Skip to main content

Computational identification of surrogate genes for prostate cancer phases using machine learning and molecular network analysis

Abstract

Background

Prostate cancer is one of the most common malignant diseases and is characterized by heterogeneity in the clinical course. To date, there are no efficient morphologic features or genomic biomarkers that can characterize the phenotypes of the cancer, especially with regard to metastasis – the most adverse outcome. Searching for effective surrogate genes out of large quantities of gene expression data is a key to cancer phenotyping and/or understanding molecular mechanisms underlying prostate cancer development.

Results

Using the maximum relevance minimum redundancy (mRMR) method on microarray data from normal tissues, primary tumors and metastatic tumors, we identifed four genes that can optimally classify samples of different prostate cancer phases. Moreover, we constructed a molecular interaction network with existing bioinformatic resources and co-identifed eight genes on the shortest-paths among the mRMR-identified genes, which are potential co-acting factors of prostate cancer. Functional analyses show that molecular functions involved in cell communication, hormone-receptor mediated signaling, and transcription regulation play important roles in the development of prostate cancer.

Conclusion

We conclude that the surrogate genes we have selected compose an effective classifier of prostate cancer phases, which corresponds to a minimum characterization of cancer phenotypes on the molecular level. Along with their molecular interaction partners, it is fairly to assume that these genes may have important roles in prostate cancer development; particularly, the un-reported genes may bring new insights for the understanding of the molecular mechanisms. Thus our results may serve as a candidate gene set for further functional studies.

Background

Prostate cancer is one of the most frequently-occurred malignant diseases affecting human health and life qualities [1]. In this cancer, metastasis (i.e. tumor cells escaping from the primary tissue and eventually colonizing a distant site) reflects the most adverse phase, which commonly results in disruption of a complex set of biological processes, causing severe bone pain and spinal cord complications [2, 3]. Due to the heterogeneity of the disease, there are currently no reliable morphologic features or genetic/genomic biomarkers that can effectively discriminate tissue-confined primary and/or metastatic tumors, thus less is known for the mechanisms underlying the development of metastatic disease.

Many efforts have been devoted to revealing the molecular mechanisms underlying the disease progression and/or identifying genetic/genomic surrogates for the tumor phenotypes. In most of the studies, the phenotype of a tumor is defined by its phase [4, 5]; and identification of molecular surrogates underlying the different tumor phases is facilitated by classification of samples from the respective phases (i.e. normal prostate, primary tumor, and metastatic tumor). Since the different phases constitute the process of disease progression, the surrogates (i.e. set of genes) that distinguish the phases (or classify samples from different phases) would certainly provide insights for understanding the molecular mechanisms of disease progression. For prostate cancer, gene expression microarray studies have characterized expression profiles of primary cancers, metastatic cancers and normal tissues [68]; in some cases, correlations between gene expressions and cancer phases have been revealed [9]. The studies have further led to the finding that differential gene expression profiles hold for metastatic androgen ablation resistant prostate cancer (AARPC) and androgen-dependent metastatic cancers [10]. In general, these results have gained important insights about metastatic prostate cancer, regarding to the changes in expressions of genes involved in various biological processes, e.g. signal transduction, cell cycle, cell adhesion, migration and mitosis, etc. [11, 12]. Nonetheless, one important problem remains: previous studies describe the correlations of expression profiles and disease phases in terms of hundreds of genes, whereas they seldom provide a convenient molecular measure (i.e. minimum predictor gene set) for accurate classification of prostate cancer phases, especially with respect to metastasis. Such a predictor gene set would be a better highlight for the mechanisms of prostate cancer.

To address this issue, we herein adopt a two-step pipeline widely-used in previous studies, which includes machine learning to identify disease-related genes and pathway analysis to reveal molecular interactions among the genes [1316]. First, we utilize the machine learning strategy for accurate classification of prostate cancer phenotypes based on gene expression microarray data. Specifically, we use the minimum redundancy – maximum relevance method (mRMR), a robust method with a broad spectrum of applications [13, 17], to serve our goal of identifying a largest-parsimony (i.e. minimum) surrogate (i.e. gene set) for prostate cancer phases. Moreover, in order to focus more on the issue of metastasis, we not only consider gene expression data of normal and (tissue-confined) primary prostate tumor tissues [7], but also include a previously published dataset of metastatic tumor samples (i.e. tissue samples excluding potentially uninformative stromal genes) in our study [11].

Furthermore, genes/proteins usually co-function with their interaction partners; thus molecular interaction partners of disease-related genes are also candidates for further studies. For this purpose, we pinpoint the identified surrogate genes in a molecular interaction network constructed based on STRING (Search Tool for the Retrieval of Interacting Genes), which is a database providing resources of molecular interaction information [18]; and we then identify by the shortest-path analysis a set of potential co-acting factors, which may serve as candidate causal genes for further experimental studies.

Materials and methods

Data source

The gene expression dataset was adopted from a research on prostate cancer by Chandran et al. [11]. The data were with the Affymatrix GPL92 platform and generated from 167 samples, which contained 77 normal tissues (NTs, including both normal prostate tissues free of pathological alterations from organ donor and normal tissues adjacent to tumor), 66 primary prostate tumors (PTs) and 24 metastatic tumors (MTs). All tissue samples were acquired from the Health Sciences Tissue Bank of the University of Pittsburgh Medical Center under stringent Institutional Review Board guidelines with appropriate informed consent [11]. The data were downloaded from NCBI Gene Expression Omnibus (GEO) with accession number GSE6919. The normalized expression data were obtained directly from the GEO website, in which the data were normalized by global scaling and analyzed with Microarray Suite version 5.0 (MAS 5.0) using Affymetrix default settings. In our machine learning procedure, we did not combine the expressions from probes to genes; instead we obtained results at the probe level directly. We focused our analysis on probes corresponding to protein coding genes.

Algorithm of mRMR & prediction engine

The minimum redundancy – maximum relevance (mRMR) algorithm was utilized herein to select surrogate genes for prostate cancer progression. The major steps of mRMR implementation were the same as we previously described [13]. The algorithm aimed to balance features’ relevance to the prediction target and the redundancy between features. Both relevance and redundancy were quantified with mutual information (MI), estimated as,

I x , y = 1 2 ln 1 ρ x , y 2
(1)

where I represented the MI and ρ was the correlation coefficient between the variables x and y.

First, assume that y was the input variable, and X = { x 1 , …, x n } was the set of input features. Given x i as the feature with the highest MI with the input variable, the feature set (S) at the current step was then initialized by x i . Second, we selected the feature x j with the best balance between highest relevance and lowest redundancy and added it to S. It was achieved by maximizing the score q as follows,

q=I x j , y 1 S x k S I x j , x k
(2)

We repeated the above steps until a desired solution length was reached. The mRMR algorithm was implemented using the R package “mRMRe” [19].

We predicted the phenotype of an individual in three ways: 1) the phenotype of its nearest neighbor; 2) the most-occurring phenotype of its five nearest neighbors; 3) the phenotype of its nearest clustering center of each phenotype group (for detailed results, see Additional file 1: Table S1). According to Chou et al.’s studies [17, 20], the distance between two individuals was calculated as follows,

d i 1 , i 2 =1 e 1 e 2 e 1 e 2
(3)

where d was the distance, i 1 and i 2 were two samples, and e 1 and e 2 were the vectors of selected features of i 1 and i 2 , respectively.

Validation & incremental feature selection

We used jackknife validation to estimate the prediction accuracy of the selected features. The advantages of jackknife comparing with other validation methods, such as independent-dataset validation and sub-dataset validation, were discussed previously [17, 20]. In jackknife validation, given X samples of a known outcome variable and N selected features, for each sample we compared the known outcome with an estimated outcome, which was computed based on the rest X - 1 samples. We defined the accuracy of a prediction using the following formula,

Accuracy= TP + TN TP + TN + FP + FN
(4)

where TP, TN, FP and FN represented the numbers of true positives, true negatives, false positives and false negatives, respectively.

Furthermore, Incremental Feature Selection (IFS) was used to determine the number of features for optimal prediction (Figure 1). As previously described [13], for N = 1 to 400 required number of features, each feature set was computed by mRMR and the prediction accuracy was estimated using Jackknife validation. The set with the best prediction accuracy and smallest feature number was regard as the final feature set. In this study, a set with four genes was chosen and its prediction accuracy is 0.7202.

Figure 1
figure 1

IFS curves determining the number of features. We used the IFS curves to determine the number of features used in mRMR computation. “Prediction 1”, “Prediction 2” and “Prediction 3” refer to the three prediction approaches, a vote of the top five nearest neighbors, the first nearest neighbor and nearest clustering center of each phenotype group, seperately. It is noted that when using four genes, the prediction accuracy reaches the maximum.

Molecular interaction network & shortest-path analysis

To reveal possible functional implications of the mRMR-selected genes, we explored the shortest-paths among the genes in a background molecular network constructed using the protein-protein interaction (PPI) data from STRING database (version 9.1) (http://string-db.org) [18]. To identify the shortest-path between two genes/proteins, we used Dijkstra’s algorithm and implemented it in the R package “igraph” [21]. The resulting sub-network of PPIs representing the shortest-paths among the four mRMR-selected genes (Figure 2) was visualized using Cytoscape (version 3.0.1) [22].

Figure 2
figure 2

PPI network of shortest-paths among the four mRMR-identified genes. We identified the shortest-paths between each pair of the four mRMR-selected genes in the backgroud molecular interaction (PPI) network constructed based on STRING. Symbols of proteins and their corresponding genes are used interchangably herein. Rectangles colored in yellow are the four mRMR-selected genes/proteins; the others represent interaction parnters locating on some shortest-paths among them.

GO and KEGG pathway annotation

We carried out functional annotation for all the genes identified by mRMR and shortest-path analysis based on GO and KEGG pathways. The functional annotations were implemented using the web service of DAVID tools (version 6.7) [23], by which existence of gene enrichments to certain functional modules/pathways could also be observed.

Results

A set of four genes presents the best accuracy for predictions of NTs vs. MTs and PTs vs. MTs

In implementation of mRMR, we consecutively tested the predictor with one feature (probes of gene expression array), two features, three features, etc., and the IFS result was provided in Figure 1. In the IFS curves, X-axis is the number of probes used for classification and Y-axis is the prediction accuracy (of the nearest-neighbor algorithm evaluated by the Jackknife validation). As shown, the accuracy for classification of NTs vs. MTs and PTs vs. MTs reaches the maximum when only four features are included, corresponding to four genes annotated in the Ensemble Biomart database (TUBB6, MYEF2, PARM1, SLC25A22). We list the genes in Table 1 and their respective expression levels in NT, PT and MT in Additional file 2: Table S2; we specifically discuss the functions of the genes in later sections.

Table 1 The four genes identified by mRMR

A PPI sub-network provides additional insights for the prostate cancer-related genes

Furthermore, we constructed an molecular interaction network with the PPI data from STRING. We traversed all pairs of any two genes from the four surrogate (protein coding) genes identified by mRMR as described above; and we then calculated the shortest paths between any pair of two proteins/genes using the Dijkstra’s algorithm. We eventually obtained a sub-network of STRING PPIs that contained all these shortest paths (Figure 2). There are a total of 14 protein-protein interactions of 12 proteins, eight of which correspond to genes other than the four mRMR-identified ones and they are all annotated in the Ensemble Biomart database. We list these genes in Table 2 as an extended set of candidate regulatory factors of prostate cancer that are possibly co-acting with the four mRMR-identified surrogate genes.

Table 2 Genes on shortest-paths among the four mRMR-identified genes

Functional annotation of the identified genes

Using the functional annotation tool DAVID, we carried out Gene Ontology (GO) and KEGG Pathway annotation for all the 12 identified genes (including the four mRMR-selected genes and another eight ones traced in the shortest-path PPI network). The results show that many genes are functioning to regulate transcription and/or transcription factor activity (Table 3), echoing the previous finding that genes expressions in (metastatic) prostate cancer are dictated by distinct transcriptional programs [12]. In addition, the genes are also involved in pathways related to steroid hormone receptor activity (Table 3). This is highly consistent with earlier studies that growth of prostate cancer cells is dependent on the male hormone (i.e. androgen) and overly prolonged changes of in vivo hormonal level (e.g. androgen deprivation therapy, ADT) causes the emergence of androgen-independent (AI) cancer cells, which result in more malignancy towards advanced or metastatic prostate cancer [24, 25]. It is obvious that function of hormone receptor plays a crucial role in prostate cancer progression; and our surrogate gene set captures this reality.

Table 3 GO annotation for genes co-identified by mRMR and shortest-path analysis

Interestingly, the identified predictor set includes genes (e.g. TUBB6, GJA1) that are annotated by both GO and KEGG pathways as relating to gap junction and regulation of cell communication (Table 4). In fact, it is long recognized that gap junction-mediated intercellular communication is required for cellular normality and breakdown of this communication is a hallmark of cancer [26, 27]. Furthermore, earlier studies have shown that intercellular communications and expressions of gap junction-forming proteins are largely reduced or not detected in prostate cancer cells [28, 29]. Therefore, our results have faithfully embodied the impact of cell communication dysregulation in prostate cancer.

Table 4 KEGG annotation for genes co-identified by mRMR and shortest-path analysis

Discussion

In the present study, we applied an informatic approach to identify molecular surrogates underlying the different phases of prostate cancer, which would facilitate deciphering the mechanism(s) of disease progression. The investigation led four genes into our sight — SLC25A22, TUBB6, MYEF2 and PARM1. Data have shown that these genes ensure the sample classification with the accuracy of more than 70% and the genes are annotated to cancer-relevant functions/pathways. Thus the reasonability of our research is suggested. In the results, two of the four genes (TUBB6, PARM1) are supported by literature for their roles in prostate cancer; nonetheless, the other two (SLC25A22, MYEF2) are less known. Therefore, due to the indication of our results, we believe that these two genes may also sustain potentially role(s) during prostate cancer progression and they are worth being focused on for further experimental studies.

SLC25A22 is named as solute carrier family 25 (mitochondrial carrier: glutamate), member 22. It is involved in the transport of glutamate across the inner mitochondrial membrane (accompanied by H+ transportation), which facilitates the malate-aspartate shuttle. The gene has also been validated by another dataset [30]. We hypothesize that the functioning of malate-aspartate shuttle can provide extra energy to cancer cells for gaining the growth advantage against native cells as well as escaping from the original site.

TUBB6 is a gene encoding a subtype of β-tubulins, the major constituent of microtube, which plays fundamental roles in cell structure maintenance, formation of the mitotic spindle, transportation of chemicals, etc. Furthermore, TUBB6 is also functionally associated with gap junctional intercellular communication (GJIC). In fact, the respective relationships between both tubulin and GJIC with metastasis of prostate/other cancers have been studied. For instance, the level of tubulin affects the metastasis of colorectal carcinoma cells [31]; and breakdowns of GJIC in a variety of cancer cells correlate their metastatic capacity [3234]. Moreover, studies have also indicated that TUBB6 itself is functionally related with the metastasis of various cancers. For instance, in a study using 60 cancer cell lines with different invasion abilities, TUBB6 is identified as an invasion-associated (IA) gene [35]. Moreover, it is also identified as one of the 38 prognostic gene expression signatures of node-positive breast cancer after systemic adjuvant chemotherapy [36]. In addition, Champine et al. have shown that one of the potential mechanisms of BRMS1-mediated metastasis suppression is the suppression of TUBB6[37].

PARM1 is named as prostate androgen-regulated mucin-like protein 1. The gene regulates telomerase protein component 1 (TLP1) expression and telomerase activity, thus enabling certain prostate cells to resist apoptosis. Multiple works have proved that PARM1 is an important causal gene of prostate cancer [3840]. It contributes to the immortalization of prostatic cancer cells, which enhances the survival advantage against the neighboring native cells and promotes metastasis. MYEF2 is myelin expression factor 2, which functions as a transcription repressor of the myelin basic protein (MBP). Our results underline the importance of the gene for prostate cancer, although no direct relationship between MYEF2 and the cancer had been established yet.

Our findings have provided a concise picture of the metastasis of prostate cancer. According to Valastyan and Weinberg, the metastatic process includes 7 steps [41]: (1) invade locally through surrounding extracellular matrix and stromal cell layers, (2) intravasate through blood vessels, (3) survive during the transportation, (4) arrest at distant organ sites, (5) extravasate into the parenchyma of distant tissues, (6) initially survive in these foreign microenvironments in order to form micrometastases, and (7) re-initiate their proliferative programs at metastatic sites. First, as a cancer cell, energy is its priority (i.e. functional relation with SLC25A22). Step (1) and (2) of metastasis need more mobility, for which tubulin will accommodate this task. PARM1 can enhance the survivability of cancer cells during the transportation and competing with the native cells in the invaded environment.

To investigate the identified surrogate genes further, we have found that they and their co-interacting genes in the PPI network contain existent or potential therapeutic targets for cancers. In fact, Conde-Pueyo et al. suggest that TUBB6 forms a sythetic lethal (SL) association with the cancer-related gene BUB1, speculating that treatments targeting the tubulin gene should be more efficient in cancers where BUB1 is mutated [42]. Moreover, tubulins are existent targets of anti-cancer drugs, e.g. Paclitaxel and vinca alkaloids (e.g. Vincristine and Vinblastine) in various cancers (including prostatic). The drugs disrupt the formations of microtubules/mitotic spindles and hence inhibit the proliferations and metastases of cancer cells [43, 44]. PARM1 is part of the Golgi apparatus that is androgen-responsive, and researches demonstrate that the Golgi apparatus embody new mechanisms of the androgen receptor (AR)-mediated signaling and they are useful biomarkers for prostate cancer diagnosis/prognosis [45]. Moreover, Golgi-targeting drugs have been shown to be effective in both androgen-dependent/-independent prostate cancers [45]. Given this foreground, PARM1 may have the potential of a therapeutic target for prostate cancer. It is also noteworthy that although the other two genes we identified (SLC25A22 and MYEF2) do not possess direct therapeutic utilities at present, they may somehow implicate theoretical clues for cancer therapies. In fact, members of the SLC25A family (e.g. SLC25A4/5/6) are existent drug targets for the treatments of bone metastases in breast cancer and metastatic bone disease [46]; since prostate cancer also exhibit bone metastasis [3], SLC25A22 may be worth being examined for potential relations to the metastatic properties of prostate cancer. Meanwhile, MYEF2 has been characterized as a downstream target modulated by the Wnt/β-catenin pathway; since inhibition of Wnt/β-catenin signaling suppresses a number of cancers (e.g. multiple myeloma, colorectal cancer, etc.) [47], the genes regulated by Wnt/β-catenin may provide insights into the mechanisms of cancer developments and therapies.

Furthermore, in the PPI partners co-identified with the surrogate genes, ESR1 (estrogen receptor 1) is a widely known therapeutic target (for selective modulators, e.g. Raloxifene, Tamoxifen, etc.) in breast cancer in female [48]; however, its roles in the prostate cancer in male have not been revealed. In addition, NR3C2 (also known as the mineralocorticoid receptor, MR) belongs to the same family with the androgen receptor (AR, also known as NR3C4). Since they are co-interacting with PARM1 and MYEF2 (Figure 2), ESR1 and NR3C2 may also participate in prostate cancer along with the existent/potential targets. Moreover, other co-identified genes via PPI are also informative for cancer researches. PCDH10 (protocadherin 10) is a potential target for demethylation drugs to achieve its reactivation, which may facilitate the therapies of a wide variety of cancers (e.g. cervical, gastric, colorectal, breast cancers and leukemias) [49, 50]. ZNF148 (also known as ZBP-89) regulates cell grwoth and apoptosis, having crucial roles in the developments of many cancers (e.g. gastric, colorectal, breast cancers). It is a potential target in cancer therapy as experiments show that ZNF148 is a tumor suppressor capable of enhancing the killing effects of several anti-cancer drugs [51]. Therefore, we hypothesize that these therapeutic targets may also have biological roles in prostate cancer, or prostate cancer may have regulators that are in common with other cancers. In addition, VIM (vimentin) is characterized as an invasion/metastasis factor in tumor cells, which is transcriptionally regulated by HIF-1[52]. GJA1 (gap junction protein, alpha 1) involves in gap junction (GJIC), which plays important roles in cancer progression/metastasis. MT-ND1 encodes a mitochondrial oxidoreductase (NADH dehydrogenase 1) involving in energy metabolism, which is intuitively crucial to cancer development. In fact, both gap junctions and mitochondria are emerging as therapeutic targets in cancers nowadays [53, 54].

The previous work of Chandran et al. has discovered hundreds of genes with differential expression profiles [11]. In order to decipher the disease more concisely, we adopt the mRMR algorithm, which can provide results with the largest parsimony. Our results have showed that prostate cancer samples can be classified with only four genes, indicating that although the cancer is a complex disease with hundreds of differentially-expressed genes, these four genes may be the primary surrogates for the mechanism (s) underlying the different cancer phases. Moreover, these four genes (along with their PPI partners) turn out having mechanistic/therapeutic implications in the prostatic or other cancers. Hence, in order to characterize the development of prostate cancer (especially metastasis) and investigate the molecular mechanism (s), these genes could firstly be focused on in further functional experiments.

Conclusion

In all, we have characterized a small-sized predictor gene set for classification of prostate cancer phases. Our results support the roles for specific genes involved in cell communication, hormone-receptor mediated pathways, and transcription regulation in (metastatic) prostate cancer. To our knowledge, the gene set we computed is of the minimal size that can rationally characterize prostate cancer phases; thus we hypothesize that these genes potentially play important roles in the molecular mechanisms of prostate cancer development. Furthermore, due to the small size, our predictor gene set can be a suitable candidate list for forthcoming functional experiments; meanwhile, it might possess potential value for other relevant studies (e.g. drug target selection).

Abbreviations

mRMR:

Minimum redundancy - maximum relevance

IFS:

Incremental feature selection

PPI:

Protein-protein interaction

NT:

Normal tissue

PT:

(tissue-confined) Primary tumor

MT:

Metastatic tumor

SLC25A22:

Solute carrier family 25 member 22

TUBB6:

Tubulin, beta 6 class V

PARM1:

Prostate androgen-regulated mucin-like protein 1

MYEF2:

Myelin expression factor 2

PPARγ:

Peroxisome proliferator-activated receptor gamma

BRMS1:

Breast cancer metastasis suppressor 1.

References

  1. American Cancer Society: Cancer Facts and Figures, 2006. 2006, Atlanta: American Cancer Society

    Google Scholar 

  2. Stewart DA, Cooper CR, Sikes RA: Changes in Extracellular Matrix (ECM) and ECM-associated proteins in the metastatic progression of prostate cancer. Reprod Biol Endocrinol. 2004, 2: 2-10.1186/1477-7827-2-2.

    Article  PubMed Central  PubMed  Google Scholar 

  3. Logothetis CJ, Lin SH: Osteoblasts in prostate cancer metastasis to bone. Nat Rev Cancer. 2005, 5 (1): 21-28. 10.1038/nrc1528.

    Article  CAS  PubMed  Google Scholar 

  4. Singh D, Febbo PG, Ross K, Jackson DG, Manola J, Ladd C, Tamayo P, Renshaw AA, D’Amico AV, Richie JP, Lander ES, Loda M, Kantoff PW, Golub TR, Sellers WR: Gene expression correlates of clinical prostate cancer behavior. Cancer Cell. 2002, 1 (2): 203-209. 10.1016/S1535-6108(02)00030-2.

    Article  CAS  PubMed  Google Scholar 

  5. Brawer MK, Deering RE, Brown M, Preston SD, Bigler SA: Predictors of pathologic stage in prostatic carcinoma. The role of neovascularity. Cancer. 1994, 73 (3): 678-687. 10.1002/1097-0142(19940201)73:3<678::AID-CNCR2820730329>3.0.CO;2-6.

    Article  CAS  PubMed  Google Scholar 

  6. Luo JH, Yu YP, Cieply K, Lin F, Deflavia P, Dhir R, Finkelstein S, Michalopoulos G, Becich M: Gene expression analysis of prostate cancers. Mol Carcinog. 2002, 33 (1): 25-35. 10.1002/mc.10018.

    Article  CAS  PubMed  Google Scholar 

  7. Chandran U, Dhir R, Ma C, Michalopoulos G, Becich M, Gilbertson J: Differences in gene expression in prostate cancer, normal appearing prostate tissue adjacent to cancer and prostate tissue from cancer free organ donors. BMC Cancer. 2005, 5 (1): 45-10.1186/1471-2407-5-45.

    Article  PubMed Central  PubMed  Google Scholar 

  8. Chetcuti A, Margan S, Mann S, Russell P, Handelsman D, Rogers J, Dong Q: Identification of differentially expressed genes in organ-confined prostate cancer by gene expression array. Prostate. 2001, 47 (2): 132-140. 10.1002/pros.1056.

    Article  CAS  PubMed  Google Scholar 

  9. Dhanasekaran SM, Barrette TR, Ghosh D, Shah R, Varambally S, Kurachi K, Pienta KJ, Rubin MA, Chinnaiyan AM: Delineation of prognostic biomarkers in prostate cancer. Nature. 2001, 412 (6849): 822-826. 10.1038/35090585.

    Article  CAS  PubMed  Google Scholar 

  10. Holzbeierlein J, Lal P, LaTulippe E, Smith A, Satagopan J, Zhang L, Ryan C, Smith S, Scher H, Scardino P, Reuter V, Gerald WL: Gene expression analysis of human prostate carcinoma during hormonal therapy identifies androgen-responsive genes and mechanisms of therapy resistance. Am J Pathol. 2004, 164 (1): 217-227. 10.1016/S0002-9440(10)63112-4.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  11. Chandran U, Ma C, Dhir R, Bisceglia M, Lyons-Weiler M, Liang W, Michalopoulos G, Becich M, Monzon F: Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process. BMC Cancer. 2007, 7 (1): 64-10.1186/1471-2407-7-64.

    Article  PubMed Central  PubMed  Google Scholar 

  12. La Tulippe E, Satagopan J, Smith A, Scher H, Scardino P, Reuter V, Gerald WL: Comprehensive gene expression analysis of prostate cancer reveals distinct transcriptional programs associated with metastatic disease. Cancer Res. 2002, 62 (15): 4499-4506.

    CAS  Google Scholar 

  13. Ma C, Dong X, Li R, Liu L: A computational study identifies HIV progression-related genes using mRMR and shortest path tracing. PLoS One. 2013, 8 (11): e78057-10.1371/journal.pone.0078057.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Li BQ, Huang T, Liu L, Cai YD, Chou KC: Identification of colorectal cancer related genes with mRMR and shortest path in protein-protein interaction network. PLoS One. 2012, 7 (4): e33393-10.1371/journal.pone.0033393.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Zhang N, Jiang M, Huang T, Cai YD: Identification of influenza a/H7N9 virus infection-related human genes based on shortest paths in a virus-human protein interaction network. Biomed Res Int. 2014, 2014: 239462-Epub

    PubMed Central  PubMed  Google Scholar 

  16. Jiang M, Chen Y, Zhang Y, Chen L, Zhang N, Huang T, Cai YD, Kong X: Identification of hepatocellular carcinoma related genes with k-th shortest paths in a protein-protein interaction network. Mol Biosyst. 2013, 9 (11): 2720-2728. 10.1039/c3mb70089e.

    Article  CAS  PubMed  Google Scholar 

  17. Chou KC: Some remarks on protein attribute prediction and pseudo amino acid composition. J Theor Biol. 2011, 273 (1): 236-247. 10.1016/j.jtbi.2010.12.024.

    Article  CAS  PubMed  Google Scholar 

  18. Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, Von Mering C, Jensen LJ: STRING v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 2013, 41 (D1): D808-D815. 10.1093/nar/gks1094.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. De Jay N, Papillon-Cavanagh S, Olsen C, El-Hachem N, Bontempi G, Haibe-Kains B: mRMRe: an R package for parallelized mRMR ensemble feature selection. Bioinformatics. 2013, 29 (18): 2365-2368. 10.1093/bioinformatics/btt383.

    Article  CAS  PubMed  Google Scholar 

  20. Chou KC, Shen HB: Predicting eukaryotic protein subcellular location by fusing optimized evidence-theoretic K-nearest neighbor classifiers. J Proteome Res. 2006, 5 (8): 1888-1897. 10.1021/pr060167c.

    Article  CAS  PubMed  Google Scholar 

  21. Csardi G, Nepusz T: The igraph software package for complex network research. Inter Journal Complex Syst. 2006, 1695: Article No:1695

    Google Scholar 

  22. Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T: Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics. 2011, 27 (3): 431-432. 10.1093/bioinformatics/btq675.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  23. Huang DW, Sherman BT, Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2008, 4 (1): 44-57. 10.1038/nprot.2008.211.

    Article  Google Scholar 

  24. Feldman BJ, Feldman D: The development of androgen-independent prostate cancer. Nat Rev Cancer. 2001, 1 (1): 34-45. 10.1038/35094009.

    Article  CAS  PubMed  Google Scholar 

  25. McLeod DG: Hormonal therapy: historical perspective to future directions. Urology. 2003, 61 (2): 3-7. 10.1016/S0090-4295(02)02393-2.

    Article  PubMed  Google Scholar 

  26. Ruch RJ: The role of gap junctional intercellular communication in neoplasia. Ann Clin Lab Sci. 1994, 24 (3): 216-231.

    CAS  PubMed  Google Scholar 

  27. Hotz-Wagenblatt A, Shalloway D: Gap junctional communication and neoplastic transformation. Crit Rev Oncogen. 1993, 4 (5): 541-558.

    CAS  Google Scholar 

  28. Tsai H, Werber J, Davia MO, Edelman M, Tanaka KE, Melman A, Christ GJ, Geliebter J: Reduced connexin 43 expression in high grade, human prostatic adenocarcinoma cells. Biochem Biophys Res Commun. 1996, 227 (1): 64-69. 10.1006/bbrc.1996.1468.

    Article  CAS  PubMed  Google Scholar 

  29. Habermann H, Ray V, Habermann W, Prins GS: Alterations in gap junction protein expression in human benign prostatic hyperplasia and prostate cancer. J Urol. 2002, 167 (2): 655-660. 10.1016/S0022-5347(01)69118-3.

    Article  PubMed  Google Scholar 

  30. Cermák V, Kosla J, Plachý J, Trejbalová K, Hejnar J, Dvořák M: The transcription factor EGR1 regulates metastatic potential of v-src transformed sarcoma cells. Cell Mol Life Sci. 2010, 67 (20): 3557-3568. 10.1007/s00018-010-0395-6.

    Article  PubMed  Google Scholar 

  31. Schaefer KL, Takahashi H, Morales VM, Harris G, Barton S, Osawa E, Nakajima A, Saubermann LJ: PPARγ inhibitors reduce tubulin protein levels by a PPARγ, PPARδ and proteasome-independent mechanism, resulting in cell cycle arrest, apoptosis and reduced metastasis of colorectal carcinoma cells. Int J Cancer. 2007, 120 (3): 702-713. 10.1002/ijc.22361.

    Article  CAS  PubMed  Google Scholar 

  32. Navolotski A, Rumjnzev A, Lu H, Proft D, Bartholmes P, Zanker KS: Migration and gap junctional intercellular communication determine the metastatic phenotype of human tunor cell lines. Cancer Lett. 1997, 118 (2): 181-187. 10.1016/S0304-3835(97)00329-7.

    Article  CAS  PubMed  Google Scholar 

  33. Nicolson GL, Dulski KM, Trosko JE: Loss of intercellular junctional communication correlates with metastatic potential in mammary adenocarcinoma cells. Proc Natl Acad Sci U S A. 1988, 85 (2): 473-476. 10.1073/pnas.85.2.473.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Saunders MM, Seraj MJ, Li Z, Zhou Z, Winter CR, Welch DR, Donahue HJ: Breast cancer metastatic potential correlates with a breakdown in homospecific and heterospecific gap junctional intercellular communication. Cancer Res. 2001, 61 (5): 1765-1767.

    CAS  PubMed  Google Scholar 

  35. Hsu YC, Chen HY, Yuan S, Yu SL, Lin CH, Wu G, Yang PC, Li KC: Genome-wide analysis of three-way interplay among gene expression, cancer cell invasion and anti-cancer compound sensitivity. BMC Med. 2013, 11 (1): 106-10.1186/1741-7015-11-106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Jézéquel P, Campone M, Roché H, Gouraud W, Charbonnel C, Ricolleau G, Magrangeas F, Minvielle S, Genève J, Martin AL, Bataille R, Campion L: A 38-gene expression signature to predict metastasis risk in node-positive breast cancer after systemic adjuvant chemotherapy: a genomic substudy of PACS01 clinical trial. Breast Cancer Res Treat. 2009, 116 (3): 509-520. 10.1007/s10549-008-0250-8.

    Article  PubMed  Google Scholar 

  37. Champine PJ, Michaelson J, Weimer BC, Welch DR, DeWald DB: Microarray analysis reveals potential mechanisms of BRMS1-mediated metastasis suppression. Clin Exp Metastasis. 2007, 24 (7): 551-565. 10.1007/s10585-007-9092-8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Cornet AM, Hanon E, Reiter ER, Bruyninx M, Nguyen VH, Hennuy BR, Hennen GP, Closset JL: Prostatic androgen repressed message-1 (PARM-1) may play a role in prostatic cell immortalisation. Prostate. 2003, 56 (3): 220-230. 10.1002/pros.10254.

    Article  CAS  PubMed  Google Scholar 

  39. Fladeby C, Gupta SN, Barois N, Lorenzo PI, Simpson JC, Saatcioglu F, Bakke O: Human PARM-1 is a novel mucin-like, androgen-regulated gene exhibiting proliferative effects in prostate cancer cells. Int J Cancer. 2008, 122 (6): 1229-1235.

    Article  CAS  PubMed  Google Scholar 

  40. Wang XY, Hao JW, Zhou RJ, Zhang XS, Yan TZ, Ding DG, Shan L: Meta-analysis of gene expression data identifies causal genes for prostate cancer. Asian Pac J Cancer Prev. 2013, 14 (1): 457-461. 10.7314/APJCP.2013.14.1.457.

    Article  PubMed  Google Scholar 

  41. Valastyan S, Weinberg RA: Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011, 147 (2): 275-292. 10.1016/j.cell.2011.09.024.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Conde-Pueyo N, Munteanu A, Sole R, Rodriguez-Caso C: Human synthetic lethal inference as potential anti-cancer target gene detection. BMC Syst Biol. 2009, 3 (1): 116-10.1186/1752-0509-3-116.

    Article  PubMed Central  PubMed  Google Scholar 

  43. Katsetos CD, Dráber P: Tubulins as therapeutic targets in cancer: from bench to bedside. Curr Pharm Des. 2012, 18 (19): 2778-2792. 10.2174/138161212800626193.

