A systematic analysis reveals gene expression alteration of serum deprivation response (SDPR) gene is significantly associated with the survival of patients with cancer

  • Authors:
    • Yingshuang Wang
    • Zhen Song
    • Ping Leng
    • Yun Liu
  • View Affiliations

  • Published online on: June 26, 2019     https://doi.org/10.3892/or.2019.7212
  • Pages: 1161-1172
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Serum deprivation response (SDPR) gene has been recently characterized as a gene signature marker or serving a tumor suppressor role in specific types of cancer. However, gene expression alterations of SDPR in various types of cancer and their relevance to clinical outcomes remain unclear. In the present study, SDPR expression was profiled using the Oncomine database, and SDPR downregulation was indicated in most types of cancer. In agreement with previously reported breast cancer cases, downregulation of SDPR was indicated to be significantly associated with poor survival in patients with lung cancer, glioma and sarcoma. To clarify why SDPR expression was altered in these types of cancer, both DNA methylation patterns and potential transcriptional factors of SDPR were analyzed. Altered DNA methylation levels of SDPR were found in 17/18 cancer types using MethHC. To the best of our knowledge, the present study for the first time, identified the CpG site (cg10082589) as one of the best survival methylation markers for lung adenocarcinoma, and the CpG site (cg07488576) for sarcoma using Methsurv. In addition, GATA binding protein 2 was identified as a potential transcription factor for SDPR, by integrating and analyzing both the co‑expressed genes and the potential transcription factor binding sites of SDPR. In the present study, the systematic analysis of SDPR provided insight for the underlying molecular mechanisms in cancer progression, revealing novel perspectives for the clinical prognostic evaluation of lung adenocarcinoma and sarcoma.

Introduction

Cancer has contributed to the rising rate in disease-associated mortality in the last few decades. The demand for early-stage diagnosis, prognosis and therapeutic targets is increasing. The serum deprivation response (SDPR) gene has been characterized to serve a critical role in breast cancer as a tumor suppressor (1,2), and recently showing a characteristic gene signature in specific types of cancer, including oral cancer, thyroid cancer and liposarcoma (35).

SDPR localizes to chr2q 32–33, also known as caveolae associated protein 2, and has been known for its role in caveolae formation (68). Its encoding protein SDPR, which is overexpressed in serum starved cells, was firstly identified as a substrate for protein kinase C (PKC) phosphorylation, an interaction that targets PKC in caveolae formation (9). Caveolae are plasma membrane microdomains involved in multiple biological processes, including lipid metabolism, endocytosis, cellular signal transduction, cell proliferation and migration (7,10).

Previous studies have gradually implicated the differential expression and tumor suppressor function of SDPR in cancer progression and metastasis; reduced SDPR expression has been observed in breast (1,2,1113), kidney (11,14) and prostate (11,15) cancer. In breast cancer, SDPR was identified as a tumor suppressor (1,2). SDPR serves an anti-metastatic function by promoting apoptosis (1), and depletion of SDPR induced epithelial-mesenchymal transition (EMT) through transforming growth factor-β (TGF-β) signaling activation (2). The reduction of SDPR expression has been reported to be associated with significantly reduced survival in patients with breast cancer, who underwent therapy (1).

In addition to breast cancer, it has been suggested that SDPR may serve as a tumor suppressor gene, with a broader clinical relevance, in other types of cancer. In oral cancer, it was identified that SDPR-negative patients had high tumor progression (5), whereas in sarcoma (SARC), a lower SDPR expression was observed in more aggressive or dedifferentiated tumor forms (4). In addition, it was suggested that the differently expressed SDPR gene can be used as a possible diagnostic marker to discriminate malignant tumors from benign formations, not only in serum from patients with kidney tumors (14), but also in follicular thyroid carcinomas (3). Nevertheless, gene expression alterations of SDPR in various types of cancer and their relevance to clinical outcomes remain unclear.

In the present study, mRNA levels of SDPR were compared in various unique tumor tissue datasets compared with normal tissue datasets, indicating that SDPR was downregulated in various types of cancer. In addition, downregulated SDPR was found to be significantly associated with the survival of the patients, not only in previously reported breast cancer cases, but also in brain, lung and soft tissue tumors. However, SDPR failed to emerge as a frequent target for gene mutational inactivation in a previous next-generation sequencing study (16). As SDPR has been reported to be hypermethylated and silenced in breast cancer cell lines, it was suggested that it is likely to be epigenetically inactivated in cancer (1). In order to examine the mechanism underlying SDPR downregulation in cancer, the present study analyzed and found SDPR gene methylation alteration between cancer and normal tissues. Furthermore, the present study investigated the methylation sites relevant to the survival of patients with lung adenocarcinoma (LUAD) and SARC. In addition, the potential transcription factor binding sites of the SDPR promoter were analyzed. The potential transcription factor GATA binding protein 2 (GATA2) was identified from the analysis of genes that are co-expressed with SDPR. The results of the present study provide additional insight in understanding the underlying molecular mechanisms of SDPR in cancer, in addition to revealing a novel approach in the clinical prognostic evaluation and treatment of LUAD and SARC.

