Next Article in Journal
FRMD7 Gene Alterations in a Pakistani Family Associated with Congenital Idiopathic Nystagmus
Next Article in Special Issue
Molecular Evolution of SARS-CoV-2 during the COVID-19 Pandemic
Previous Article in Journal
High Levels of Diversity in Anopheles Subgenus Kerteszia Revealed by Species Delimitation Analyses
Previous Article in Special Issue
The Evolutionary History of a DNA Methylase Reveals Frequent Horizontal Transfer and Within-Gene Recombination
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Tumor Androgen Receptor Protein Level Is Positively Associated with a Better Overall Survival in Melanoma Patients

1
College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
2
Department of Dermatology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
3
Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
*
Author to whom correspondence should be addressed.
Genes 2023, 14(2), 345; https://doi.org/10.3390/genes14020345
Submission received: 10 December 2022 / Revised: 19 January 2023 / Accepted: 25 January 2023 / Published: 28 January 2023
(This article belongs to the Special Issue Feature Papers: Molecular Genetics and Genomics 2023)

Abstract

:
Androgen receptor (AR) is expressed in numerous tissues and serves important biologic functions in skin, prostate, immune, cardiovascular, and neural systems, alongside sexual development. Several studies have associated AR expression and patient survival in various cancers, yet there are limited studies examining the relationship between AR expression and cutaneous melanoma. This study used genomics and proteomics data from The Cancer Proteome Atlas (TCPA) and The Cancer Genome Atlas (TCGA), with 470 cutaneous melanoma patient data points. Cox regression analyses evaluated the association between AR protein level with overall survival and revealed that a higher level of AR protein was positively associated with a better overall survival (OS) (p = 0.003). When stratified by sex, the AR association with OS was only significant for both sexes. The multivariate Cox models with justifications of sex, age of diagnosis, stage of disease, and Breslow depth of the tumor confirmed the AR-OS association in all patients. However, the significance of AR was lost when ulceration was included in the model. When stratified by sex, the multivariate Cox models indicated significant role of AR in OS of female patients but not in males. AR-associated genes were identified and enrichment analysis revealed shared and distinct gene network in male and female patients. Furthermore, AR was found significantly associated with OS in RAS mutant subtypes of melanoma but not in BRAF, NF1, or triple-wild type subtypes of melanoma. Our study may provide insight into the well-known female survival advantage in melanoma patients.

1. Introduction

Melanoma incidence continues to increase worldwide; in the US, it has increased by 320% since 1975 [1]. Research on how sex hormones and their receptors impact melanoma have not resulted in solid conclusions. Androgen receptor (AR), for example, was recently reported to exhibit effects of promoting cell proliferation, melanoma metastasis, and drug resistance in melanoma cells and mouse models [2,3,4]. While molecular studies and mouse models have provided much interesting information, we are interested in investigating whether AR was differentially expressed in melanoma tumors from men and women, and whether the tumor AR levels are associated with patient overall survival (OS). This study shall shed insight into a long-observed phenomena, i.e., the female survival advantage of melanoma patients [5,6].
As a male sex hormone receptor, the gene AR is located on the X chromosome [7]. Two androgenic hormones that are able to bind to AR include testosterone (T), and its metabolite dihydrotestosterone (DHT), and they are active in human skin in endocrine and paracrine manner [8,9,10]. AR and these hormones exert their genomic effects via induction of transcriptional activities, and non-genomics activity through signal transduction, both of which are best studied in human prostate cancer [11,12,13].
Sex differences in cancer incidence have also been documented in several cancers, [14]. For instance, higher incidence rates of lung, liver, stomach, esophageal, and bladder malignancies alongside cutaneous melanoma are found in males compared to females [15,16,17,18]. Aside from lifestyle, the characterization of the molecular differences in cancer between male and female malignancies highlights the sex-based variations of gene expression on a molecular level [19]. Nonetheless, there remains a lack of complete understanding of what role AR signaling plays in most hormone-independent cancers alongside cutaneous melanoma.
Current literature has explored possible pathways into AR’s effects, both harmful and protective, on cutaneous melanoma development. One proposed mechanism explains how AR and the protein Early Growth Response 1 (EGR1) increase melanoma proliferation through coordinated transcriptional regulation of several growth-regulatory genes, including the repression of EGR1-mediated transcriptional activation of p21Waf1/Cip1, a known tumor suppressor gene [20]. Another mechanism suggesting melanoma progression includes altering the miRNA-539-3p/USP13 signaling to reduce de-ubiquitination of MITF protein, increasing MITF degradation, and allowing further invasion [4]. Other mechanisms of AR’s role in cancer risks have been proposed to provide a potential protective effect, specifically cancer-associated fibroblast (CAF) activation. Decreased AR expression in primary human dermal fibroblasts (HDFs) derived from multiple individuals led to early steps of CAF activation. The discovered mechanism includes the development of a complex in which AR combines with CSL/RBP-Jκ to normally repress the transcription of key CAF effector gene [21].
The conflicting findings of AR’s protective or harmful role in melanoma progression shows the complexity of several convergent mechanisms likely implicated in melanoma’s AR dependency. In this paper, we use The Cancer Genome Atlas (TCGA) and The Cancer Proteome Atlas (TCPA) to evaluate the relationship between AR gene and AR protein expression in human cutaneous melanoma, and their association with OS in patients. Genomic network was further explored in an attempt to understand the sex-differentiated roles of AR in patient overall survival.

2. Materials and Methods

2.1. The Source of Data

The data source used for all analyses (TCGA-SKCM) were obtained from The Cancer Genome Atlas (TCGA), with mRNA sequencing (RNA-Seq) data downloaded from Broad Firehose GDAC (http://gdac.broadinstitute.org/, accessed on 14 June 2022), and proteomics data from The Cancer Proteome Atlas (TCPA) (https://tcpaportal.org/, accessed on 14 June 2022). The RNA-Seq data were retrieved as RSEM (RNA-Seq by Expectation-Maximization) and Z scores [22]. Patient ID, sex, age of diagnosis, follow-up time, and survival status were also downloaded from the Broad GDAC site. The database contained 480 tumors from 471 patients with cutaneous melanoma. Protein expression data were available for 355 tumors. If patient duplicates were encountered, the data for metastatic tumor was selected and the primary tumor data were discarded. Tumor stages are grouped into early (Stage I and Stage II) or late stage (Stage III and Stage VI) or used as denoted in the dataset as Stage 0–4.

2.2. Statistical Methods

All statistics were analyzed using Stata 17. Linear regression was used to examine the association of mRNA and protein levels in tumor samples. AR levels (mRNA or protein) were compared between sex by Student t-test and/or rank-sum test. Cox regression analyses were performed to evaluate the association between AR protein level with overall survival. The AR-high and AR-low groups were defined by the median of AR protein level (−0.718). The regression model was further stratified by sex or adjusted to age, tumor stage (early and late), Breslow depth, and ulceration status of the tumors. Sex was also used as an adjusting co-variable in the overall model. The overall survival was defined as the period from date of diagnosis until death from any cause. Significance levels are set at 0.05 (two-sided) for all analysis.

2.3. Gene Network Analysis

AR-co-expressed genes (based on RNA-Seq) were extracted from the cBioportal website (https://www.cbioportal.org/, accessed on 14 June 2022) using sex-stratified patient information. The AR-co-expressed genes were processed using adjusted q values of 0.05, followed by cutoff value of Spearman’s coefficient of 0.3 [23]. A set of genes that are uniquely associated with AR in male and female tumors were identified and then subjected to a functional enrichment analysis using g:profiler web-based analysis tools (https://biit.cs.ut.ee/gprofiler/, accessed on 14 June 2022).

