidence of the inverse relationship between DM and PC (7476). However, some other results in population-based cohort study concur with the findings of the present study suggesting that the relationship between DM and high-grade PC has a positive correlation (77,78). The potential explanation for this hypothesis is related to the activity of the patient such as exercise, body mass index (79,80), glycemic control (78) and ethnicity (81) and further large scale studies are required to understand the proper mechanisms controlling the correlation in each case.

The data presented in the present study shed light on the potential role of these two SNPs in HSD3B1gene as the promising marker for the prediction of PC incidence. The future perspective for is to illustrate the effect of both studied SNPs in different advanced cases of PC such as CRPC through the activity of HSD3B1 and to study their role in susceptibility and resistance of prostate patients to treatments such as abiraterone.

The present study has suggested a potential impact of SNPs in HSD3B1 gene (rs6203 and rs33937873) individually and in combination in relation to the risk of PC in Egyptian patients. These results deserve the trial on a larger study in the context of PC susceptibility to shed the light on the function of HSD3B1 and associated allelic variants in correlation to other androgen-metabolizing enzymes. Moreover, the present study needs to be applied using both in vitro and in vivo models to confirm the hypothesis and elucidate the role of the gene and its corresponding protein in PC pathogenesis.

Acknowledgements

The authors would like to thank Professor Sameh Soror (Helwan University, Centre of Scientific Excellence ‘Helwan Structural Biology Research’, Helwan University, Cairo) for the opportunity to use the facilities/equipment available in the Helwan Structural Biology Research Laboratory.

Funding

Funding: No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

HH, HBA, YMA and DMA designed the study, performed the experiments, and wrote and revised the manuscript. HBA, YMA and HH analyzed the datasets. HH searched the literature. HH, HBA YMA and DMA confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All subjects gave their written informed consent to participate in the study. The study was approved by Scientific Research Ethics Committee of the Faculty of Pharmacy, Helwan university (approval number 03H2021).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Culp MB, Soerjomataram I, Efstathiou JA, Bray F and Jemal A: Recent global patterns in prostate cancer incidence and mortality rates. Eur Urol. 77:38–52. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA, Penning TM, Febbo PG and Balk SP: Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res. 66:2815–2825. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Knepper PA, Collins JA and Frederick R: Effects of dexamethasone, progesterone, and testosterone on IOP and GAGs in the rabbit eye. Investig Ophthalmol Vis Sci. 26:1093–1100. 1985.PubMed/NCBI

4 

Ettinger SL, Sobel R, Whitmore TG, Akbari M, Bradley DR, Gleave ME and Nelson CC: Dysregulation of sterol response element-binding proteins and downstream effectors in prostate cancer during progression to androgen independence. Cancer Res. 64:2212–2221. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Heemers H, Maes B, Foufelle F, Heyns W, Verhoeven G and Swinnen JV: Androgens stimulate lipogenic gene expression in prostate cancer cells by activation of the sterol regulatory element-binding protein cleavage activating protein/sterol regulatory element-binding protein pathway. Mol Endocrinol. 15:1817–1828. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Vis AN and Schröder FH: Key targets of hormonal treatment of prostate cancer. Part 1: The androgen receptor and steroidogenic pathways. BJU Int. 104:438–448. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Hamad A, Kluk M, Fox J, Park M and Turner JE: The effects of aromatase inhibitors and selective estrogen receptor modulators on eye development in the zebrafish (Danio rerio). Curr Eye Res. 32:819–827. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Deb S, Chin MY, Pham S, Adomat H, Hurtado-Coll A, Gleave ME and Tomlinson Guns ES: Steroidogenesis in peripheral and transition zones of human prostate cancer tissue. Int J Mol Sci. 22:4872021. View Article : Google Scholar : PubMed/NCBI

9 

Torres MJ, López-Moncada F, Herrera D, Indo S, Lefian A, Llanos P, Tapia J, Castellón EA and Contreras HR: Endothelin-1 induces changes in the expression levels of steroidogenic enzymes and increases androgen receptor and testosterone production in the PC3 prostate cancer cell line. Oncol Rep. 46:1712021. View Article : Google Scholar : PubMed/NCBI