Materials and methods

Gene expression analysis using oncomine platform

The profile of SDPR gene expression level in various types of cancer was identified in Oncomine™ Platform and the detailed datasets are available online (https://www.oncomine.org/) (17). By comparing with normal tissues, the mRNA expression-fold of SDPR in cancer tissues was obtained using the parameters of P-value <10−4 or 0.01, fold-change >2, and gene ranking in the top 10%. To adjust the false discovery rate, the P-values were corrected by using the Benjamini-Hochberg procedure (B-H method) (18) in R, version 3.5.0 (https://www.r-project.org/).

Prognoscan database analysis

The association between SDPR expression and survival in different types of cancer was analyzed using the PrognoScan database (http://www.abren.net/PrognoScan/) (19), and presented as a Kaplan-Meier plot, in which survival curves for high (red) and low (blue) expression groups dichotomized at the optimal cut-point were plotted. The P-values were adjusted for multiple correlation testing using the Miller and Siegmund formula (20), according to Prognoscan database and shown as a corrected P-value (Pcor). The threshold was adjusted to corrected P-values at <0.05.

MethHC database analysis

The MethHC database was used in the analysis of SDPR DNA methylation alternation in cancer. MethHC (http://MethHC.mbc.nctu.edu.tw/) is a systematic database integrating DNA methylation data from The Cancer Genome Atlas (TCGA; http://cancergenome.nih.gov/abouttcga/policies/informedconsent), which includes >6,000 DNA methylation data generated by Illumina HumanMethylation450K BeadChip in 18 types of cancer. The methylation status of DNA was represented as β-values (0–1) (21), and the average β-value of SDPR was presented as a boxplot by comparing the transcript expression in tumor samples and matched normal samples in all 18 types of cancer. To adjust the false discovery rate, the P-values were corrected using the B-H method in R software.

Methsurv database analysis

Methsurv database (https://biit.cs.ut.ee/methsurv) was utilized for survival analysis in different types of cancer based on SDPR methylation patterns. In Methsurv, the gene methylation data was from TCGA Genome Data Analysis Center Firehose (http://gdac.broadinstitute.org/) (22), using the HM450K array, which covers 486,428 CpGs. The methylation status of DNA was represented as β-values (0–1) (23). The methylation pattern was annotated by probes indicating subregions of the query gene, according to the annotation file (Human genome build 27) provided by Illumina [TSS to-200 nucleotides upstream of TSS (‘TS200’); covering-200 to-1500 nucleotides upstream of TSS (‘TSS1500’; first exon (‘1st exon’); ‘5′UTR’, ‘body’ and ‘3′UTR’]. Clustering analysis was plotted and visualized using a heatmap by integrating Methsurv settings with ClustVis (https://biit.cs.ut.ee/clustvis/) (24). The survival analysis of each type of cancer between the low-methylated and the high-methylated groups in specific methylation sites was visualized using Kaplan-Meier plots. Multivariable survival analysis was performed using a Cox proportional-hazards model. Age and sex were used as covariates in the multivariable prediction models. The hazard ratio (HR) with 95% CI was derived from Cox fitting. The goodness-of-fit of the Cox proportional hazard model was assessed using a likelihood-ratio (LR) test, and presented as an LR P-value. The methylation status of SDPR in different LUAD clinical stage samples was shown as violin plots after grouping samples according to stage.

Identification of potential transcription factor for SDPR

The transcription start site (TSS) of SDPR was indicated by the University of California, Santa Cruz (UCSC) Genome Browser (http://genome.ucsc.edu), and the DNA sequence from 2,000 bp nucleotides upstream and 500 bp nucleotides downstream of the TSS was used as a potential promoter sequence for SDPR. PROMO at the ALGGEN server was subsequently used to identify the putative transcription factor binding sites in this promoter sequence (25). The co-expression profiles of the SDPR gene in LUAD were identified and presented as the pattern of a heat map using the Oncomine database (17), in which the node correlation value is computed as the average of all pair-wise correlations among genes. The node correlation value >0.5 was used to define significant co-expressing genes. Finally, the intersection of the above two profiles was investigated in order to identify the potential transcription factor regulating SDPR expression.

Results

Downregulation of SDPR in various types of cancer

The expression of SDPR is nearly ubiquitous in normal tissues and increased expression levels have been reported in the heart and lungs, while lower expression levels have been indicated in the kidney, the brain, the pancreas, skeletal muscle and the liver (8). To explore the gene expression alteration of SDPR in tumor tissues, the present study compared the mRNA levels of SDPR in various unique tumor tissue datasets compared with normal tissue datasets from the Oncomine™ Platform (17). Consistent with previous studies, the analysis showed that the expression of SDPR was downregulated in breast (1,1113), kidney (11,14) and prostate cancer (11,15), and SARC (4,26). In addition, the present results identified that SDPR is downregulated in bladder, lung, cervical, colorectal, gastric, ovarian and pancreatic cancer (Fig. 1).

Furthermore, gene expression levels of SDPR in cancer subtypes were estimated as a fold-change and gene rank (Table I). The fold change was from-44.083 (invasive ductal breast carcinoma) to-2.102 (cervical squamous cell carcinoma). The gene rank was between 6 and 1% (in the top %). For instance, the entire subtype dataset of breast cancer showed 1% downregulated gene ranks. Downregulated gene expression of SDPR was also observed in lung cancer (in top 3–1%), pancreatic cancer (in top 1%) and SARC (in top 4–1%).