3. Results

3.1. The Sex Difference of AR Gene Expression at mRNA and Protein Level

The TCGA SKCM dataset was downloaded from the Broad Institute Firehose website. The baseline characterizations of patients are listed in Table 1. The protein quantification data are available for 353 patients and the mRNA data are available for all 471 patients. The mRNA level of AR was compared between tumors from male and female sources using log transformed RSEM. A total of 21 female tumors and 34 male tumors did not show detectable levels of mRNA (RSEM = 0), but they were also included in the analysis. Student t-test showed no sex difference in mean of log transformed RSEM values (p = 0.10). However, when the protein levels were used for sex comparison, tumors from females (N = 144) showed a significant lower level of AR protein than those from males (N = 208) (p = 0.0099) (Table 2).
We then investigated whether tumor mRNA and protein levels of AR are positively associated. In fact, a linear regression model between AR protein and log-transformed RSEM data showed significant positive association of AR at mRNA and protein level (p < 0.0001 for all samples together, p = 0.003 for females and p < 0.0001 for males) (Table 2, Figure S1).

3.2. Tumor AR Protein Levels Are Positively Associated with Patient Overall Survival

The sex difference in survival is well known for melanoma. In order to examine whether AR plays a role in such sex difference, melanoma patients are grouped by their tumor AR protein levels. “AR-high” group of patients have tumor AR levels greater than median AR (−0.718) for the entire cohort, while “AR-low” group of patients have tumor AR levels lower than the median AR. Our initial Kaplan–Meier survival analysis suggested that higher AR levels were associated with better OS (Figure 1), and this result seemed true for all patients (log rank test p = 0.0025), for male patients (p = 0.046), or for female patients (p = 0.0107) (Figure 1a–c). Cox regression analysis (simple variate analysis) further revealed that higher AR levels were significantly associated with better OS in females (p = 0.012) or for all patients (p = 0.003), and the significance level was reduced to 0.047 (p value) in male patients (Table 3). Cox proportional assumption was tested based on the Schoenfeld residues, and a p value of 0.24 was returned, indicating Cox analysis was a proper method for survival analysis for this dataset, which is consistent with our previous report [24]. In this dataset, sex alone was not a significant determinant for overall survival (Cox regression HR = 1.14, p = 0.39) (Table A1).
In the multivariate analysis, sex was used as a co-variable, and the AR association with overall survival was additionally adjusted by age of diagnosis, stage of disease (either stage 0 to stage 4 or early and late stages, as described in Section 2) (Table 3, Model 1–3). The association of AR with overall survival stayed significant after adjusting to these factors. When the presence of ulceration was added in the multivariate analysis, the association lost its significance (HR = 0.80, p = 0.29) (Table 3, Model 4). Interestingly, ulceration alone was significantly associated with overall survival (HR = 1.8, p = 0.001), and the significance remained after adjusting to age of diagnosis, stage of disease, and sex (HR = 1.46, p = 0.04).
These results may suggest an impact of AR on the ulceration status. However, tumors with or without ulceration did not show significant difference in the AR protein levels (p = 0.23 in Student t-test).
Another important prognostic factor for melanoma survival is Breslow depth. We grouped Breslow depth according to the AJCC TNM staging standards (1 = <1 mm; 2 = 1.01–2 mm; 3 = 2.01–4 mm; 4 = >4 mm) and included this variable in our multivariate COX analysis. The AR association with overall survival remained significant when the result was adjusted to Breslow depth, along with other factors (HR = 0.59, p = 0.008) (Table 3, Model 5).

3.3. The Sex Difference in the AR Association with OS

Table 4 shows that the AR protein level is not significantly associated with overall survival in men, even though the high AR is significantly associated with overall survival in women. Sex was then used as a stratification variable and the multivariate Cox models in male and female patients were analyzed separately, with age, stage, ulceration status, and Breslow depth as adjusting co-variables. AR levels were not associated with men’s OS in any of the models, but they are associated with women’s overall survival in all models except for Model 4 where ulceration was justified.
Since testosterone levels are known to change with men’s age, we also examined whether AR levels in tumors were different in older versus younger patients (≤50 vs. >50 years). A Student t-test was used to evaluate the AR protein levels, and no difference in means was found (p = 0.89 for men and 0.13 for women).

3.4. The Differential AR Gene Network in Tumors from Men and Women

In order to understand how AR expression levels are associated with patient overall survival in women but not in men, the TGCA SKCM mRNA data were used to extract the AR co-expressed genes using the online tool from the cBioportal website. The entire genome was included and the co-expressed genes were identified using a cutoff q value of q < 0.05. 6413 genes from men and 3384 genes from women were retained for further comparison. When the Spearman’s co-efficient for AR-association was set at ρ > 0.34, then 75 genes in women and 202 genes in men were retained for further comparison. Among these genes, 44 were unique for women, 171 were unique for men (Table A2), and 31 were shared by tumors from both sexes (Table A3, Figure S2). The 10 most significant genes for each sex are included in Table 5.
The 44 and 171 genes identified in the female and male tumors, respectively, are subjected to enrichment analysis using an integrated web-based tool termed g:profiler (https://biit.cs.ut.ee/gprofiler/gost, accessed on 14 June 2022). Genes were ordered All significant enrichments for females and partial of that for males are listed in Table 6. For female tumors, AR is significantly associated with GO:MF (molecular function), GO:CC (cellular component), and TF (transcription factor) functions. For male tumors, AR is significantly associated with a wide range of functions, including 99 GO:BP (biological process), 19 GO:CC, 10 GO:MF, 8 TF, 1 Reactome (Neurophilin interactions with VEGF and VEGFR), and 4 WP (Wikipathways). Only the top three significant functions in each category are shown in the table.
The shared 31 genes in male and female tumors were used for the same profiling analysis, 18 enriched functions were identified, which are listed in Table A4.

3.5. The Role of AR in Overall Survival in Four Melanoma Subtypes

The TCGA melanoma team classified this cohort of patients into four distinct subtypes with distinct somatic mutations in the tumors [25]. We obtained the classification information at patient level from their supplemental tables. A total of 316 patients were included in the analysis, but due to some tumors lacking AR protein data, only 230 patients were included in the survival analysis. Very interestingly, only in the RAS (mainly NRAS, but also including several mutants in KRAS and HRAS) mutants, did AR show significant association with overall survival (p = 0.013) (Table 7). The significance remained after adjusting to age of diagnosis and stage of disease (p = 0.047). When only sex is adjusted, the significant also remained (p = 0.022), but it was reduced to borderline (p = 0.057) when both sex and age are included.