10 

Hou Z, Yang T, Mei Z, Zhang S, Gao Y, Chen X, Tan Q, Zhu X, Xu C, Lian J, et al: Tracing steroidogenesis in prostate biopsy samples to unveil prostate tissue androgen metabolism characteristics and potential clinical application. J Steroid Biochem Mol Biol. 210:1058592021. View Article : Google Scholar : PubMed/NCBI

11 

Cunningham JM, Hebbring SJ, McDonnell SK, Cicek MS, Christensen GB, Wang L, Jacobsen SJ, Cerhan JR, Blute ML, Schaid DJ and Thibodeau SN: Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer. Cancer Epidemiol Biomark Prev. 16:969–978. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Holzbeierlein J, Lal P, LaTulippe E, Smith A, Satagopan J, Zhang L, Ryan C, Smith S, Scher H, Scardino P, et al: Gene expression analysis of human prostate carcinoma during hormonal therapy identifies androgen-responsive genes and mechanisms of therapy resistance. Am J Pathol. 164:217–227. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Crawford ED, Heidenreich A, Lawrentschuk N, Tombal B, Pompeo ACL, Mendoza-Valdes A, Miller K, Debruyne FMJ and Klotz L: Androgen-targeted therapy in men with prostate cancer: Evolving practice and future considerations. Prostate Cancer Prostatic Dis. 22:24–38. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Shiota M, Yokomizo A and Naito S: Pro-survival and anti-apoptotic properties of androgen receptor signaling by oxidative stress promote treatment resistance in prostate cancer. Endocr Relat Cancer. 19:R243–R253. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, True LD and Nelson PS: Maintenance of intratumoral androgens in metastatic prostate cancer: A mechanism for castration-resistant tumor growth. Cancer Res. 68:4447–4454. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Schiffer L, Barnard L, Baranowski ES, Gilligan LC, Taylor AE, Arlt W, Shackleton CHL and Storbeck KH: Human steroid biosynthesis, metabolism and excretion are differentially reflected by serum and urine steroid metabolomes: A comprehensive review. J Steroid Biochem Mol Biol. 194:1054392019. View Article : Google Scholar : PubMed/NCBI

17 

Gsur A, Feik E and Madersbacher S: Genetic polymorphisms and prostate cancer risk. World J Urol. 21:414–423. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Evaul K, Li R, Papari-Zareei M, Auchus RJ and Sharifi N: 3beta-hydroxysteroid dehydrogenase is a possible pharmacological target in the treatment of castration-resistant prostate cancer. Endocrinol. 151:3514–3520. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Mason JI, Keeney DS, Bird IM, Rainey WE, Morohashi K, Leers-Sucheta S and Melner MH: The regulation of 3 beta-hydroxysteroid dehydrogenase expression. Steroids. 62:164–168. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Simard J, Ricketts ML, Gingras S, Soucy P, Feltus FA and Melner MH: Molecular biology of the 3beta-hydroxysteroid dehydrogenase/delta5-delta4 isomerase gene family. Endocr Rev. 26:525–582. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Rhéaume E, Lachance Y, Zhao HF, Breton N, Dumont M, de Launoit Y, Trudel C, Luu-The V, Simard J and Labrie F: Structure and expression of a new complementary DNA encoding the almost exclusive 3 beta-hydroxysteroid dehydrogenase/delta 5-delta 4-isomerase in human adrenals and gonads. Mol Endocrinol. 5:1147–1157. 1991. View Article : Google Scholar : PubMed/NCBI

22 

Schiffer L, Arlt W and Storbeck KH: Intracrine androgen biosynthesis, metabolism and action revisited. Mol Cell Endocrinol. 465:4–26. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Silva SA, Gobbo MG, Pinto-Fochi ME, Rafacho A, Taboga SR, Almeida EA, Góes RM and Ribeiro DL: Prostate hyperplasia caused by long-term obesity is characterized by high deposition of extracellular matrix and increased content of MMP-9 and VEGF. Int J Exp Pathol. 96:21–30. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Edlund M, Sung SY and Chung LW: Modulation of prostate cancer growth in bone microenvironments. J Cell Biochem. 91:686–705. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Trudel D, Fradet Y, Meyer F, Harel F and Têtu B: Significance of MMP-2 expression in prostate cancer: An immunohistochemical study. Cancer Res. 63:8511–8515. 2003.PubMed/NCBI

26 

Xie T, Dong B, Yan Y, Hu G and Xu Y: Association between MMP-2 expression and prostate cancer: A meta-analysis. Biomed Rep. 4:241–245. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Gong Y, Chippada-Venkata UD and Oh WK: Roles of matrix metalloproteinases and their natural inhibitors in prostate cancer progression. Cancers (Basel). 6:1298–1327. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Abdelaal MR, Soror SH, Elnagar MR and Haffez H: Revealing the potential application of EC-synthetic retinoid analogues in anticancer therapy. Molecules. 26:5062021. View Article : Google Scholar : PubMed/NCBI

29 

Nemeth JA, Yousif R, Herzog M, Che M, Upadhyay J, Shekarriz B, Bhagat S, Mullins C, Fridman R and Cher ML: Matrix metalloproteinase activity, bone matrix turnover, and tumor cell proliferation in prostate cancer bone metastasis. J Natl Cancer Inst. 94:17–25. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Overall CM and López-Otín C: Strategies for MMP inhibition in cancer: Innovations for the post-trial era. Nat Rev Cancer. 2:657–672. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Chang KH, Li R, Kuri B, Lotan Y, Roehrborn CG, Liu J, Vessella R, Nelson PS, Kapur P, Guo X, et al: A gain-of-function mutation in DHT synthesis in castration-resistant prostate cancer. Cell. 154:1074–1084. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Sabharwal N and Sharifi N: HSD3B1 genotypes conferring adrenal-restrictive and adrenal-permissive phenotypes in prostate cancer and beyond. Endocrinology. 160:2180–2188. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Chen ZTY, Wang IJ, Liao YT, Shih YF and Lin LLK: Polymorphisms in steroidogenesis genes, sex steroid levels, and high myopia in the Taiwanese population. Mol Vis. 17:2297–2310. 2011.PubMed/NCBI

34 

Cho LY, Yang JJ, Ko KP, Ma SH, Shin A, Choi BY, Han DS, Song KS, Kim YS, Chang SH, et al: Genetic susceptibility factors on genes involved in the steroid hormone biosynthesis pathway and progesterone receptor for gastric cancer risk. PLoS One. 7:e476032012. View Article : Google Scholar : PubMed/NCBI

35 

Carmichael S, Witte J, Ma C, Lammer E and Shaw G: Hypospadias and variants in genes related to sex hormone biosynthesis and metabolism. Andrology. 2:130–137. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Shimodaira M, Nakayama T, Sato I, Sato N, Izawa N, Mizutani Y, Furuya K and Yamamoto T: Estrogen synthesis genes CYP19A1, HSD3B1, and HSD3B2 in hypertensive disorders of pregnancy. Endocrine. 42:700–707. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Shimodaira M, Nakayama T, Sato N, Aoi N, Sato M, Izumi Y, Soma M and Matsumoto K: Association of HSD3B1 and HSD3B2 gene polymorphisms with essential hypertension, aldosterone level, and left ventricular structure. Eur J Endocrinol. 163:671–810. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Gao H, Xu J, Ma Q, Tang F, Ga Q, Li Y, Guan W, Ge R and Yang YZ: Association between the polymorphism of steroid hormone metabolism genes and high-altitude pulmonary edema in the Chinese Han population. Int J Gen Med. 15:787–794. 2022. View Article : Google Scholar : PubMed/NCBI

39 

Mohler JL, Armstrong AJ, Bahnson RR, Boston B, Busby JE, D'Amico AV, Eastham JA, Enke CA, Farrington T, Higano CS, et al: Prostate cancer, version 3.2012: Featured updates to the NCCN guidelines. J Natl Compr Canc Netw. 10:1081–1087. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Cormio L, Lucarelli G, Netti GS, Stallone G, Selvaggio O, Troiano F, Di Fino G, Sanguedolce F, Bufo P, Grandaliano G and Carrieri G: Post-void residual urinary volume is an independent predictor of biopsy results in men at risk for prostate cancer. Anticancer Res. 35:2175–2182. 2015.PubMed/NCBI

41 

Jeong CW, Hong SK, Byun SS, Jeon SS, Seo SI, Lee HM, Ahn H, Kwon DD, Ha HK, Kwon TG, et al: Selection criteria for active surveillance of patients with prostate cancer in Korea: A multicenter analysis of pathology after radical prostatectomy. Cancer Res Treat. 50:265–274. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Solanki AA, Schroth CA, Authier C, Carlson K, Garraway I, Haegerich T, Henry E, Jones JA, Joseph R, Koppes T, et al: Veterans affairs seamless phase II/III randomized trial of standard systemic therapy with or without PET-directed local therapy for oligorecurrent prostate cancer (VA STARPORT). J Clin Oncol. 40 (Suppl 6):TPS2032022. View Article : Google Scholar

43 

Jiang Y, Meyers TJ, Emeka AA, Cooley LF, Cooper PR, Lancki N, Helenowski I, Kachuri L, Lin DW, Stanford JL, et al: Genetic factors associated with prostate cancer conversion from active surveillance to treatment. HGG Adv. 3:1000702022.PubMed/NCBI

44 

Barrett JC, Fry B, Maller J and Daly MJ: Haploview: Analysis and visualization of LD and haplotype maps. Bioinformatics. 21:263–265. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Litwin MS and Tan HJ: The diagnosis and treatment of prostate cancer: A review. JAMA. 317:2532–2542. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Adhyam M and Gupta AK: A review on the clinical utility of PSA in cancer prostate. Indian J Surg Oncol. 3:120–129. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Wu G, Huang S, Nastiuk KL, Li J, Gu J, Wu M, Zhang Q, Lin H and Wu D: Variant allele of HSD3B1 increases progression to castration-resistant prostate cancer. Prostate. 75:777–782. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Sharifi N, McPhaul MJ and Auchus RJ: ‘Getting from here to there’-mechanisms and limitations to the activation of the androgen receptor in castration-resistant prostate cancer. J Investig Med. 58:938–944. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Dai C, Heemers H and Sharifi N: Androgen signaling in prostate cancer. Cold Spring Harb Perspect Med. 7:a0304522017. View Article : Google Scholar : PubMed/NCBI

50 

Mendell JT and Dietz HC: When the message goes awry: Disease-producing mutations that influence mRNA content and performance. Cell. 107:411–414. 2001. View Article : Google Scholar : PubMed/NCBI

51 

Nicholson P, Yepiskoposyan H, Metze S, Zamudio Orozco R, Kleinschmidt N and Mühlemann O: Nonsense-mediated mRNA decay in human cells: Mechanistic insights, functions beyond quality control and the double-life of NMD factors. Cell Mol Life Sci. 67:677–700. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Beaudoin JD and Perreault JP: 5′-UTR G-quadruplex structures acting as translational repressors. Nucleic Acids Res. 38:7022–7036. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Robert F and Pelletier J: Exploring the impact of single-nucleotide polymorphisms on translation. Front Genet. 507:5072018. View Article : Google Scholar : PubMed/NCBI

54 

Das SC, Rahman M and Das Gupta S: In-silico analysis unravels the structural and functional consequences of non-synonymous SNPs in the human IL-10 gene. Egypt J Med Hum Genet. 23:1–14. 2022. View Article : Google Scholar

55 

Kucukkal TG, Petukh M, Li L and Alexov E: Structural and physico-chemical effects of disease and non-disease nsSNPs on proteins. Curr Opin Struct Biol. 32:18–24. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Gebert M, Jaśkiewicz M, Moszyńska A, Collawn JF and Bartoszewski R: The effects of single nucleotide polymorphisms in cancer RNAi therapies. Cancers (Basel). 12:31192020. View Article : Google Scholar : PubMed/NCBI

57 

Park JY, Tanner JP, Sellers TA, Huang Y, Stevens CK, Dossett N, Shankar RA, Zachariah B, Heysek R and Pow-Sang J: Association between polymorphisms in HSD3B1 and UGT2B17 and prostate cancer risk. Urology. 70:374–379. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Han FF, Ren LL, Xuan LL, Lv YL, Liu H, Gong LL, An ZL and Liu LH: HSD3B1 variant and androgen-deprivation therapy outcome in prostate cancer. Cancer Chemother Pharmacol. 87:103–112. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Setlur SR, Chen CX, Hossain RR, Ha JS, Van Doren VE, Stenzel B, Steiner E, Oldridge D, Kitabayashi N, Banerjee S, et al: Genetic variation of genes involved in dihydrotestosterone metabolism and the risk of prostate cancer. Cancer Epidemiol Biomark Prev. 19:229–239. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Huang J, Huang D and Na R: The association between genetic variants in HSD3B1 and clinical management of PCa. J Transl Genet Genom. 5:240–249. 2021.PubMed/NCBI

61 

Devgan SA, Henderson BE, Yu MC, Shi CY, Pike MC, Ross RK and Reichardt JK: Genetic variation of 3 beta-hydroxysteroid dehydrogenase type II in three racial/ethnic groups: Implications for prostate cancer risk. Prostate. 33:9–12. 1997. View Article : Google Scholar : PubMed/NCBI

62 

Kosaka T, Miyajima A and Oya M: Is DHT production by 5α-reductase friend or foe in prostate cancer? Front Oncol. 4:2472014. View Article : Google Scholar : PubMed/NCBI

63 

Stangl-Kremser J, Lemberger U, Hassler MR, Bruchbacher A, Ilijazi D, Garstka N, Kramer G, Haitel A, Abufaraj M and Shariat SF: Prevalence and prognostic value of the polymorphic variant 1245A>C of HSD3B1 in castration-resistant prostate cancer. Clin Genitourin Cancer. 17:389–394. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Johns L and Houlston R: A systematic review and meta-analysis of familial prostate cancer risk. BJU Int. 91:789–794. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Barber L, Gerke T, Markt SC, Peisch SF, Wilson KM, Ahearn T, Giovannucci E, Parmigiani G and Mucci LA: Family history of breast or prostate cancer and prostate cancer risk. Clin Cancer Res. 24:5910–5917. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Chen YC, Page JH, Chen R and Giovannucci E: Family history of prostate and breast cancer and the risk of prostate cancer in the PSA era. Prostate. 68:1582–1591. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Thomas JA II, Gerber L, Moreira DM, Hamilton RJ, Bañez LL, Castro-Santamaria R, Andriole GL, Isaacs WB, Xu J and Freedland SJ: Prostate cancer risk in men with prostate and breast cancer family history: Results from the REDUCE study (R1). J Intern Med. 272:85–92. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Cunningham GR, Ashton CM, Annegers JF, Souchek J, Klima M and Miles B: Familial aggregation of prostate cancer in African-Americans and white Americans. Prostate. 56:256–262. 2003. View Article : Google Scholar : PubMed/NCBI

69 

Steinberg GD, Carter BS, Beaty TH, Childs B and Walsh PC: Family history and the risk of prostate cancer. Prostate. 17:337–347. 1990. View Article : Google Scholar : PubMed/NCBI

70 

Acheampong E, Adu EA, Obirikorang C, Amoah G, Afriyie OO, Yorke J and Yeboah FA: Association of genetic variants with prostate cancer in Africa: A concise review. Egypt J Med Hum Genet. 22:1–9. 2021. View Article : Google Scholar

71 

Kasper JS, Liu Y and Giovannucci E: Diabetes mellitus and risk of prostate cancer in the health professionals follow-up study. Int J Cancer. 124:1398–1403. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Bonovas S, Filioussi K and Tsantes A: Diabetes mellitus and risk of prostate cancer: A meta-analysis. Diabetologia. 47:1071–1078. 2004. View Article : Google Scholar : PubMed/NCBI

73 

Feng X, Song M, Preston MA, Ma W, Hu Y, Pernar CH, Stopsack KH, Ebot EM, Fu BC, Zhang Y, et al: The association of diabetes with risk of prostate cancer defined by clinical and molecular features. Br J Cancer. 123:657–665. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Chan JM, Latini DM, Cowan J, Duchane J and Carroll PR: History of diabetes, clinical features of prostate cancer, and prostate cancer recurrence-data from CaPSURE (United States). Cancer Causes Control. 16:789–797. 2005. View Article : Google Scholar : PubMed/NCBI

75 

Au Yeung SL and Schooling CM: Impact of glycemic traits, type 2 diabetes and metformin use on breast and prostate cancer risk: A Mendelian randomization study. BMJ Open Diabetes Res Care. 7:e0008722019. View Article : Google Scholar : PubMed/NCBI

76 

Wu C, Moreira DM, Gerber L, Rittmaster RS, Andriole GL and Freedland SJ: Diabetes and prostate cancer risk in the REDUCE trial. Prostate Cancer Prostatic Dis. 14:326–331. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Li Q, Kuriyama S, Kakizaki M, Yan H, Sone T, Nagai M, Sugawara Y, Ohmori-Matsuda K, Hozawa A, Nishino Y and Tsuji I: History of diabetes mellitus and the risk of prostate cancer: The Ohsaki cohort study. Cancer Causes Control. 21:1025–1032. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Park J, Cho SY, Lee YJ, Lee SB, Son H and Jeong H: Poor glycemic control of diabetes mellitus is associated with higher risk of prostate cancer detection in a biopsy population. PLoS One. 9:e1047892014. View Article : Google Scholar : PubMed/NCBI

79 

Leitzmann MF, Ahn J, Albanes D, Hsing AW, Schatzkin A, Chang SC, Huang WY, Weiss JM, Danforth KN, Grubb RL III, et al: Diabetes mellitus and prostate cancer risk in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Causes Control. 19:1267–1276. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Fall K, Garmo H, Gudbjörnsdottir S, Stattin P and Zethelius B: Diabetes mellitus and prostate cancer risk; a nationwide case-control study within PCBaSe Sweden. Cancer Epidemiol Biomarkers Prev. 22:1102–1109. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Moreira DM, Anderson T, Gerber L, Thomas JA, Bañez LL, McKeever MG, Hoyo C, Grant D, Jayachandran J and Freedland SJ: The association of diabetes mellitus and high-grade prostate cancer in a multiethnic biopsy series. Cancer Causes Control. 22:977–983. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2022
Volume 26 Issue 2

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
Amrousy YM, Haffez H, Abdou DM and Atya HB: Role of single nucleotide polymorphisms of the HSD3B1 gene (rs6203 and rs33937873) in the prediction of prostate cancer risk. Mol Med Rep 26: 271, 2022
APA
Amrousy, Y.M., Haffez, H., Abdou, D.M., & Atya, H.B. (2022). Role of single nucleotide polymorphisms of the HSD3B1 gene (rs6203 and rs33937873) in the prediction of prostate cancer risk. Molecular Medicine Reports, 26, 271. https://doi.org/10.3892/mmr.2022.12787
MLA
Amrousy, Y. M., Haffez, H., Abdou, D. M., Atya, H. B."Role of single nucleotide polymorphisms of the HSD3B1 gene (rs6203 and rs33937873) in the prediction of prostate cancer risk". Molecular Medicine Reports 26.2 (2022): 271.
Chicago
Amrousy, Y. M., Haffez, H., Abdou, D. M., Atya, H. B."Role of single nucleotide polymorphisms of the HSD3B1 gene (rs6203 and rs33937873) in the prediction of prostate cancer risk". Molecular Medicine Reports 26, no. 2 (2022): 271. https://doi.org/10.3892/mmr.2022.12787