Table I.

SDPR expression in cancer.

Table I.

SDPR expression in cancer.

A, Bladder cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)
Sanchez-Carbayo et al, 2006Superficial bladder cancer<0.001−6.264128(34)
Kim et al, 2010Superficial bladder cancer<0.001−3.0763126(35)
Sanchez-Carbayo et al, 2006Infiltrating bladder urothelial carcinoma<0.001−3.517281(34)
Kim et al, 2010Infiltrating bladder urothelial carcinoma<0.001−2.315662(35)

B, Breast cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Curtis et al, 2012Invasive lobular breast carcinoma<0.001−4.8571148(12)
TCGA Breast, Current studyInvasive lobular breast carcinoma<0.001−10.124136
Curtis et al, 2012Tubular breast carcinoma<0.001−5.474167(12)
Curtis et al, 2012Medullary breast carcinoma<0.001−8.162132(12)
Curtis et al, 2012Invasive ductal breast carcinoma<0.001−44.08311,556(12)
TCGA Breast, Current studyInvasive ductal breast carcinoma<0.001−25.3471389
Curtis et al, 2012Mucinous breast carcinoma<0.001−5.711146(12)

C, Cervical cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Biewenga et al, 2008Cervical squamous cell carcinoma<0.001−2.102540(36)
Scotto et al, 2008Cervical squamous cell carcinoma<0.001−2.123332(37)

D, Colorectal cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Skrzypczak et al, 2010Colorectal adenocarcinoma<0.001−3.48445(38)
Ki et al, 2007Colon adenocarcinoma<0.001−2.413150(39)

E, Gastric cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

D'Errico et al, 2009Gastric intestinal type adenocarcinoma<0.001−2.126326(40)
Cho et al, 2011Diffuse gastric adenocarcinoma<0.001−4.626431(41)

F, Leukemia

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Haferlach et al, 2010Pro-B acute lymphoblastic<0.001−4.65370(42)
Haferlach et al, 2010Acute myeloid leukemia<0.001−2.4424542(42)
Haferlach et al, 2010B-cell acute lymphoblastic Leukemia<0.001−4.5134147(42)
Haferlach et al, 2010T-Cell acute lymphoblastic leukemia<0.001−4.3664174(42)
Haferlach et al, 2010B-cell childhood acute lymphoblastic Leukemia<0.001−4.1875359(42)

G, Lung cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Garber et al, 2001Lung aenocarcinoma<0.001−9.404240(43)
Okayama et al, 2012Lung aenocarcinoma<0.001−5.9761226(44)
Selamat et al, 2012Lung aenocarcinoma<0.001−6.617158(45)
Su et al, 2007Lung aenocarcinoma<0.001−3.803227(46)
Landi et al, 2008Lung aenocarcinoma<0.001−4.783258(47)
Hou et al, 2010Lung aenocarcinoma<0.001−7.204345(48)
Garber et al, 2001Squamous cell lung carcinoma<0.001−11.994113(43)
Wachi et al, 2005Squamous cell lung carcinoma<0.001−5.83315(49)
Hou et al, 2010Squamous cell lung carcinoma<0.001−8.845227(48)
Garber et al, 2001Large cell lung carcinoma<0.001−9.98114(43)
Crabtree et al, 2009Large cell lung carcinoma<0.001−10.929319(50)

H, Other cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Yoshihara et al, 2009Uterine Corpus Leiomyoma<0.001−2.418150(51)

I, Ovarian cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

TCGA Ovarian, Current studyOvarian serous adenocarcinoma<0.001−38.254137
Pei et al, 2009Ovarian serous cystadenocarcinoma<0.001−3.5194586(52)

J, Pancreatic cancer

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Barretina et al, 2010Pancreatic carcinoma<0.001−2.616136(26)

K, Sarcoma

Author, yearCancer subtypeP-valueFold changeGene rank (%)Samples (n)(Refs.)

Barretina et al, 2010Pleomorphic myxofibrosarcoma<0.001−8.44713(26)
Barretina et al, 2010Pleomorphic liposarcoma<0.001−4.486123(26)
Barretina et al, 2010Dedifferentiated liposarcoma<0.001−4.804146(26)
Barretina et al, 2010 Myxofibrosarcoma<0.001−6.96131(26)
Barretina et al, 2010Leiomyosarcoma<0.001−3.417226(26)
Barretina et al, 2010Myxoid/round cell liposarcoma<0.001−2.673420(26)
SDPR gene expression alteration and survival of patients

The PrognoScan platform, which integrates published cancer microarray datasets with clinical annotation (19), was used in the systematic meta-analysis and to determine the prognostic value of SDPR in multiple datasets. Survival analysis consists of two steps, patient grouping and comparing the determined risks of these groups. Since gene expression is continuous data, the PrognoScan platform employed a minimum P-value approach for grouping patients in the survival analysis, which determines the optimal cut-off point in the continuous gene-expression measurement.