4. Discussion

The findings of this study suggest that a higher level of tumor AR protein is positively associated with a better overall survival in cutaneous melanoma patients, which remains true after adjusting to age of diagnosis, stage of disease, sex of patients, and Breslow depth of the tumors. However, when patients are stratified by sex, the significant association was found only in female patients, but not in male patients, even though sex itself is not significantly associated with overall survival in this dataset. Additionally, when ulceration status is included in the model, the significance of AR association with OS was lost, suggesting that ulceration is still the most effective prognostic factor for melanoma OS. A statistical test of an interaction of AR with ulceration status revealed only borderline significance (p = 0.10, not shown in results). Nevertheless, our finding is significant, as this is one of the first studies to show an association of tumor AR level with overall survival in melanoma patients.
A previous report suggested an opposite role of AR in melanoma patient survival, i.e., higher AR was associated with worse survival [4]. That report did not specify melanoma subtype. The samples were collected in China, while the melanoma subtype in China is different than that in US—Chinese melanoma cases are mostly acral melanoma, which are distinct in oncogenic causes and pathological pathways than the US cases, which are mostly superficial spreading melanoma [26,27].
For melanoma, similar to many other cancer types, females in general show a survival advantage even after adjusting to many other prognostic factors. The underlying mechanism may be multi-fold, and we have been interested in the roles of sex hormones in such situations. Sex hormones and their receptors play critical roles in many pathophysiological conditions and impact many oncogenic pathways and cellular functions. AR was recently studied in melanoma cells, with a function of promoting proliferation, tumorigenesis, metastasis, and drug resistance [2,3,4], which is opposite to our findings.
The possible explanation may be directly linked to the androgen levels as the majority function of AR is linked to locally available testosterone and dihydrotestosterone. Therefore, in most cases, we must study the function of AR/T or AR/DHT together. It is particularly important to study the sex-specific impact, as men and women are distinctly different in the circulating T or DHT levels. Our study also showed a distinct gene network in tumors from male and female patients, further strengthening the importance of sex-specific investigation. Another possible reason is related to how to interpret the data. In one study, loss of AR led to more DNA damage [2], suggesting that AR played a protective role in genome integrity. When this occurs in normal melanocytes, one would expect AR serves as a tumor suppressor, as it was found in a subset of breast cancer [28]. This is also what our study suggests.
It is also noticeable that AR plays distinct functions in the male and female tumors, with shared functions in both sexes. The enriched functions are much broader in male tumors, indicating male-biased significance of AR. Since AR is involved in many more gene networks in males, the ability of these functions to maintain a relative cellular balance may be strengthened, which may help to explain why AR in men did not show a significant association with overall survival.
The weakness of this study is that we used only the TCGA data, with no replicating dataset. Therefore, this study requires further validation in a different patient cohort. As noted in one of our previous study [24], the patient sex did not show a significant association with OS, which is not the usual case for melanoma patients. That is the limitation of the patient cohort as well, and requires further replication.

5. Conclusions

The overall conclusion of this study is that tumor AR protein levels are associated with better OS in female patients, and not in male patients. We have also identified shared and distinct AR-associated gene networks in male and female tumors, which suggests AR exhibits common function in all tumors, and also exhibits distinct function in tumors from male and female patients. This study is the first to include data from a large database source with over 350 data points from melanoma patients’ tumors. Most of previous studies of AR in melanoma suggested that AR promoted tumor proliferation, metastasis, and drug resistance. Our study suggests that the role of AR should be considered in sex-specific manner, and in females, AR could be protective. Further investigation on these shared and distinct functions of AR in melanoma patients will help us to develop precise treatment strategies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14020345/s1, Figure S1: The RPPA protein levels (Y axis, normally distributed) were plotted against log-transformed AR mRNA (RSEM readings). The red line is regression fitting line. Figure S2. Venn diagram of the shared and distinct AR-co-expressed genes in male and female tumors.