    Article  CAS  PubMed  Google Scholar 

  44. Jordan A, Hadfield JA, Lawrence NJ, McGown AT: Tubulin as a target for anticancer drugs: agents which interact with the mitotic spindle. Med Res Rev. 1998, 18 (4): 259-296. 10.1002/(SICI)1098-1128(199807)18:4<259::AID-MED3>3.0.CO;2-U.

    Article  CAS  PubMed  Google Scholar 

  45. Migita T, Inoue S: Implications of the Golgi apparatus in prostate cancer. Int J Biochem Cell Biol. 2012, 44 (11): 1872-1876. 10.1016/j.biocel.2012.06.004.

    Article  CAS  PubMed  Google Scholar 

  46. Rask-Andersen M, Masuram S, Fredriksson R, Schiöth HB: Solute carriers as drug targets: current use, clinical trials and prospective. Mol Aspects Med. 2013, 34 (2–3): 702-710.

    Article  CAS  PubMed  Google Scholar 

  47. Yao H, Ashihara E, Strovel JW, Nakagawa Y, Kuroda J, Nagao R, Tanaka R, Yokota A, Takeuchi M, Hayashi Y, Shimazaki C, Taniwaki M, Strand K, Padia J, Hirai H, Kimura S, Maekawa T: AV-65, a novel Wnt/β-catenin signal inhibitor, successfully suppresses progression of multiple myeloma in a mouse model. Blood Cancer J. 2011, 1 (11): e43-10.1038/bcj.2011.41.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  48. Holst F, Stahl PR, Ruiz C, Hellwinkel O, Jehan Z, Wendland M, Lebeau A, Terracciano L, Al-Kuraya K, Janicke F, Sauter G, Simon R: Estrogen receptor alpha (ESR1) gene amplification is frequent in breast cancer. Nat Genet. 2007, 39 (5): 655-660. 10.1038/ng2006.

    Article  CAS  PubMed  Google Scholar 

  49. Narayan G, Freddy AJ, Xie D, Liyanage H, Clark L, Kisselev S, Un Kang J, Nandula SV, McGuinn C, Subramaniyam S, Alobeid B, Satwani P, Savage D, Bhagat G, Murty VV: Promoter methylation-mediated inactivation of PCDH10 in acute lymphoblastic leukemia contributes to chemotherapy resistance. Genes Chromosomes Cancer. 2011, 50 (12): 1043-1053. 10.1002/gcc.20922.

    Article  CAS  PubMed  Google Scholar 

  50. Ying J, Li H, Seng TJ, Langford C, Srivastava G, Tsao SW, Putti T, Murray P, Chan ATC, Tao Q: Functional epigenetics identifies a protocadherin PCDH10 as a candidate tumor suppressor for nasopharyngeal, esophageal and multiple other carcinomas with frequent methylation. Oncogene. 2006, 25 (7): 1070-1080. 10.1038/sj.onc.1209154.

    Article  CAS  PubMed  Google Scholar 

  51. Zhang CZ, Chen GG, Lai PB: Transcription factor ZBP-89 in cancer growth and apoptosis. Biochim Biophys Acta. 2010, 1806 (1): 36-41.

    CAS  PubMed  Google Scholar 

  52. Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003, 3 (10): 721-732. 10.1038/nrc1187.

    Article  CAS  PubMed  Google Scholar 

  53. Kandouz M, Batist G: Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets. 2010, 14 (7): 681-692. 10.1517/14728222.2010.487866.

    Article  CAS  PubMed  Google Scholar 

  54. Don AS, Hogg PJ: Mitochondria as cancer drug targets. Trends Mol Med. 2004, 10 (8): 372-378. 10.1016/j.molmed.2004.06.005.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This study is supported by the Ministry of Science and Technology Grant of P.R. China, No. 2012AA02A602.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lei Liu.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Conceiving the research: RL, XD, CM and LL. Data analysis: RL, XD and CM. Manuscript writing: RL, XD, CM and LL. All authors read and approved the final manuscript.

Rudong Li, Xiao Dong, Chengcheng Ma contributed equally to this work.

Electronic supplementary material

12976_2014_481_MOESM1_ESM.xlsx

Additional file 1: Table S1: Prediction accuracies for each individuals in IFS. The file includes detailed prediction data for the normal condition (denoted by“A”), primary tumor (“B”) and metastatic tumor (“C”). “Prediction 1 - 3” have the same meaning as in Figure 1, and the prediction statuses are shown. The file is in the format of electronic data sheet (Microsoft Excel *.xlsx). (XLSX 17 KB)

12976_2014_481_MOESM2_ESM.xlsx

Additional file 2: Table S2: Expression profiles of the four mRMR-selected genes. The file shows expression levels of the four mRMR-selected genes in the three class of samples. The file is in the format of Microsoft Excel *.xlsx. (XLSX 10 KB)

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, R., Dong, X., Ma, C. et al. Computational identification of surrogate genes for prostate cancer phases using machine learning and molecular network analysis. Theor Biol Med Model 11, 37 (2014). https://doi.org/10.1186/1742-4682-11-37

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1742-4682-11-37

Keywords