In the present study, PrognoScan indicated a significant association between microarray SDPR expression and cancer prognosis in several tests: Brain (1/8), breast (10/40), lung (6/19) and soft tissue (1/1). In all these 18 tests, low SDPR expression was associated with poor survival (data not shown), suggesting its protective function in cancer malignancy. Decreased SDPR expression was significantly associated with decreased overall survival (OS) and relapse-free survival (RFS) of patients with breast cancer (Fig. 2A and B), consistent with a previous study (1). In addition, SDPR downregulation was significantly associated with OS and RFS in patients with lung cancer adenocarcinoma (Fig. 2C and D), OS in patients with glioma (Fig. 2E) and distant-recurrence free survival (DRFS) in liposarcoma (Fig. 2F).

SDPR is hypermethylated in specific types of cancer

In a previous next-generation sequencing study, SDPR was not identified as a frequent mutational gene target (16) and it was also observed that SDPR was epigenetically silenced in breast cancer cell lines (1). Therefore, the present study hypothesized that DNA methylation alteration, which is an important epigenetic regulator for transcription, may be one of the mechanisms for SDPR gene expression differences. In order to reveal the underlying molecular mechanisms responsible for SDPR downregulation, MethHC was used to identify differential DNA methylation data between tumor and non-tumor tissues, which included 18 human types of cancer in >6,000 samples. MethHC is a database that systematically integrates DNA methylation and mRNA expression data from TCGA. The methylation profile of SDPR across 18 tumors is presented in Fig. 3. In this profile, significantly SDPR gene methylation differences were observed in most types of cancer (17/18). Consistent with previously published experimental data, SDPR was significantly hypermethylated in breast invasive carcinoma compared with normal breast tissues (1). In addition, SDPR was observed to be significantly hypermethylated in bladder urothelial carcinoma, cervical squamous cell carcinoma, head and neck squamous cell carcinoma, LUAD, lung squamous cell carcinoma (LUSC), pancreatic adenocarcinoma, prostate adenocarcinoma, SARC, skin cutaneous melanoma, stomach adenocarcinoma and uterine corpus endometrial carcinoma (P<0.005). Statistical differences were also found in colon adenocarcinoma, rectum adenocarcinoma and thyroid carcinoma (P<0.05). These results suggested that DNA methylation may be responsible for SDPR downregulation in these types of cancer. As shown in Fig. 1, SDPR was downregulated in kidney cancer. However, significant SDPR hypomethylation was found in both kidney renal clear cell carcinoma (KIRC) and kidney renal papillary cell carcinoma (KIRP) (P<0.005), suggesting the existence of other regulatory pathways.

Gene methylation of SDPR is associated with patient survival in LUAD and SARC

As downregulation of SDPR gene expression was observed in lung cancer and SARC, SDPR was indicated to be hypermethylated compared with normal tissues, and downregulation of SDPR expression was significantly associated with poor patient survival in LUAD and SARC, MethSurv was used to identify whether hypermethylation of SDPR was associated with patient survival in LUAD or SARC. In addition, since the differential methylation levels at the CpG island are tissue-specific, the DNA methylation patterns were analyzed, taking into consideration the differential methylation levels in different gene subregions (Fig. 4A). MethSurv was the first database that indicated an association with overall survival and DNA methylation patterns, in which the methylation levels are from TCGA methylation profile using the HM450k array. Based on the UCSC database, the CpG sites were grouped into gene subregions: ‘TSS200’, ‘TSS1500’, ‘1st exon’, ‘5′UTR’, ‘body’ and ‘3′UTR’ (23). As shown in Fig. 4B and C, clustering visualization in the form of heat maps evaluated the association of methylation levels with the available patient characteristics and gene subregions in LUAD and SARC. According to the heatmaps, methylation sites cg18843739, cg10082589 and cg17809945 in the ‘TSS1500’ subregion showed differential methylation levels in patients with LUAD (461 samples). Similar to patients with SARC, the differently methylated sites were cg07488576 in the ‘1st Exon’ subregion and cg18843739 in the ‘TSS1500’ subregion (261 samples).

MethSurv was used to identify which methylation sites were significantly associated with patient survival. By analyzing all 11 methylation sites in LUAD hypermethylation, it was indicated that two sites in LUAD (cg17809945 and cg10082589, located in the ‘TSS1500’ subregion of SDPR) were significantly associated with poor overall survival (Fig. 4D and E). In SARC, 2 out of 13 methylation sites (cg07488576 in the ‘1st Exon’ subregion and cg18843739 in the ‘TSS1500’ subregion) were identified to be associated with poor survival (Fig. 4F and G). The results were consistent with the heatmap presented in Fig. 4B and C. In this analysis, CpG (cg10082589; HR=1.887; 95% CI: 1.371–2.596; LR test P-value=0.001) was identified as the optimal survival methylation marker for LUAD (Fig. 4E) and CpG (cg07488576; HR=2.406; 95% CI: 1.608–3.599; LR test P-value=0.00001) was identified as the optimal survival methylation marker for SARC (Fig. 4F). In addition, when LUAD samples were grouped according to clinical stage, the methylation levels of cg17809945 (Fig. 4H) and cg10082589 (Fig. 4I) were indicated to be higher in late stages, suggesting that they may be involved in LUAD progression. However, no association between methylation levels of CpG sites and clinical stages was observed in SARC (data not shown).