Author Contributions

Conceptualization, F.L.-S. and T.S.P.; methodology, F.L.-S. and C.-Y.C.; validation, C.-Y.C.; formal analysis, F.L.-S., J.L. and C.-Y.C.; investigation, N.S. and J.K. resources, F.L.-S.; data curation, F.L.-S.; writing—original draft preparation, N.S. and J.K.; writing—review and editing, F.L.-S. and T.S.P.; visualization, F.L.-S.; supervision, F.L.-S. and T.S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data used in this study are publicly available; the sites are listed in the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Sex difference in overall survival.
Table A1. Sex difference in overall survival.
VariablesHR95%Conf.p Value
Model 1AR0.650.470.900.009
age1.031.021.040
sex1.120.811.560.495
Model 2AR0.670.480.950.025
age1.021.011.040
sex0.950.671.340.763
stage (0–4)1.391.141.700.001
Model 3AR0.680.480.960.029
age1.031.011.040
sex0.940.661.330.733
stage (early, late)1.691.192.400.003
Model 4AR0.800.541.200.286
age1.021.011.040.001
sex0.880.581.320.536
stage (early, late)1.841.212.790.004
ulceration1.440.932.220.101
Model 5AR0.590.400.870.008
age1.021.011.040.001
sex0.900.611.340.615
stage (early, late)1.511.012.250.044
Breslow depth1.501.221.850
Table A2. Sex-specific AR co-expressed genes in tumors.
Table A2. Sex-specific AR co-expressed genes in tumors.
GeneSpearman’s Coefficientp Valueq ValueSexApproved Gene NameHGNC IDCytoband
USP470.351.50 × 10−60.000403Fubiquitin specific peptidase 47HGNC:2007611p15.3
CTR90.328.70 × 10−60.000864FCTR9 homolog, Paf1/RNA polymerase II complex componentHGNC:1685011p15.4
THAP120.321.20 × 10−50.001011FTHAP domain containing 12HGNC:944011q13.5
ZNF1430.321.60 × 10−50.001217Fzinc finger protein 143HGNC:1292811p15.4
COPB10.311.70 × 10−50.001233FCOPI coat complex subunit β 1HGNC:223111p15.2
ASB120.310.000020.001376Fankyrin repeat and SOCS box containing 12HGNC:19763Xq11.2
THAP7-AS1−0.312.10 × 10−50.001378FTHAP7 antisense RNA 1HGNC:4101322q11.21
NR2F1-AS10.312.20 × 10−50.001426FNR2F1 antisense RNA 1HGNC:486225q15
MED170.312.70 × 10−50.00163Fmediator complex subunit 17HGNC:237511q21
TOM1−0.33.30 × 10−50.001835Ftarget of myb1 membrane trafficking proteinHGNC:1198222q12.3
C11ORF95/ZFTA0.33.50 × 10−50.001909Fzinc finger translocation associatedHGNC:2844911q13.1
HECTD2-AS1−0.30.000040.002025FHECTD2 antisense RNA 1HGNC:4867910q23.32
NDUFB8−0.34.20 × 10−50.002074FNADH:ubiquinone oxidoreductase subunit B8HGNC:770310q24.31
MARCH8/MARCHF80.431.00 × 10−142.40 × 10−11Mmembrane associated ring-CH-type finger 8HGNC:2335610q11.21-q11.22
RNF1520.393.80 × 10−122.00 × 10−9Mring finger protein 152HGNC:2681118q21.33
GDF100.382.00 × 10−116.30 × 10−9Mgrowth differentiation factor 10HGNC:421510q11.22
PLXNA40.374.90 × 10−111.20 × 10−8Mplexin A4HGNC:91027q32.3
ZNHIT2−0.361.40 × 10−102.50 × 10−8Mzinc finger HIT-type containing 2HGNC:117711q13.1
ZC4H20.362.50 × 10−104.00 × 10−8Mzinc finger C4H2-type containingHGNC:24931Xq11.2
NDST20.355.70 × 10−107.50 × 10−8MN-deacetylase and N-sulfotransferase 2HGNC:768110q22.2
TCHH0.356.90 × 10−108.60 × 10−8MtrichohyalinHGNC:117911q21.3
PCDHB60.359.70 × 10−101.10 × 10−7Mprotocadherin β 6HGNC:86915q31.3
LBHD1−0.351.10 × 10−91.20 × 10−7MLBH domain containing 1HGNC:2835111q12.3
SHISA60.351.10 × 10−91.20 × 10−7Mshisa family member 6HGNC:3449117p12
LRRC550.351.20 × 10−91.30 × 10−7Mleucine rich repeat containing 55HGNC:3232411q12.1
TIMM10−0.351.20 × 10−91.30 × 10−7Mtranslocase of inner mitochondrial membrane 10HGNC:1181411q12.1
PTPRD0.342.30 × 10−92.10 × 10−7Mprotein tyrosine phosphatase receptor type DHGNC:96689p24.1-p23
CDH230.342.70 × 10−92.40 × 10−7Mcadherin related 23HGNC:1373310q22.1
CDH80.342.80 × 10−92.40 × 10−7Mcadherin 8HGNC:176716q21
DPYSL20.342.90 × 10−92.50 × 10−7Mdihydropyrimidinase like 2HGNC:30148p21.2
PCDHB18P0.343.10 × 10−92.60 × 10−7Mprotocadherin β 18 pseudogeneHGNC:145485q31.3
CDC37−0.343.20 × 10−92.60 × 10−7Mcell division cycle 37, HSP90 cochaperoneHGNC:173519p13.2
SSTR10.343.40 × 10−92.80 × 10−7Msomatostatin receptor 1HGNC:1133014q21.1
PCDHB100.343.70 × 10−93.00 × 10−7Mprotocadherin β 10HGNC:86815q31.3
EDNRA0.344.10 × 10−93.30 × 10−7Mendothelin receptor type AHGNC:31794q31.22-q31.23
EPB41L4A-DT0.335.10 × 10−93.80 × 10−7MEPB41L4A divergent transcriptHGNC:256435q22.2
BACH20.335.80 × 10−94.10 × 10−7MBTB domain and CNC homolog 2HGNC:140786q15
SEC24A0.335.80 × 10−94.10 × 10−7MSEC24 homolog A, COPII coat complex componentHGNC:107035q31.1
ZNF4230.335.90 × 10−94.10 × 10−7Mzinc finger protein 423HGNC:1676216q12.1
STAT30.337.00 × 10−94.90 × 10−7Msignal transducer and activator of transcription 3HGNC:1136417q21.2
C12ORF45−0.337.10 × 10−94.90 × 10−7MNOP protein chaperone 1HGNC:2862812q23.3
GALNT170.337.30 × 10−95.00 × 10−7Mpolypeptide N-acetylgalactosaminyltransferase 17HGNC:163477q11.22
OLFML2B0.337.50 × 10−95.10 × 10−7Molfactomedin like 2BHGNC:245581q23.3
TMEM69−0.338.10 × 10−95.40 × 10−7Mtransmembrane protein 69HGNC:280351p34.1
MRPL16−0.339.60 × 10−96.20 × 10−7Mmitochondrial ribosomal protein L16HGNC:1447611q12.1
CYP4X10.331.00 × 10−86.50 × 10−7Mcytochrome P450 family 4 subfamily X member 1HGNC:202441p33|1
EPHB10.331.00 × 10−86.40 × 10−7MEPH receptor B1HGNC:33923q22.2
METTL17−0.331.00 × 10−86.40 × 10−7Mmethyltransferase like 17HGNC:1928014q11.2
SYT150.331.00 × 10−86.50 × 10−7Msynaptotagmin 15HGNC:1716710q11.22
LMOD10.331.10 × 10−86.70 × 10−7Mleiomodin 1HGNC:66471q32.1
PCDHGA50.331.30 × 10−87.60 × 10−7Mprotocadherin γ subfamily A, 5HGNC:87035q31.3
SUCNR10.331.30 × 10−87.60 × 10−7Msuccinate receptor 1HGNC:45423q25.1
MTMR120.321.50 × 10−88.50 × 10−7Mmyotubularin related protein 12HGNC:181915p13.3
FGF100.321.60 × 10−89.20 × 10−7Mfibroblast growth factor 10HGNC:36665p12
NLGN4Y0.321.60 × 10−89.00 × 10−7Mneuroligin 4 Y-linkedHGNC:15529Yq11.221
PENK0.321.60 × 10−89.10 × 10−7MproenkephalinHGNC:88318q12.1
TMEM1300.321.60 × 10−89.20 × 10−7Mtransmembrane protein 130HGNC:254297q22.1
ZNF7780.321.60 × 10−89.10 × 10−7Mzinc finger protein 778HGNC:2647916q24.3
PBX10.321.80 × 10−80.000001MPBX homeobox 1HGNC:86321q23.3
COA4−0.321.90 × 10−81.10 × 10−6Mcytochrome c oxidase assembly factor 4 homologHGNC:2460411q13.4
NPNT0.321.90 × 10−81.10 × 10−6MnephronectinHGNC:274054q24
GPR200.322.10 × 10−81.10 × 10−6MG protein-coupled receptor 20HGNC:44758q24.3
TSPAN180.322.10 × 10−81.10 × 10−6Mtetraspanin 18HGNC:2066011p11.2
SLITRK40.322.20 × 10−81.20 × 10−6MSLIT and NTRK like family member 4HGNC:23502Xq27.3
UQCR11−0.322.30 × 10−81.20 × 10−6Mubiquinol-cytochrome c reductase, complex III subunit XIHGNC:3086219p13.3
NFATC30.322.50 × 10−81.30 × 10−6Mnuclear factor of activated T cells 3HGNC:777716q22.1
TRMT112−0.322.50 × 10−81.30 × 10−6MtRNA methyltransferase activator subunit 11-2HGNC:2694011q13.1
EPHA70.323.50 × 10−81.70 × 10−6MEPH receptor A7HGNC:33906q16.1
ATP8B10.323.60 × 10−81.80 × 10−6MATPase phospholipid transporting 8B1HGNC:370618q21.31
FLT40.323.60 × 10−81.80 × 10−6Mfms related receptor tyrosine kinase 4HGNC:37675q35.3
PTAFR0.313.80 × 10−81.90 × 10−6Mplatelet activating factor receptorHGNC:95821p35.3
MAGED4B0.314.30 × 10−80.000002MMAGE family member D4BHGNC:22880Xp11.22
NAP1L20.314.30 × 10−80.000002Mnucleosome assembly protein 1 like 2HGNC:7638Xq13.2
NEURL1B0.314.70 × 10−82.20 × 10−6Mneuralized E3 ubiquitin protein ligase 1BHGNC:354225q35.1
TMEM132E0.315.20 × 10−82.40 × 10−6Mtransmembrane protein 132EHGNC:2699117q12
MRPL21−0.315.60 × 10−82.50 × 10−6Mmitochondrial ribosomal protein L21HGNC:1447911q13.3
SAP30L0.315.70 × 10−82.50 × 10−6MSAP30 likeHGNC:256635q33.2
ILDR20.316.40 × 10−82.80 × 10−6Mimmunoglobulin like domain containing receptor 2HGNC:181311q24.1
SLCO3A10.317.00 × 10−80.000003Msolute carrier organic anion transporter family member 3A1HGNC:1095215q26.1
LAMTOR5−0.317.10 × 10−80.000003Mlate endosomal/lysosomal adaptor, MAPK and MTOR activator 5HGNC:179551p13.3
MMRN20.317.10 × 10−80.000003Mmultimerin 2HGNC:1988810q23.2
ANK10.317.40 × 10−83.10 × 10−6Mankyrin 1HGNC:4928p11.21
UQCC3−0.317.60 × 10−83.20 × 10−6Mubiquinol-cytochrome c reductase complex assembly factor 3HGNC:3439911q12.3
OTULIN0.317.70 × 10−83.20 × 10−6MOTU deubiquitinase with linear linkage specificityHGNC:251185p15.2
UNC5C0.317.70 × 10−83.20 × 10−6Munc-5 netrin receptor CHGNC:125694q22.3
SCN2B0.317.90 × 10−83.20 × 10−6Msodium voltage-gated channel β subunit 2HGNC:1058911q23.3
CASTOR20.318.20 × 10−83.30 × 10−6Mcytosolic arginine sensor for mTORC1 subunit 2HGNC:370737q11.23
ARHGAP440.318.60 × 10−83.50 × 10−6MRho GTPase activating protein 44HGNC:2909617p12
COX8A−0.318.70 × 10−83.50 × 10−6Mcytochrome c oxidase subunit 8AHGNC:229411q13.1
FOXJ20.319.90 × 10−83.90 × 10−6Mforkhead box J2HGNC:2481812p13.31
ATP1A20.311.00 × 10−73.90 × 10−6MATPase Na+/K+ transporting subunit α 2HGNC:8001q23.2
ZBTB40.311.00 × 10−70.000004Mzinc finger and BTB domain containing 4HGNC:2384717p13.1
SEZ6L0.31.20 × 10−74.50 × 10−6Mseizure related 6 homolog likeHGNC:1076322q12.1
SATB10.31.30 × 10−74.90 × 10−6MSATB homeobox 1HGNC:105413p24.3
PCDHA90.31.40 × 10−75.10 × 10−6Mprotocadherin α 9HGNC:86755q31.3
SCN3B0.31.40 × 10−74.90 × 10−6Msodium voltage-gated channel β subunit 3HGNC:2066511q24.1
TMEM223−0.31.40 × 10−70.000005Mtransmembrane protein 223HGNC:2846411q12.3
ABCA80.31.50 × 10−75.20 × 10−6MATP binding cassette subfamily A member 8HGNC:3817q24.2
PCDH120.31.50 × 10−75.20 × 10−6Mprotocadherin 12HGNC:86575q31.3
ZNF4360.31.50 × 10−75.20 × 10−6Mzinc finger protein 436HGNC:208141p36.12
RELN0.31.60 × 10−75.60 × 10−6MreelinHGNC:99577q22.1
PGM50.31.70 × 10−75.80 × 10−6Mphosphoglucomutase 5HGNC:89089q21.11
SLC25A22−0.31.70 × 10−75.70 × 10−6Msolute carrier family 25 member 22HGNC:1995411p15.5
Table A3. AR-co-expressed genes in tumors in both sexes.
Table A3. AR-co-expressed genes in tumors in both sexes.
GeneSpearman’s_Menp_Menq_MenSpearman_Womenp_Womenq_WomenApproved NameHGNC IDLocation
NHSL20.531.2 × 10−222.4 × 10−180.3812651.3 × 10−70.000108NHS like 2HGNC:33737Xq13.1
ADAMTS120.486.7 × 10−184.5 × 10−140.419314.7 × 10−90.000025ADAM metallopeptidase with thrombospondin type 1 motif 12HGNC:146055p13.3-p13.2
RUNX1T10.441.7 × 10−155.8 × 10−120.3921575.2 × 10−80.000065RUNX1 partner transcriptional co-repressor 1HGNC:15358q21.3
ZNF3660.442.8 × 10−158 × 10−120.3448862.1 × 10−60.000458zinc finger protein 366HGNC:183165q13.1
FBN10.431.1 × 10−142.4 × 10−110.4143727.3 × 10−90.000025fibrillin 1HGNC:360315q21.1