Prediction of transcription factors regulating SDPR expression in LUAD

Since epigenetic patterns may not be the only reason for gene expression alternations in cancer, transcriptional regulation by deregulated transcription factors was investigated. The UCSC database was used to identify the promoter sequences of SDPR and PROMO was subsequently used at the ALGGEN server showing potential transcriptional factors on the promoter (Fig. 5A). Meanwhile, genes that were co-expressed with SDPR were analyzed using the Oncomine database, which were subsequently grouped into normal lung tissue and LUAD (Fig. 5B). By comparing the potential transcriptional factors with genes significantly co-expressed with SDPR (node correlation value >0.5), GATA binding protein 2 (GATA2; node correlation value=0.807) was identified as a potential transcription factor. GATA2 is a member of the GATA family, serves as a transcriptional activator during development and carcinogenesis, and was indicated to be epigenetically repressed in both human and mouse lung tumors (27). This result suggested that GATA2 may be a potential transcription factor regulating SDPR gene expression in LUAD.

Discussion

SDPR has been previously reported to show characteristic gene signatures in specific types of cancer, including breast (1,2,1113), thyroid (3,28), oral (5) and kidney (11,14) cancer, and SARC (4,26). In breast cancer, SDPR was identified as a novel tumor suppressor, which was significantly associated with patient survival (1,2). However, a systemic profile of SDPR alterations or analysis of its relevance in clinical outcomes in different types of cancer has yet to be performed, to the best of our knowledge. In the present study, SDPR downregulation was observed in bladder, breast, lung, kidney, cervical, colorectal, gastric, ovarian and pancreatic cancer. Consistent with previous studies (1,2), SDPR downregulation was significantly associated with patient survival in breast cancer. Furthermore, the analysis also indicated that decreased expression of SDPR was significantly associated with poor OS and RFS in patients with adenocarcinoma of lung cancer, OS in patients with glioma and DRFS in liposarcoma.

In breast cancer, SDPR, which is partially silenced by DNA methylation, has been elucidated to execute an anti-metastatic function by promoting apoptosis (1), and depletion of SDPR-induced EMT through TGF-β signaling activation, according to previously published experimental studies (1,2). To clarify why SDPR expression was altered in other types of cancer, the gene methylation level of SDPR was subsequently profiled in 18 types of cancer and was compared with normal tissues. It was indicated to be significantly altered in 17 out of 18 types of cancer.

If DNA methylation of SDPR is involved in its downregulation in LUAD or SARC, it might be associated with patient survival. As not all of the methylation sites are responsible for gene expression, the most significant methylation sites require further investigation. Therefore, the present study analyzed the DNA methylation patterns in LUAD and SARC, considering the differential methylation levels in different gene subregions of SDPR. To the best of our knowledge, the present study for the first time, indicated that CpG (cg10082589) serves as the optimal survival methylation marker for LUAD, and CpG (cg07488576) serves as the optimal survival methylation marker for SARC. Furthermore, when LUAD samples were grouped according to clinical stage, the methylation level of CpG (cg10082589) and CpG (cg1780995) were higher in late stages, suggesting that they may be involved in LUAD progression.

In addition, by analyzing both the co-expressed genes with SDPR and the putative transcription factor binding sites on SDPR, GATA2 was identified as a potential transcription factor for SDPR transcription in LUAD. Transcriptional factor GATA2 regulates genes critical for embryonic development, self-renewal maintenance (29), functionality of blood-forming (30) and lymphatic vessel valve development (31). GATA2 has been reported to be frequently epigenetically repressed in both human and mouse lung tumors, and aberrant GATA2 methylation occurred early during lung carcinogenesis (27). GATA2 may serve a role in the downregulation of SDPR in LUAD.

Analysis of the associations between SDPR expression and DNA methylation with patient survival in LUSC was additionally performed. However, neither the OS nor disease-free survival of patients had been observed to be significantly associated with SDPR expression alternations, according to the Prognoscan database. Furthermore, similar to LUAD, the same 13 CpG sites grouped in gene subregions based on the UCSC database were analyzed using MethSurv. However, the results indicated no significant association between DNA methylation data and patient survival (data not shown). Since gene expression patterns differ in the subtypes of lung cancer, this may provide novel insight for the examination of potential mechanisms of SDPR function in LUAD.

SDPR was also downregulated in kidney cancer. Significant hypomethylation was found in both KIRC and KIRP, suggesting other regulatory pathways. By comparing methylation patterns with patient survival in KIRC and KIRP, the present study indicated that hypomethylation in the ‘TSS200’ and ‘TSS1500’ subregions of SDPR was significantly associated with longer survival time, while hypomethylation in the gene ‘body’ and the ‘3′UTR’ subregions was associated with poor OS (data not shown). The function of DNA methylation status seems to vary in context; this may be due to hypermethylation in the promoter region inducing the downregulation of gene expression, while hypermethylation in the gene body may not block and may even stimulate transcription elongation, and the gene body methylation may have an impact on splicing (32). In addition, long non-coding RNA (lncRNA) SDPR-antisense (SDPR-AS) has been verified to be co-expressed with SDPR, and elevated lncRNA SDPR-AS increases the OS in renal cell carcinoma, suggesting the possibility that lncRNAs may serve a regulatory role in the SDPR pathway (33).

The specific methylation CpG sites, which are significantly associated with patient survival, require further study in order to verify the present results, such as pyrosequencing and reverse transcription-quantitative PCR. The potential transcription factor GATA2 should also be experimentally investigated to determine whether it is responsible in SDPR transactivation. Despite taking age and sex into consideration as covariates in survival analysis based on the Methsurv database, due to the lack of available clinical data, the survival analysis based on PrognoScan is univariable. Since several factors, such as co-morbidities, performance status and treatments, may potentially affect the prognosis of patients with cancer, it is important to consider all potential relevant specific features for specific cancer types in future clinical investigations of SDPR.

In summary, the present study suggested that the role of SDPR as a tumor suppressor may have broader clinical relevance beyond breast cancer. The present study on SDPR may help to examine its underlying molecular mechanism in cancer progression, reveal novel perspectives for prognostic evaluation in specific cancer, and provide insight for further research in the field.

Acknowledgements

Not applicable.

Funding

The present study was supported by Foundation of Sichuan Educational Committee, People's Republic of China (grant nos. 17ZA0164 and 18CZ0010) and Foundation of Chengdu University of Traditional Chinese Medicine, China (grant nos. ZRQN1660 and CGZH1709).

Availability of data and materials

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

Authors' contributions

YW and YL designed the present study. YW analyzed the gene expressing data, patient survival data and the potential transcriptional factors of SDPR. ZS analyzed the co-expressing genes with SDPR. YL and PL analyzed the gene methylation data. YW wrote the manuscript and YL revised it. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ozturk S, Papageorgis P, Wong CK, Lambert AW, Abdolmaleky HM, Thiagalingam A, Cohen HT and Thiagalingam S: SDPR functions as a metastasis suppressor in breast cancer by promoting apoptosis. Proc Natl Acad Sci USA. 113:638–643. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Tian Y, Yu Y, Hou LK, Chi JR, Mao JF, Xia L, Wang X, Wang P and Cao XC: Serum deprivation response inhibits breast cancer progression by blocking transforming growth factor-β signaling. Cancer Sci. 107:274–280. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Poma AM, Giannini R, Piaggi P, Ugolini C, Materazzi G, Miccoli P, Vitti P and Basolo F: A six-gene panel to label follicular adenoma, low- and high-risk follicular thyroid carcinoma. Endocr Connect. 7:124–132. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Codenotti S, Vezzoli M, Poliani PL, Cominelli M, Monti E and Fanzani A: Cavin-2 is a specific marker for detection of well-differentiated liposarcoma. Biochem Biophys Res Commun. 493:660–665. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Unozawa M, Kasamatsu A, Higo M, Fukumoto C, Koyama T, Sakazume T, Nakashima D, Ogawara K, Yokoe H, Shiiba M, et al: Cavin-2 in oral cancer: A potential predictor for tumor progression. Mol Carcinog. 55:1037–1047. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Hansen CG, Bright NA, Howard G and Nichols BJ: SDPR induces membrane curvature and functions in the formation of caveolae. Nat Cell Biol. 11:807–814. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Nassar ZD and Parat MO: Cavin family: New players in the biology of caveolae. Int Rev Cell Mol Biol. 320:235–305. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Gustincich S, Vatta P, Goruppi S, Wolf M, Saccone S, Della Valle G, Baggiolini M and Schneider C: The human serum deprivation response gene (SDPR) maps to 2q32-q33 and codes for a phosphatidylserine-binding protein. Genomics. 57:120–129. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Baig A, Bao X, Wolf M and Haslam RJ: The platelet protein kinase C substrate pleckstrin binds directly to SDPR protein. Platelets. 20:446–457. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Gupta R, Toufaily C and Annabi B: Caveolin and cavin family members: Dual roles in cancer. Biochimie. 107:188–202. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Li X, Jia Z, Shen Y, Ichikawa H, Jarvik J, Nagele RG and Goldberg GS: Coordinate suppression of Sdpr and Fhl1 expression in tumors of the breast, kidney, and prostate. Cancer Sci. 99:1326–1333. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, et al: The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 486:346–352. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Bai L, Deng X, Li Q, Wang M, An W, Deli A, Gao Z, Xie Y, Dai Y and Cong YS: Down-regulation of the cavin family proteins in breast cancer. J Cell Biochem. 113:322–328. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Gianazza E, Chinello C, Mainini V, Cazzaniga M, Squeo V, Albo G, Signorini S, Di Pierro SS, Ferrero S, Nicolardi S, et al: Alterations of the serum peptidome in renal cell carcinoma discriminating benign and malignant kidney tumors. J Proteomics. 76:125–140. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Altintas DM, Allioli N, Decaussin M, de Bernard S, Ruffion A, Samarut J and Vlaeminck-Guillem V: Differentially expressed androgen-regulated genes in androgen-sensitive tissues reveal potential biomarkers of early prostate cancer. PLoS One. 8:e662782013. View Article : Google Scholar : PubMed/NCBI

16 

Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr and Kinzler KW: Cancer genome landscapes. Science. 339:1546–1558. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, Barrette TR, Anstet MJ, Kincead-Beal C, Kulkarni P, et al: Oncomine 3.0: Genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 9:166–180. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: A practical and powerful approach to multiple testing. J R Statist Soc B. 57:289–300. 1995.

19 

Mizuno H, Kitada K, Nakai K and Sarai A: PrognoScan: A new database for meta-analysis of the prognostic value of genes. BMC Med Genomics. 2:182009. View Article : Google Scholar : PubMed/NCBI

20 

Miller R and Siegmund D: Maximally selected chi square statistics. Biometrics. 38:1011–1016. 1982. View Article : Google Scholar

21 

Huang WY, Hsu SD, Huang HY, Sun YM, Chou CH, Weng SL and Huang HD: MethHC: A database of DNA methylation and gene expression in human cancer. Nucleic Acids Res 43 (Database Issue). D856–D861. 2015. View Article : Google Scholar

22 

Broad Institute TCGA Genome Data Analysis Center (2016), . Firehose stddata__2016_01_28 run. Broad Institute of MIT and Harvard. Doi: 10.7908/C11G0KM9.

23 

Modhukur V, Iljasenko T, Metsalu T, Lokk K, Laisk-Podar T and Vilo J: MethSurv: A web tool to perform multivariable survival analysis using DNA methylation data. Epigenomics. 10:277–288. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Metsalu T and Vilo J: ClustVis: A web tool for visualizing clustering of multivariate data using principal component analysis and heatmap. Nucleic Acids Res 43W. W566–W570. 2015. View Article : Google Scholar

25 

Farré D, Roset R, Huerta M, Adsuara JE, Roselló L, Albà MM and Messeguer X: Identification of patterns in biological sequences at the ALGGEN server: PROMO and MALGEN. Nucleic Acids Res. 31:3651–3653. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos-Quintana M, Decarolis PL, Shah K, Socci ND, Weir BA, Ho A, et al: Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet. 42:715–721. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Tessema M, Yingling CM, Snider AM, Do K, Juri DE, Picchi MA, Zhang X, Liu Y, Leng S, Tellez CS and Belinsky SA: GATA2 is epigenetically repressed in human and mouse lung tumors and is not requisite for survival of KRAS mutant lung cancer. J Thorac Oncol. 9:784–793. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Borup R, Rossing M, Henao R, Yamamoto Y, Krogdahl A, Godballe C, Winther O, Kiss K, Christensen L, Høgdall E, et al: Molecular signatures of thyroid follicular neoplasia. Endocr Relat Cancer. 17:691–708. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Krendl C, Shaposhnikov D, Rishko V, Ori C, Ziegenhain C, Sass S, Simon L, Müller NS, Straub T, Brooks KE, et al: GATA2/3-TFAP2A/C transcription factor network couples human pluripotent stem cell differentiation to trophectoderm with repression of pluripotency. Proc Natl Acad Sci USA. 114:E9579–E9588. 2017. View Article : Google Scholar : PubMed/NCBI

30 

de Pater E, Kaimakis P, Vink CS, Yokomizo T, Yamada-Inagawa T, van der Linden R, Kartalaei PS, Camper SA, Speck N and Dzierzak E: Gata2 is required for HSC generation and survival. J Exp Med. 210:2843–2850. 2013. View Article : Google Scholar

31 

Kazenwadel J, Betterman KL, Chong CE, Stokes PH, Lee YK, Secker GA, Agalarov Y, Demir CS, Lawrence DM, Sutton DL, et al: GATA2 is required for lymphatic vessel valve development and maintenance. J Clin Invest. 125:2979–2994. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Jones PA: Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat Rev Genet. 13:484–492. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Ni W, Song E, Gong M, Li Y, Yao J and An R: Downregulation of lncRNA SDPR-AS is associated with poor prognosis in renal cell carcinoma. Onco Targets Ther. 10:3039–3047. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Sanchez-Carbayo M, Socci ND, Lozano J, Saint F and Cordon-Cardo C: Defining molecular profiles of poor outcome in patients with invasive bladder cancer using oligonucleotide microarrays. J Clin Oncol. 24:778–789. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Kim WJ, Kim EJ, Kim SK, Kim YJ, Ha YS, Jeong P, Kim MJ, Yun SJ, Lee KM, Moon SK, et al: Predictive value of progression-related gene classifier in primary non-muscle invasive bladder cancer. Mol Cancer. 9:32010. View Article : Google Scholar : PubMed/NCBI

36 

Biewenga P, Buist MR, Moerland PD, Ver Loren van Themaat E, van Kampen AH, ten Kate FJ and Baas F: Gene expression in early stage cervical cancer. Gynecol Oncol. 108:520–526. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Scotto L, Narayan G, Nandula SV, Arias-Pulido H, Subramaniyam S, Schneider A, Kaufmann AM, Wright JD, Pothuri B, Mansukhani M and Murty VV: Identification of copy number gain and overexpressed genes on chromosome arm 20q by an integrative genomic approach in cervical cancer: Potential role in progression. Genes Chromosomes Cancer. 47:755–765. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Skrzypczak M, Goryca K, Rubel T, Paziewska A, Mikula M, Jarosz D, Pachlewski J, Oledzki J and Ostrowski J: Modeling oncogenic signaling in colon tumors by multidirectional analyses of microarray data directed for maximization of analytical reliability. PLoS One. 5(pii): e130912010. View Article : Google Scholar : PubMed/NCBI

39 

Ki DH, Jeung HC, Park CH, Kang SH, Lee GY, Lee WS, Kim NK, Chung HC and Rha SY: Whole genome analysis for liver metastasis gene signatures in colorectal cancer. Int J Cancer. 121:2005–2012. 2007. View Article : Google Scholar : PubMed/NCBI

40 

D'Errico M, de Rinaldis E, Blasi MF, Viti V, Falchetti M, Calcagnile A, Sera F, Saieva C, Ottini L, Palli D, et al: Genome-wide expression profile of sporadic gastric cancers with microsatellite instability. Eur J Cancer. 45:461–469. 2009. View Article : Google Scholar

41 

Cho JY, Lim JY, Cheong JH, Park YY, Yoon SL, Kim SM, Kim SB, Kim H, Hong SW, Park YN, et al: Gene expression signature-based prognostic risk score in gastric cancer. Clin Cancer Res. 17:1850–1857. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Haferlach T, Kohlmann A, Wieczorek L, Basso G, Kronnie GT, Béné MC, De Vos J, Hernández JM, Hofmann WK, Mills KI, et al: Clinical utility of microarray-based gene expression profiling in the diagnosis and subclassification of leukemia: Report from the international microarray innovations in leukemia study group. J Clin Oncol. 28:2529–2537. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna-Gengelbach M, van de Rijn M, Rosen GD, Perou CM, Whyte RI, et al: Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci USA. 98:13784–13789. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, Furuta K, Tsuta K, Shibata T, Yamamoto S, et al: Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72:100–111. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Selamat SA, Chung BS, Girard L, Zhang W, Zhang Y, Campan M, Siegmund KD, Koss MN, Hagen JA, Lam WL, et al: Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res. 22:1197–1211. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Su LJ, Chang CW, Wu YC, Chen KC, Lin CJ, Liang SC, Lin CH, Whang-Peng J, Hsu SL, Chen CH and Huang CY: Selection of DDX5 as a novel internal control for Q-RT-PCR from microarray data using a block bootstrap re-sampling scheme. BMC Genomics. 8:1402007. View Article : Google Scholar : PubMed/NCBI

47 

Landi MT, Dracheva T, Rotunno M, Figueroa JD, Liu H, Dasgupta A, Mann FE, Fukuoka J, Hames M, Bergen AW, et al: Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One. 3:e16512008. View Article : Google Scholar : PubMed/NCBI

48 

Hou J, Aerts J, den Hamer B, van Ijcken W, den Bakker M, Riegman P, van der Leest C, van der Spek P, Foekens JA, Hoogsteden HC, et al: Gene expression-based classification of non-small cell lung carcinomas and survival prediction. PLoS One. 5:e103122010. View Article : Google Scholar : PubMed/NCBI

49 

Wachi S, Yoneda K and Wu R: Interactome-transcriptome analysis reveals the high centrality of genes differentially expressed in lung cancer tissues. Bioinformatics. 21:4205–4208. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Crabtree JS, Jelinsky SA, Harris HA, Choe SE, Cotreau MM, Kimberland ML, Wilson E, Saraf KA, Liu W, McCampbell AS, et al: Comparison of human and rat uterine leiomyomata: Identification of a dysregulated mammalian target of rapamycin pathway. Cancer Res. 69:6171–6178. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Yoshihara K, Tajima A, Komata D, Yamamoto T, Kodama S, Fujiwara H, Suzuki M, Onishi Y, Hatae M, Sueyoshi K, et al: Gene expression profiling of advanced-stage serous ovarian cancers distinguishes novel subclasses and implicates ZEB2 in tumor progression and prognosis. Cancer Sci. 100:1421–1428. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Pei H, Li L, Fridley BL, Jenkins GD, Kalari KR, Lingle W, Petersen G, Lou Z and Wang L: FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer Cell. 16:259–266. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2019
Volume 42 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Song Z, Leng P and Liu Y: A systematic analysis reveals gene expression alteration of serum deprivation response (SDPR) gene is significantly associated with the survival of patients with cancer. Oncol Rep 42: 1161-1172, 2019
APA
Wang, Y., Song, Z., Leng, P., & Liu, Y. (2019). A systematic analysis reveals gene expression alteration of serum deprivation response (SDPR) gene is significantly associated with the survival of patients with cancer. Oncology Reports, 42, 1161-1172. https://doi.org/10.3892/or.2019.7212
MLA
Wang, Y., Song, Z., Leng, P., Liu, Y."A systematic analysis reveals gene expression alteration of serum deprivation response (SDPR) gene is significantly associated with the survival of patients with cancer". Oncology Reports 42.3 (2019): 1161-1172.
Chicago
Wang, Y., Song, Z., Leng, P., Liu, Y."A systematic analysis reveals gene expression alteration of serum deprivation response (SDPR) gene is significantly associated with the survival of patients with cancer". Oncology Reports 42, no. 3 (2019): 1161-1172. https://doi.org/10.3892/or.2019.7212