LAMA20.432.4 × 10−144.2 × 10−110.3705033.1 × 10−70.000181laminin subunit α 2HGNC:64826q22.33
CDKL50.424.2 × 10−146 × 10−110.3407642.9 × 10−60.000514cyclin dependent kinase like 5HGNC:11411Xp22.13
MAN2A10.428.3 × 10−141 × 10−100.351741.3 × 10−60.000378mannosidase α class 2A member 1HGNC:68245q21.3
LTBP20.428.5 × 10−141 × 10−100.3521131.3 × 10−60.000378latent transforming growth factor β binding protein 2HGNC:671514q24.3
TSHZ20.412.2 × 10−132 × 10−100.345212.1 × 10−60.000458teashirt zinc finger homeobox 2HGNC:1301020q13.2
PPM1L0.413 × 10−132.5 × 10−100.3490591.6 × 10−60.000416protein phosphatase, Mg2+/Mn2+ dependent 1LHGNC:163813q25.33-q26.1
REST0.413.9 × 10−133 × 10−100.3591027.4 × 10−70.000303RE1 silencing transcription factorHGNC:99664q12
SVEP10.392.9 × 10−121.7 × 10−90.4157566.5 × 10−90.000025sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1HGNC:159859q31.3
SON0.396.4 × 10−123 × 10−90.3437582.3 × 10−60.00047SON DNA and RNA binding proteinHGNC:1118321q22.11
JCAD0.398.3 × 10−123.4 × 10−90.343322.4 × 10−60.000472junctional cadherin 5 associatedHGNC:2928310p11.23
SLIT30.398.6 × 10−123.5 × 10−90.3417092.7 × 10−60.000496slit guidance ligand 3HGNC:110875q34-q35.1
PEAK10.381.4 × 10−115.1 × 10−90.3639215.1 × 10−70.000249pseudopodium enriched atypical kinase 1HGNC:2943115q24.3
TMEM200A0.381.4 × 10−115.1 × 10−90.3688473.5 × 10−70.000194transmembrane protein 200AHGNC:210756q23.1
SLC12A60.381.7 × 10−115.7 × 10−90.3723622.6 × 10−70.000173solute carrier family 12 member 6HGNC:1091415q14
PGR0.382.2 × 10−116.6 × 10−90.3528751.2 × 10−60.000377progesterone receptorHGNC:891011q22.1
CCDC800.383.5 × 10−119.1 × 10−90.4072691.4 × 10−80.000029coiled-coil domain containing 80HGNC:306493q13.2
AKAP130.374.3 × 10−111.1 × 10−80.3474551.8 × 10−60.000437A-kinase anchoring protein 13HGNC:37115q25.3
SELENOP0.375.6 × 10−111.3 × 10−80.3577338.2 × 10−70.000311selenoprotein PHGNC:107515p12
KAT6A0.362.6 × 10−104 × 10−80.3441762.2 × 10−60.000468lysine acetyltransferase 6AHGNC:130138p11.21
BICC10.362.6 × 10−104.1 × 10−80.4038041.9 × 10−80.000032BicC family RNA binding protein 1HGNC:1935110q21.1
FHL10.364 × 10−105.7 × 10−80.3595187.2 × 10−70.000303four and a half LIM domains 1HGNC:3702Xq26.3
SETD70.357.3 × 10−108.9 × 10−80.3497841.5 × 10−60.000403SET domain containing 7, histone lysine methyltransferaseHGNC:304124q31.1
DCN0.351 × 10−91.2 × 10−70.3436842.3 × 10−60.00047decorinHGNC:270512q21.33
OGN0.351.3 × 10−91.4 × 10−70.3557989.5 × 10−70.000339osteoglycinHGNC:81269q22.31
PDGFRA0.342 × 10−91.9 × 10−70.3451072.1 × 10−60.000458platelet derived growth factor receptor αHGNC:88034q12
TTBK20.342.2 × 10−92.1 × 10−70.3489581.6 × 10−60.000416tau tubulin kinase 2HGNC:1914115q15.2
Table A4. Enriched function using AR-co-expressed genes in both sexes.
Table A4. Enriched function using AR-co-expressed genes in both sexes.
SourceTerm_NameTerm_IDAdjusted_p_ValueTerm_SizeQuery_SizeIntersection_SizeIntersections
GO:MFextracellular matrix structural constituentGO:00052010.001116174275FBN1, LAMA2, OGN, LTBP2, DCN
GO:MFglycosaminoglycan bindingGO:00055390.009894244305FBN1, CCDC80, LTBP2, DCN, SLIT3
GO:MFheparin bindingGO:00082010.043013173304FBN1, CCDC80, LTBP2, SLIT3
GO:BPcellular response to vascular endothelial growth factor stimulusGO:00359240.00253562294ADAMTS12, PDGFRA, DCN, JCAD
GO:BPanatomical structure morphogenesisGO:00096530.00287727223115ADAMTS12, SVEP1, FBN1, SLC12A6, LAMA2, PEAK1, FHL1, PGR, MAN2A1, AKAP13, PDGFRA, DCN, JCAD, SLIT3, CDKL5
GO:BPcirculatory system developmentGO:00723590.00295711093010SVEP1, FBN1, BICC1, SLC12A6, REST, AKAP13, PDGFRA, DCN, JCAD, SLIT3
GO:BPdevelopmental processGO:00325020.00659964243122ADAMTS12, SVEP1, FBN1, BICC1, RUNX1T1, NHSL2, SLC12A6, LAMA2, PEAK1, FHL1, REST, SELENOP, PGR, MAN2A1, TTBK2, AKAP13, PDGFRA, KAT6A, DCN, JCAD, SLIT3, CDKL5
GO:BPregulation of cellular response to vascular endothelial growth factor stimulusGO:19025470.00677123293ADAMTS12, DCN, JCAD
GO:BPsystem developmentGO:00487310.00985743693118ADAMTS12, SVEP1, FBN1, BICC1, SLC12A6, LAMA2, REST, SELENOP, PGR, MAN2A1, TTBK2, AKAP13, PDGFRA, KAT6A, DCN, JCAD, SLIT3, CDKL5
GO:BPanatomical structure developmentGO:00488560.03384558363120ADAMTS12, SVEP1, FBN1, BICC1, SLC12A6, LAMA2, PEAK1, FHL1, REST, SELENOP, PGR, MAN2A1, TTBK2, AKAP13, PDGFRA, KAT6A, DCN, JCAD, SLIT3, CDKL5
GO:BPmulticellular organism developmentGO:00072750.04213948233118ADAMTS12, SVEP1, FBN1, BICC1, SLC12A6, LAMA2, REST, SELENOP, PGR, MAN2A1, TTBK2, AKAP13, PDGFRA, KAT6A, DCN, JCAD, SLIT3, CDKL5
GO:BPvascular endothelial growth factor signaling pathwayGO:00380840.04312842293PDGFRA, DCN, JCAD
GO:BPcell adhesionGO:00071550.044906152144ADAMTS12, SVEP1, FBN1, CCDC80
GO:CCextracellular matrixGO:00310120.000983565186ADAMTS12, FBN1, CCDC80, LAMA2, OGN, LTBP2
GO:CCexternal encapsulating structureGO:00303120.000993566186ADAMTS12, FBN1, CCDC80, LAMA2, OGN, LTBP2
GO:CCbasement membraneGO:00056040.0017269993FBN1, CCDC80, LAMA2
GO:CCcollagen-containing extracellular matrixGO:00620230.002734429276FBN1, CCDC80, LAMA2, OGN, LTBP2, DCN
HPMicrospherophakiaHP:00309610.0450023182FBN1, LTBP2

References

  1. Saginala, K.; Barsouk, A.; Aluru, J.S.; Rawla, P.; Barsouk, A. Epidemiology of Melanoma. Med. Sci. 2021, 9, 63. [Google Scholar] [CrossRef]
  2. Ma, M.; Ghosh, S.; Tavernari, D.; Katarkar, A.; Clocchiatti, A.; Mazzeo, L.; Samarkina, A.; Epiney, J.; Yu, Y.R.; Ho, P.C.; et al. Sustained androgen receptor signaling is a determinant of melanoma cell growth potential and tumorigenesis. J. Exp. Med. 2021, 218, e20201137. [Google Scholar] [CrossRef] [PubMed]
  3. Vellano, C.P.; White, M.G.; Andrews, M.C.; Chelvanambi, M.; Witt, R.G.; Daniele, J.R.; Titus, M.; McQuade, J.L.; Conforti, F.; Burton, E.M.; et al. Androgen receptor blockade promotes response to BRAF/MEK-targeted therapy. Nature 2022, 606, 797–803. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, Y.; Ou, Z.; Sun, Y.; Yeh, S.; Wang, X.; Long, J.; Chang, C. Androgen receptor promotes melanoma metastasis via altering the miRNA-539-3p/USP13/MITF/AXL signals. Oncogene 2017, 36, 1644–1654. [Google Scholar] [CrossRef] [PubMed]
  5. Reintgen, D.S.; Paull, D.E.; Seigler, H.F.; Cox, E.B.; McCarty, K.S., Jr. Sex related survival differences in instances of melanoma. Surg. Gynecol. Obstet. 1984, 159, 367–372. [Google Scholar] [PubMed]
  6. Micheli, A.; Ciampichini, R.; Oberaigner, W.; Ciccolallo, L.; de Vries, E.; Izarzugaza, I.; Zambon, P.; Gatta, G.; De Angelis, R.; Group, E.W. The advantage of women in cancer survival: An analysis of EUROCARE-4 data. Eur. J. Cancer 2009, 45, 1017–1027. [Google Scholar] [CrossRef]
  7. Brown, C.J.; Goss, S.J.; Lubahn, D.B.; Joseph, D.R.; Wilson, E.M.; French, F.S.; Willard, H.F. Androgen receptor locus on the human X chromosome: Regional localization to Xq11-12 and description of a DNA polymorphism. Am. J. Hum. Genet. 1989, 44, 264–269. [Google Scholar]
  8. Ju, Q.; Tao, T.; Hu, T.; Karadag, A.S.; Al-Khuzaei, S.; Chen, W. Sex hormones and acne. Clin. Dermatol. 2017, 35, 130–137. [Google Scholar] [CrossRef] [PubMed]
  9. Bienenfeld, A.; Azarchi, S.; Lo Sicco, K.; Marchbein, S.; Shapiro, J.; Nagler, A.R. Androgens in women: Androgen-mediated skin disease and patient evaluation. J. Am. Acad. Dermatol. 2019, 80, 1497–1506. [Google Scholar] [CrossRef]
  10. Slominski, A.; Zbytek, B.; Nikolakis, G.; Manna, P.R.; Skobowiat, C.; Zmijewski, M.; Li, W.; Janjetovic, Z.; Postlethwaite, A.; Zouboulis, C.C.; et al. Steroidogenesis in the skin: Implications for local immune functions. J. Steroid. Biochem. Mol. Biol. 2013, 137, 107–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Palvimo, J.J.; Reinikainen, P.; Ikonen, T.; Kallio, P.J.; Moilanen, A.; Janne, O.A. Mutual transcriptional interference between RelA and androgen receptor. J. Biol. Chem. 1996, 271, 24151–24156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sato, N.; Sadar, M.D.; Bruchovsky, N.; Saatcioglu, F.; Rennie, P.S.; Sato, S.; Lange, P.H.; Gleave, M.E. Androgenic induction of prostate-specific antigen gene is repressed by protein-protein interaction between the androgen receptor and AP-1/c-Jun in the human prostate cancer cell line LNCaP. J. Biol. Chem. 1997, 272, 17485–17494. [Google Scholar] [CrossRef] [Green Version]
  13. Wilkenfeld, S.R.; Lin, C.; Frigo, D.E. Communication between genomic and non-genomic signaling events coordinate steroid hormone actions. Steroids 2018, 133, 2–7. [Google Scholar] [CrossRef]
  14. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  15. Yang, L.; Zheng, R.; Wang, N.; Yuan, Y.; Liu, S.; Li, H.; Zhang, S.; Zeng, H.; Chen, W. Incidence and mortality of stomach cancer in China, 2014. Chin. J. Cancer Res. 2018, 30, 291–298. [Google Scholar] [CrossRef] [PubMed]
  16. Uhlenhopp, D.J.; Then, E.O.; Sunkara, T.; Gaduputi, V. Epidemiology of esophageal cancer: Update in global trends, etiology and risk factors. Clin. J. Gastroenterol. 2020, 13, 1010–1021. [Google Scholar] [CrossRef] [PubMed]
  17. Sayiner, M.; Golabi, P.; Younossi, Z.M. Disease Burden of Hepatocellular Carcinoma: A Global Perspective. Dig. Dis. Sci. 2019, 64, 910–917. [Google Scholar] [CrossRef]
  18. Saginala, K.; Barsouk, A.; Aluru, J.S.; Rawla, P.; Padala, S.A.; Barsouk, A. Epidemiology of Bladder Cancer. Med. Sci. 2020, 8, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Yuan, Y.; Liu, L.; Chen, H.; Wang, Y.; Xu, Y.; Mao, H.; Li, J.; Mills, G.B.; Shu, Y.; Li, L.; et al. Comprehensive Characterization of Molecular Differences in Cancer between Male and Female Patients. Cancer Cell 2016, 29, 711–722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Schmidt, K.; Carroll, J.S.; Yee, E.; Thomas, D.D.; Wert-Lamas, L.; Neier, S.C.; Sheynkman, G.; Ritz, J.; Novina, C.D. The lncRNA SLNCR Recruits the Androgen Receptor to EGR1-Bound Genes in Melanoma and Inhibits Expression of Tumor Suppressor p21. Cell Rep. 2019, 27, 2493–2507.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Clocchiatti, A.; Ghosh, S.; Procopio, M.G.; Mazzeo, L.; Bordignon, P.; Ostano, P.; Goruppi, S.; Bottoni, G.; Katarkar, A.; Levesque, M.; et al. Androgen receptor functions as transcriptional repressor of cancer-associated fibroblast activation. J. Clin. Investig. 2018, 128, 5531–5548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Li, B.; Dewey, C.N. RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011, 12, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. De Siqueira Santos, S.; Takahashi, D.Y.; Nakata, A.; Fujita, A. A comparative study of statistical methods used to identify dependencies between gene expression signals. Brief. Bioinform. 2014, 15, 906–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Liu-Smith, F.; Lu, Y. Opposite Roles of BAP1 in Overall Survival of Uveal Melanoma and Cutaneous Melanoma. J. Clin. Med. 2020, 9, 411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cancer Genome Atlas, N. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [Green Version]
  26. Guo, J.; Qin, S.; Liang, J.; Lin, T.; Si, L.; Chen, X.; Chi, Z.; Cui, C.; Du, N.; Fan, Y.; et al. Chinese Guidelines on the Diagnosis and Treatment of Melanoma (2015 Edition). Ann. Transl. Med. 2015, 3, 322. [Google Scholar] [CrossRef]
  27. Hall, K.H.; Rapini, R.P. Acral Lentiginous Melanoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  28. Hickey, T.E.; Selth, L.A.; Chia, K.M.; Laven-Law, G.; Milioli, H.H.; Roden, D.; Jindal, S.; Hui, M.; Finlay-Schultz, J.; Ebrahimie, E.; et al. The androgen receptor is a tumor suppressor in estrogen receptor-positive breast cancer. Nat. Med. 2021, 27, 310–320. [Google Scholar] [CrossRef]
Figure 1. Kaplan–Meier survival curves for patients with high and low AR protein levels. (a) For both male and female patients; (b) for female patients; (c) for male patients (p values are derived from log rank test).
Figure 1. Kaplan–Meier survival curves for patients with high and low AR protein levels. (a) For both male and female patients; (b) for female patients; (c) for male patients (p values are derived from log rank test).
Genes 14 00345 g001
Table 1. Baseline characterization of patients.
Table 1. Baseline characterization of patients.
FemaleMaleMissing * Total
Number of patients1802901 (sex)471
Number of tumors1832961 (sex)480
Tumors with available AR RPPA data1442081 (sex)353
Tumors with available AR mRNA data1802891 (mRNA)471
Number of primary tumors45640109
Number of metastatic tumors1382321 (sex)371
Age at diagnosis (years)58.5 ± 1.258.0 ± 0.99 (age)58.2 ± 0.7
Stage of disease
stage 025 7
stage 12552 77
stage 26193 154
stage 367104 171
stage 4815 23
missing17211 (sex)39
total180290 471
Stage of disease **
early88150 238
late75119 194
missing17211 (sex)39
Ulceration
no5789 146
yes67100 167
missing 56101 158
Breslow depth
<1.0 mm2036 56
1.0–2.0 mm2753 80
2.0–4.0 mm3344 77
>4.0 mm6083 143
missing40741 (sex)115
* missing means the number of patients missing the corresponding data, e.g., first row, 1 (sex) means 1 patient missing sex information. ** stages 0–2 are defined as early stage, while stages 3–4 are late stage.
Table 2. The sex difference in AR gene expression.
Table 2. The sex difference in AR gene expression.
mRNAProtein
SexFemaleMaleTotalFemaleMaleTotal
N180289469144208352
Mean−0.077−0.053−0.062−0.744−0.665−0.691
Std.err.0.0650.0480.0390.0230.0190.015
Median−0.347−0.288−0.312−0.762−0.695−0.718
95% CI−0.205−0.148−0.138−0.789−0.703−0.727
0.0510.0410.013−0.698−0.627−0.668
p value (sex difference)0.77 0.0099
mRNA vs. protein (linear regression)Female: coefficient: 0.10 ± 0.03, p = 0.003
Male: coefficient: 0.11 ± 0.02, p < 0.0001
All: coefficient: 0.11 ± 0.017, p < 0.0001
Table 3. AR is significantly associated with overall survival in melanoma patients.
Table 3. AR is significantly associated with overall survival in melanoma patients.
AnalysisPatients/ModelHR95% CIp Value *Variable(s) Included
Simple variateFemale0.490.280.860.012AR
Male0.660.440.990.047AR
All0.610.440.840.003AR
MultivariateModel 10.650.470.900.009AR, sex, age
Model 20.670.480.950.025AR, sex, age, stage (0–4)
Model 30.680.480.960.029AR, sex, age, stage (early, late)
Model 40.800.541.200.29AR, sex, age, stage (early, late), ulceration
Model 50.590.400.870.008AR, sex, age, stage (early, late), Breslow depth (4 category)
*, p value: for AR.
Table 4. AR protein level for survival between female and male sexes.
Table 4. AR protein level for survival between female and male sexes.
FemaleMale
VariablesHR[95% Conf. Interval]p ValueHR[95% Conf. Interval]p Value
Model 1AR0.510.290.900.0210.700.471.060.092
age1.031.021.0501.021.011.040.003
Model 2AR0.490.270.890.020.800.521.240.328
age1.031.011.050.0011.021.001.030.028
stage (0–4)1.270.911.790.1651.461.141.870.003
Model 3AR0.480.260.870.0160.840.541.310.443
age1.031.011.0501.021.001.030.017
stage (early, late)1.360.772.420.2891.961.263.080.003
Model 4AR0.580.281.190.1351.020.601.730.951
age1.031.011.050.0091.021.001.040.106
Stage (early, late)1.500.772.910.232.161.263.710.005
ulceration1.280.642.580.4891.600.912.810.1
Model 5AR0.490.250.960.0390.650.391.090.102
age1.031.011.050.0041.021.001.040.088
Stage (early, late)1.580.862.900.1431.380.792.410.258
Breslow Depth1.270.921.740.1491.711.282.280
Table 5. Top 10 most significant sex-specific AR co-expressed genes in tumors.
Table 5. Top 10 most significant sex-specific AR co-expressed genes in tumors.
GeneSpearman’s Coefficientp Valueq ValueSexApproved Gene NameHGNC IDLocation
KMT2A0.424.3 × 10−90.000025Flysine methyltransferase 2AHGNC:713211q23.3
NECTIN30.418.8 × 10−90.000025Fnectin cell adhesion molecule 3HGNC:176643q13.13
ROR10.411.4 × 10−80.000029Freceptor tyrosine kinase like orphan receptor 1HGNC:102561p31.3
MACF10.411.6 × 10−80.000029Fmicrotubule actin crosslinking factor 1HGNC:136641p34.3
CBL0.395.5 × 10−80.000065FCbl proto-oncogeneHGNC:154111q23.3
AKAP20.397 × 10−80.000078FA-kinase anchoring protein 2HGNC:3729q31.3
KERA0.397.5 × 10−80.00008FkeratocanHGNC:630912q21.33
PRDM100.398.3 × 10−80.000082FPR/SET domain 10HGNC:1399511q24.3
MAML20.389.9 × 10−80.00009Fmastermind like transcriptional coactivator 2HGNC:1625911q21
ZFP910.381 × 10−70.00009FZFP91 zinc finger protein, atypical E3 ubiquitin ligaseHGNC:1498311q12.1
SLIT20.497.6 × 10−197.7 × 10−15Mslit guidance ligand 2HGNC:110864p15.31
ITGA80.451.1 × 10−154.5 × 10−12Mintegrin subunit α 8HGNC:614410p13
PREX20.451.1 × 10−154.5 × 10−12Mphosphatidylinositol-3,4,5-trisphosphate dependent Rac exchange factor 2HGNC:229508q13.2
MARCHF80.431 × 10−142.4 × 10−11Mmembrane associated ring-CH-type finger 8HGNC:2335610q11.21-q11.22
RALGAPA20.431.3 × 10−142.6 × 10−11MRal GTPase activating protein catalytic subunit α 2HGNC:1620720p11.23
ZDHHC150.432.5 × 10−144.2 × 10−11Mzinc finger DHHC-type palmitoyltransferase 15HGNC:20342Xq13.3
IL6ST0.424 × 10−146 × 10−11Minterleukin 6 cytokine family signal transducerHGNC:60215q11.2
PCSK50.428.5 × 10−141 × 10−10Mproprotein convertase subtilisin/kexin type 5HGNC:87479q21.13
MAN1A10.429.3 × 10−141 × 10−10Mmannosidase α class 1A member 1HGNC:68216q22.31
ASXL30.421.2 × 10−131.3 × 10−10MASXL transcriptional regulator 3HGNC:2935718q12.1
Table 6. The sex-specific AR-associated enrichment of gene function.
Table 6. The sex-specific AR-associated enrichment of gene function.
SexSourceTerm_IdAdjusted_p_ValueTerm_SizeQuery_SizeIntersection_SizeTerm_Name
FemaleGO:MFGO:00428000.02883218142histone methyltransferase activity (H3-K4 specific)
GO:MFGO:01063630.042024211protein-cysteine methyltransferase activity
GO:CCGO:00432960.009441154274apical junction complex
TFTF:M09984_10.00703356964327Factor: MAZ; motif: GGGGGAGGGGGNGRGRRRGNRG; match class: 1
TFTF:M12654_10.0323914432Factor: PRDM15; motif: NYCCRNTCCRGGTTTTSC; match class: 1
TFTF:M09834_10.03279929503917Factor: ZNF148; motif: NNNNNNCCNNCCCCTCCCCCACCCN; match class: 1
MaleGO:MFGO:00468725.32 × 10−6427113157metal ion binding
GO:MFGO:00055095.9 × 10−672613021calcium ion binding
GO:MFGO:00431691.2 × 10−5436413157cation binding
GO:BPGO:00487311.65 × 10−11436916378system development
GO:BPGO:00488566.64 × 10−11583616391anatomical structure development
GO:BPGO:00071552.2 × 10−10152116743cell adhesion
GO:CCGO:00058873.25 × 10−10164915641integral component of plasma membrane
GO:CCGO:00312263.56 × 10−10173115642intrinsic component of plasma membrane
GO:CCGO:00719443.05 × 10−8627016085cell periphery
REACREAC:R-HSA-1943060.0052734152Neurophilin interactions with VEGF and VEGFR
WPWP:WP48230.00402844113Genes controlling nephrogenesis
WPWP:WP39430.0040816112Robo4 and VEGF signaling pathways crosstalk
WPWP:WP50650.0051935152SARS-CoV-2 altering angiogenesis via NRP1
TFTF:M00695_13.82 × 10−87194169104Factor: ETF; motif: GVGGMGG; match class: 1
TFTF:M12345_10.0005217357422Factor: Zbtb37; motif: NYACCGCRNTCACCGCR; match class: 1
TFTF:M011990.0023768683169105Factor: RNF96; motif: BCCCGCRGCC
Table 7. Role of AR in overall survival in four subtypes of melanoma.
Table 7. Role of AR in overall survival in four subtypes of melanoma.
HR[95% Conf. Interval]p ValueN **
BRAF_Hotspot_Mutants0.620.201.150.336106
RAS_Hotspot_Mutants0.440.230.840.013 *67
NF1_Any_Mutants0.730.262.040.55125
Triple_WT0.970.392.450.95032
* p value for RAS subtype = 0.047 after adjusting to age and stage of patients. ** N: number of patients in each subtype included in survival analysis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Singh, N.; Khatib, J.; Chiu, C.-Y.; Lin, J.; Patel, T.S.; Liu-Smith, F. Tumor Androgen Receptor Protein Level Is Positively Associated with a Better Overall Survival in Melanoma Patients. Genes 2023, 14, 345. https://doi.org/10.3390/genes14020345

AMA Style

Singh N, Khatib J, Chiu C-Y, Lin J, Patel TS, Liu-Smith F. Tumor Androgen Receptor Protein Level Is Positively Associated with a Better Overall Survival in Melanoma Patients. Genes. 2023; 14(2):345. https://doi.org/10.3390/genes14020345

Chicago/Turabian Style

Singh, Nupur, Jude Khatib, Chi-Yang Chiu, Jianjian Lin, Tejesh Surender Patel, and Feng Liu-Smith. 2023. "Tumor Androgen Receptor Protein Level Is Positively Associated with a Better Overall Survival in Melanoma Patients" Genes 14, no. 2: 345. https://doi.org/10.3390/genes14020345

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop