Skip to main content

CoMI: consensus mutual information for tissue-specific gene signatures

Abstract

Background

The gene signatures have been considered as a promising early diagnosis and prognostic analysis to identify disease subtypes and to determine subsequent treatments. Tissue-specific gene signatures of a specific disease are an emergency requirement for precision medicine to improve the accuracy and reduce the side effects. Currently, many approaches have been proposed for identifying gene signatures for diagnosis and prognostic. However, they often lack of tissue-specific gene signatures.

Results

Here, we propose a new method, consensus mutual information (CoMI) for analyzing omics data and discovering gene signatures. CoMI can identify differentially expressed genes in multiple cancer omics data for reflecting both cancer-related and tissue-specific signatures, such as Cell growth and death in multiple cancers, Xenobiotics biodegradation and metabolism in LIHC, and Nervous system in GBM. Our method identified 50-gene signatures effectively distinguishing the GBM patients into high- and low-risk groups (log-rank pā€‰=ā€‰0.006) for diagnosis and prognosis.

Conclusions

Our results demonstrate that CoMI can identify significant and consistent gene signatures with tissue-specific properties and can predict clinical outcomes for interested diseases. We believe that CoMI is useful for analyzing omics data and discovering gene signatures of diseases.

Background

Genome-wide gene expression profiling has been used to identify genetic signatures that could be associated with the outcome of cancer patients [1]. Several different gene signatures have been developed, and many of these approaches have been shown to better define the prognosis of cancer patients as compared with conventional clinical and pathological characteristics of the tumors [1]. Some studies began with genome-wide gene expression profiling from microarray datasets or next-generation sequencing (NGS). For example, the PAM50 gene signature can use to classify breast tumors into one of these four subtypes and to predict clinical outcomes [2, 3]. Moreover, the gene signature identified from specific tissues is a promising avenue to maximize efficacy in target tissues while minimizing the safety risks of affecting unrelated tissues [4].

Many methods have been proposed to identify the gene signatures between normal and disease states [5]. The simple and common way is to use the T-test, which is a statistics-based method. According to the distributions between two states, the T-test evaluates the probability (p-value) in each gene. The significant difference of gene expression is often defined if the probability is less than 0.05 or 0.01. Another method is calculating the fold change (FC) of gene expression, which is not a statistical test and has no associated values for indicating the level of confidence in the genes as differentially expressed or not [6]. Therefore, some methods were developed, such as Significance Analysis of Microarrays (SAM) and Cancer Outlier Profile Analysis (COPA), to choose the biomarkers in high confidence [7,8,9]. The SAM approach proposed false discovery rate (FDR) to reduce genes showing significantly different expression by chance (i.e., false positive) [7, 10]. The COPA, based on the hypothesis of tumor heterogeneity, identified candidate genes overexpressing in subsets of samples by using median and median absolute deviation of gene expression profiles [8]. In another study, Parker et al. determined 50 genes (i.e., PAM50) for classified four breast subtypes with clinical means by using Predictive Analysis of Microarray (PAM) algorithm [2, 11], but this approach relied on sample labels that was difficult to propose new subtype of diseases. Recently, Gentles et al. applied CIBERSORT computational methods and PRECOG tools to identify cancer prognostic biomarkers and therapeutic targets [12], however, they only focused on the genes that related to clinical outcomes, but the ones might not be suitable for diagnosis due to the non-significant gene expression changes between the sample subgroups [13].

In this study, we hypothesize that the biological processes of tissues/organs are dysregulated during tumorigenesis, and the perturbed genes (i.e., tissue-specific genes) are often involved in corresponding functions of tissues [14,15,16]. To address these issues, we propose consensus mutual information (CoMI) to analyze omics data and to identify gene signatures. We utilized mutual information (MI) and gene expression distance as the basis to find the significantly and consistently expressed genes and gene signatures between normal and disease states. For multiple cancer omics data, our identified gene signatures could reflect cancer-related signatures and have tissue-specific properties (mean odds ratioā€‰=ā€‰2.89). Based on our previously developed global omics data analysis method [17], our CoMI identified gene signatures not only involved in common cancer-related progress, such as Cell growth and death, but also reflect tissue unique functions of Xenobiotics biodegradation and metabolism in LIHC and Nervous system in GBM. For clinical prognosis, a 50-gene signature identified by CoMI could distinguish the GBM patients into high- and low-risk groups based on gene expression patterns, and could predict clinical outcomes at 12-month survival (log-rank pā€‰=ā€‰0.006). We believe that our method and results are useful for analyzing omics data, discovering gene signatures with tissue-specific properties, and predicting clinical outcomes of diseases.

Results

CoMI for identifying gene signatures in multiple cancers

To identify consistent patterns of gene expression for gene signatures development in multiple cancers, we utilized CoMI to analyze genome-wide gene expression profiles in LIHC, GBM, BLCA, BRCA, and COAD. To evaluate the cancer associations of selected genes from CoMI in multiple cancer datasets, we collected 1675 cancer-related genes derived from HPA database [18]. The results show that the gene signatures selected from the different ranking cut-off of CoMI have a high probability to be cancer-related genes compared with T-test, FC, and Significance Analysis of Microarrays (SAM) (Fig.Ā 1). For example, the top-ranked 200 significant genes were 190, 182, 181, and 126 genes for CoMI, T-test, FC, and SAM, respectively.

Fig. 1
figure 1

The prediction accuracies between CoMI, T-test, FC, and SAM in cancer-related genes. The mean precision of cancer-related genes recorded in HPA was selected by CoMI (red line), T-test (blue line), FC (black line), and SAM (yellow line). The predicting precision was the average of genes selected from different cut-offs in LIHC, GBM, BLCA, BRCA, and COAD

To investigate the statistical meanings of CoMI, we calculated Spearman's correlation coefficient (Ļ) between CoMI scores and FC values, SAM scores and p-values of T-test on all of 20,531 genes in NGS profiles of TCGA, respectively (Additional file 1: Fig. S1). For these methods, the average Ļ values of these five cancer types were 0.81 (FC), 0.95 (T-test), and 0.94 (SAM). These results suggest that CoMI has statistical meanings and is able to identify significantly and consistently expressed genes and disease-related gene signatures from omics data.

Tissue-specific properties of gene signatures

To investigate the biological meanings of gene signatures identified by CoMI in different cancers, we collected tissue-specific genes from HPA with protein annotation of tissue specificity in liver, brain, urinary bladder, breast, or colon. Here, we used the odds ratio to assess whether gene signatures have tissue-specific properties or not in five cancers (Fig.Ā 2 and Additional file 1: Fig. S2). In the comparison of CoMI and T-test, the average odds ratios of gene signatures selected from eight kinds of top-ranked thresholds were 2.89, 5.40, 1.90, 1.74, and 2.99 in LIHC, GBM, BLCA, BRCA, and COAD, respectively. For example, the top-ranked 200 genes selected by CoMI and T-test in GBM were 89 and 23 with annotated brain specificity, respectively. The odds were 0.8 (89/111) and 0.13 (23/177), then, the odds ratio was calculated as 6.15 (0.8/0.13). The results were similar when compared to SAM (Additional file 1: Fig. S2). Besides, we observed that CoMI has a lower ranking on average of descending order than the ones of T-test and SAM in all of the tissue-specific genes in each cancer (Additional file 1: Figs. S3 and S4). These results demonstrate that gene signatures identified by CoMI are more like to have tissue-specific properties.

Fig. 2
figure 2

The odds ratios of tissue-specific genes selected by CoMI and T-test in different cancer types. The results of tissue-specific genes identified by CoMI and T-test in five cancer types, including LIHC (blue), GBM (orange), BLCA (yellow), BRCA (purple), and COAD (green)

Furthermore, we used our previously developed global omics data analysis method, Hierarchical System Biology Model (HiSBiM), to investigate the involved pathways as well as biological subsystems and systems of gene signatures [17]. We observed that biological subsystems of the 200-gene signatures identified by CoMI and T-test were involved in common cancer-related pathways, such as Cell growth and death and Replication and repair in five cancer types (Fig.Ā 3). In particular, the gene signature of LIHC identified by CoMI was enriched in Xenobiotics biodegradation and metabolism (meta-z scoreā€‰=ā€‰3.72) and Lipid metabolism (meta-z scoreā€‰=ā€‰2.74), which could reflect unique functions to liver tissue. In GBM, we observed that neurotransmission-related functions were highly enriched, such as Nervous system (meta-z scoreā€‰=ā€‰7.49) and Signaling molecules and interaction (meta-z scoreā€‰=ā€‰2.46). In addition, the Digestive system (meta-z scoreā€‰=ā€‰7.03) was highly regulated in COAD.

Fig. 3
figure 3

The genes identified from different cancers reflected tissue-specific pathways. The subsystem-level meta-z scores of the top-ranked 200 genes identified by CoMI and T-test in A LIHC, B GBM, C BLCA, D BRCA, and E COAD. The red triangle was denoted if meta-z scoresā€‰>ā€‰2 and CoMIā€‰>ā€‰T-test, the green square was represented if meta-z scoresā€‰>ā€‰2 and T-testā€‰>ā€‰CoMI

We also evaluated the tissue-specific properties of CoMI and COPA [9] based on HPA database and HiSBiM analysis. The results show that the genes identified by CoMI are related to both common cancer-related pathways and tissue-specific properties, such as Endocrine system (meta-z scoreā€‰=ā€‰5.40) in BRCA (Additional file 1: Fig. S5). Additionally, CoMI outperformed COPA and our results indicated that CoMI not only can identify tissue-specific gene signatures in different cancers, but also can reflect corresponding biological pathways and functions unique to those tissues.

Clinical prognosis of gene signatures

The grade IV astrocytomas (i.e., GBM) are an aggressive class of brain cancer, it is difficult to treat and has a poor median 12-month overall survival, and the urgent need to develop a prognostic gene signature [19]. We selected a 50-gene signature with significant and consistent expressions by using our CoMI from the gene expression profile in GBM, as well as, five of those genes were brain tissue specificity (Table 1). According to the gene expression patterns, we found that these 50 genes could cluster 156 tumor samples into four groups (Fig.Ā 4A). According to the 12-month overall survival analysis, the patients in GBM-C1 group (nā€‰=ā€‰15) have a significantly lower survival probability (30%) than GBM-C3 group (62%; log-rank pā€‰=ā€‰0.006; Fig.Ā 4B). Moreover, we found that 70% of CoMI top-ranked 10 genes can distinguish patients into high- and low-risk groups (log-rank pā€‰<ā€‰0.01), such as PBK (log-rank pā€‰=ā€‰0.0058) and CCNB2 (log-rank pā€‰=ā€‰0.009), however, only five of 10 genes with log-rank pā€‰<ā€‰0.01 in T-test (Additional file 1: Fig. S6). These results indicated that our identified 50-gene signature could predict clinical outcomes and provide the available clues for developing the new therapeutic strategies in GBM.

Table 1 The 50-gene signature identified by CoMI in GBM
Fig. 4
figure 4

Glioblastoma multiforme prognostic gene signature. A The hierarchal clustering of a 50-gene signature identified by CoMI in glioblastoma multiforme (GBM) tumor samples. Based on the gene expression patterns, the expression profile could be divided into four groups, including C1 (blue), C2 (green), C3 (orange), and C4 (purple). B The 12-month overall survival analysis in four groups of GBM patients. The inner table showed p-value of log-rank test compared between four groups of patients.

Discussion

Genome-wide gene expression profiling has been used to identify genetic signatures that could be associated with the outcome of cancer patients [1]. Some studies using genome-wide gene expression profiling for developing gene signatures, and many of these approaches have shown to better define the prognosis of cancer patients, such as the PAM50 gene signature can use to classify breast tumors into four subtypes and to predict clinical prognosis [2, 3].

In this paper, we propose consensus mutual information (CoMI) to analyze omics data and to identify gene signatures. For multiple cancer omics data, we used CoMI to identify gene signatures, and those genes could reflect cancer-related signatures and have tissue-specific properties. In general, normal cells perform the function they are meant to perform, whereas cancer cells may not execute these functions. For example, cancerous thyroid cells may not produce thyroid hormone [20], and cancerous white blood cells are not functioning as they should [21]. On the other hand, the genes recorded in cancer hallmarks are often activated in cancer cells [22].

Here, we hypothesize that tissue-specific genes are often related to lost-of-tissue functions which are often down-regulated in the disease states. Our CoMI can identify these tissue-specific genes which are significant change (SDist score) and have consensus expressed values (SMI) within cancer and normal samples. The changes between cancer and normal samples were quantified by considering global gene expression values for all genes in omics data. Based on our scoring function, CoMI identified the liver-specific genes, such as CYP1A2 (CoMI scoreā€‰=ā€‰0.99 and FCā€‰=ā€‰āˆ’Ā 7.76), CYP2C8 (CoMI scoreā€‰=ā€‰0.71 and FCā€‰=ā€‰āˆ’Ā 4.14), and CYP3A4 (CoMI scoreā€‰=ā€‰0.55 and FCā€‰=ā€‰āˆ’Ā 5.85), are the members of the cytochrome P450 family involving in Lipid metabolism (meta-z scoreā€‰=ā€‰2.74) as well as Xenobiotics biodegradation and metabolism (meta-z scoreā€‰=ā€‰3.72) in LIHC. Additionally, CoMI also identified gene SLC22A1 (CoMI scoreā€‰=ā€‰0.53 and FCā€‰=ā€‰āˆ’Ā 4.81) involving in liver unique functions, that is, bile secretion of Digestive system (meta-z scoreā€‰=ā€‰4.54). Conversely, MAP2K1, an essential component of the MAP kinase signal transduction pathway related to cancer hallmark, was not a tissue-specific gene. We found that MAP2K1 significantly changed in most cancers, but its expression values are not consistent (CoMI scoreā€‰=ā€‰0.20). These results show that our CoMI identified gene signatures not only involved in cancer-related progress, such as Cell growth and death, but also reflect tissue unique functions of Nervous system (meta-z scoreā€‰=ā€‰7.49) in GBM. For clinical prognosis, our identified 50-gene signature could stratify GBM patients into high- and low-risk groups, and could predict clinical outcomes with 12-month survival.

There were some limitations to the current study. Firstly, the omics data (i.e., NGS) is only collected from TCGA, and different sources and platforms, such as microarray, are needed to use to validate our method and identified gene signatures. Secondly, the results of this study are mostly based on bioinformatics analysis and predictions, and further experiments are needed to prove the tissue-specific properties of our identified gene signatures. Thirdly, although we identified prognostic gene signatures, those genes remain to be further explored in our future work.

Conclusion

In summary, we have proposed CoMI for analyzing omics data and discovering gene signatures. Our method accomplished the identification of genes and gene signatures, which have consistently and significantly changed between normal and disease states. Our results indicated that CoMI could identify gene signatures with tissue-specific properties for interested diseases, and is able to be applied to predict clinical prognosis.

Methods

To identify significantly and consistently expressed genes and gene signatures, we proposed a method, consensus mutual information (called CoMI), for analyzing omics data between normal and disease samples (Fig.Ā 5A). We first calculated gene expression variations for each gene in a given omics data (Fig.Ā 5B). For the evaluating of the consensus gene expression in normal and tissue states, we transferred the continuous gene expression to discrete integer symbols based on expression variations and intensities (Fig.Ā 5C). Finally, we computed CoMI score for all genes, which can be used as a measure for significantly and consistently expressed genes and gene signatures between two states (Fig.Ā 5D).

Fig. 5
figure 5

Overview of consensus mutual information. A Flowchart describing the main procedure. B We firstly evaluated the standard deviation (\(\sigma_{i}\)) of gene expression intensity for each gene in a given omics data. C We then computing the average standard deviation (\(\overline{\sigma }\)) from \(\sigma_{i}\) of all genes, as well as expression means of normal and disease samples (i.e., Ī¼N and Ī¼D), respectively, in each gene. For a given gene i in sample j, we assigned expression intensity into 7 integral symbols by considering the \(\overline{\sigma }\) and its Ī¼N and Ī¼D. D The gene expression values were converted to discretized integer symbol ranging from 0 to 6. The highly expressed genes were assigned to the highest symbol 6 and lowly expressed genes were assigned to the lowest symbol 0. (D) The calculation of consensus mutual information (CoMI) values of all genes

Omics and validation data

Here, we collected omics data (i.e. NGS) in LIHC, GBM, BLCA, BRCA, and COAD from The Cancer Genome Atlas (TCGA) databases [23]. These datasets contain 228 normal and 2315 tumor samples. To evaluate our method and compare to T-test and FC method, we collected the 1675 cancer-related genes derived from the Human Protein Atlas (HPA) database [18]. The tissue-specific genes were collected from HPA with protein annotation of tissue specificity in liver (409 genes), brain (1313 genes), urinary bladder (56 genes), breast (82 genes), or colon (147 genes). Finally, we calculated the p-values (i.e., T-test), fold changes, and SAM scores of all genes by using limma and samr R package [24, 25].

Discretization

To consider the readily quantifiable and significant expressions in the disease state of genes and gene signatures, we firstly evaluated the standard deviation (\(\sigma_{i}\)) of expression intensity of the gene i (Fig.Ā 5B). Then, we calculated the average standard deviation (\(\overline{\sigma }\)) from \(\sigma_{i}\) of all genes (Fig.Ā 5C). For each gene, computing expression means of normal (Ī¼N) and disease (Ī¼D) samples, respectively. For a given gene i in sample j, we utilize the average standard deviation (\(\overline{\sigma }\)), average expression (Ī¼N,i and Ī¼D,i) to assign expression intensity (EIi,j) into an integral symbol (ESi,j) by using the following equations:

$$ES_{i,j} = \left\{ \begin{aligned} & 6, \;\;if EI_{i,j} > \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 2.5\overline{\sigma } \\ & 5, \;\;if \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 1.5\overline{\sigma } < EI_{i,j} \le \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 2.5\overline{\sigma } \\ & 4, \;\;if \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 0.5\overline{\sigma } < EI_{i,j} \le \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 1.5\overline{\sigma } \\ & 3, \;\;if \frac{{\mu_{D,i} +\mu_{N,i} }}{2} - 0.5\overline{\sigma } < EI_{i,j} \le \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 0.5\overline{\sigma } \\ & 2, \;\;if \frac{{\mu_{D,i} +\mu_{N,i} }}{2} - 1.5\overline{\sigma } < EI_{i,j} \le \frac{{\mu_{D,i} +\mu_{N,i} }}{2} - 0.5\overline{\sigma } \\ & 1, \;\;if \frac{{\mu_{D,i} +\mu_{N,i} }}{2} - 2.5\overline{\sigma } < EI_{i,j} \le \frac{{\mu_{D,i} +\mu_{N,i} }}{2} - 1.5\overline{\sigma } \\ & 0,\;\; if EI_{i,j} \le \frac{{\mu_{D,i} +\mu_{N,i} }}{2} - 2.5\overline{\sigma } \\ \end{aligned} \right.$$
(1)

The gene expression values were converted to discretized integer symbols ranging from 0 to 6. The highly expressed gene was assigned to the symbol 6 and lowly expressed gene was assigned to the symbol 0. For instance, the \(\overline{\sigma }\) is 1.04 in LIHC, and the Ī¼N and Ī¼D of the gene NAT2 are 10.4 and 5.7, respectively. The expression intensity (EI) of NAT2 is 10.8 in sample TCGA-DD-AAE3-01A-11R-A41C-07, which satisfies the \(EI_{i,j} > \frac{{\mu_{D,i} +\mu_{N,i} }}{2} + 2.5\overline{\sigma }\) (i.e., 10.7), therefore, we assign the ESNAT2,TCGA-DD-AAE3-01A-11R-A41C-07 value to 6.

CoMI: Consensus mutual information

For each gene i in a given omics data, we evaluated its consensus mutual information (CoMI) and to identify significantly and consistently expressed genes and gene signatures between normal and disease states (Fig.Ā 5D). The CoMI of a gene is defined as:

$$CoMI = S_{MI} \times S_{Dist}$$
(2)

where SMI is gene expression difference between two states using mutual information; SDist is the gene expression distance between two states by using mean distance. For the gene i, the SMI is given as:

$$S_{MI} = \mathop \sum \limits_{y = 1}^{Y} \mathop \sum \limits_{x = 1}^{X} p\left( {x,y} \right){\text{log}}\left( {\frac{{p\left( {x,y} \right)}}{p\left( x \right)p\left( y \right)}} \right)$$
(3)

where p(x,y) is the probability of gene i in symbol x and state y; p(x) is the fraction of gene i in symbol x, and p(y) is the fraction of samples in state y. Y is the number of states (here, Yā€‰=ā€‰2 for normal and disease states), and X is the number of symbols (here, Xā€‰=ā€‰7). The SDist is given as:

$$S_{Dist} = \left( {\frac{{\mathop \sum \nolimits_{n \in y1} ES_{i,n} }}{N} - \frac{{\mathop \sum \nolimits_{d \in y2} ES_{i,d} }}{D}} \right)$$
(4)

where N and D are numbers of samples in the normal (y1) and disease (y2) states, respectively; \(ES_{i,n}\) and \(ES_{i,d}\) are the integral symbols of gene i at the sample for normal and disease states, respectively.

According to the equation of mutual information, the SMI is related to the ratio between normal and cancer samples. SMI is also related to the overlap of integer symbols between normal and cancer samples. For example, SMIā€‰=ā€‰1, while p(normal)ā€‰=ā€‰p(cancer) and there is no overlap of integer symbols between normal and cancer samples; SMI is from 0.3 to 0.5, while p(normal)ā€‰=ā€‰p(cancer) and half of the integer symbols in normal and cancer samples are the same; SMIā€‰=ā€‰1, while p(normal)ā€‰=ā€‰p(cancer) and there is no overlap of integer symbols between normal and cancer samples; SMIā€‰=ā€‰0.44, while 10ā€‰Ć—ā€‰p(normal)ā€‰=ā€‰p(cancer) and there is no overlap of integer symbols between normal and cancer samples; SMI is from 0.3 to 0.4, while 10ā€‰Ć—ā€‰p(normal)ā€‰=ā€‰p(cancer) and half of the integer symbols in normal samples are the same as integer symbols in cancer samples; SMIā€‰=ā€‰0.28, while 20ā€‰Ć—ā€‰p(normal)ā€‰=ā€‰p(cancer) and there is no overlap of integer symbols between normal and cancer samples. In our collected NGS data in LIHC, GBM, BLCA, BRCA, and COAD from TCGA databases, there are 228 normal and 2315 tumor samples and the expected maxima of SMI might be 0.44. Here, we assumed that CoMIā€‰>ā€‰0.6 might be a suitable cut-off which could ensure only half of the integer symbols in normal samples are the same as integer symbols in cancer samples and the SDist is greater than 2 (Additional file 1: Fig. S7).

Moreover, we found that both SMI and SDist could be used as good indexes to identify the genes with tissue-specific properties (Additional file 1: Fig. S8). SDist focuses on the distance between two states and is more related to the tissue-specific genes in our five data sets. In this study, we provide a scoring system that has a reliable discretizing method and consider both distances between two states and mutual information to identify gene signatures. Using SMI and SDist could evaluate the distance, significantly and consistently expressed value of the gene between normal and disease states under the same scale.

Availability of data and materials

The omics data of LIHC, GBM, BLCA, BRCA, and COAD and clinical information were collected from TCGA cohort of Genomic Data Commons Data Portal database (https://portal.gdc.cancer.gov/). Kaplanā€“Meier analysis was performed by using R tool which could be obtained from R Project (https://www.r-project.org/).

Abbreviations

CoMI:

Consensus mutual information

FC:

Fold change

NGS:

Next-generation sequencing

HPA:

Human protein atlas

TCGA:

The cancer genome atlas

LIHC:

Liver hepatocellular carcinoma

GBM:

Glioblastoma multiforme

BLCA:

Bladder urothelial carcinoma

BRCA:

Breast invasive carcinoma

COAD:

Colon adenocarcinoma

References

  1. Normanno N, De Luca A, Carotenuto P, Lamura L, Oliva I, Dā€™Alessio A. Prognostic applications of gene expression signatures in breast cancer. Oncology. 2009;77(Suppl 1):2ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  2. Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27(8):1160ā€“7.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  3. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747ā€“52.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  4. Ryaboshapkina M, Hammar M. Tissue-specific genes as an underutilized resource in drug discovery. Sci Rep. 2019;9(1):7233.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  5. Cui X, Churchill GA. Statistical tests for differential expression in cDNA microarray experiments. Genome Biol. 2003;4(4):210.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  6. Jain N, Thatte J, Braciale T, Ley K, Oā€™Connell M, Lee JK. Local-pooled-error test for identifying differentially expressed genes with a small number of replicated microarrays. Bioinformatics. 2003;19(15):1945ā€“51.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  7. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98(9):5116ā€“21.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  8. Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, Varambally S, Cao X, Tchinda J, Kuefer R, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310(5748):644ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  9. MacDonald JW, Ghosh D. COPA-cancer outlier profile analysis. Bioinformatics. 2006;22(23):2950ā€“1.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  10. Galitski T, Saldanha AJ, Styles CA, Lander ES, Fink GR. Ploidy regulation of gene expression. Science. 1999;285(5425):251ā€“4.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Tibshirani R, Hastie T, Narasimhan B, Chu G. Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci U S A. 2002;99(10):6567ā€“72.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  12. Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938ā€“45.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  13. Madu CO, Lu Y. Novel diagnostic biomarkers for prostate cancer. J Cancer. 2010;1:150ā€“77.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  14. Gaude E, Frezza C. Tissue-specific and convergent metabolic transformation of cancer correlates with metastatic potential and patient survival. Nat Commun. 2016;7:13041.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  15. Sonawane AR, Platig J, Fagny M, Chen CY, Paulson JN, Lopes-Ramos CM, DeMeo DL, Quackenbush J, Glass K, Kuijjer ML. Understanding tissue-specific gene regulation. Cell Rep. 2017;21(4):1077ā€“88.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  16. Shlomi T, Cabili MN, Herrgard MJ, Palsson BO, Ruppin E. Network-based prediction of human tissue-specific metabolism. Nat Biotechnol. 2008;26(9):1003ā€“10.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  17. Yang WY, Rao PS, Luo YC, Lin HK, Huang SH, Yang JM, Yuh CH. Omics-based investigation of diet-induced obesity synergized with HBx, Src, and p53 mutation accelerating hepatocarcinogenesis in Zebrafish model. Cancers (Basel). 2019;11(12):1899.

    ArticleĀ  Google ScholarĀ 

  18. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjostedt E, Asplund A, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347(6220):1260419.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  19. Clark PA, Iida M, Treisman DM, Kalluri H, Ezhilan S, Zorniak M, Wheeler DL, Kuo JS. Activation of multiple ERBB family receptors mediates glioblastoma cancer stem-like cell resistance to EGFR-targeted inhibition. Neoplasia. 2012;14(5):420ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  20. Carter Y, Sippel RS, Chen H. Hypothyroidism after a cancer diagnosis: etiology, diagnosis, complications, and management. Oncologist. 2014;19(1):34ā€“43.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  21. Chandran R, Hakki M, Spurgeon S. Infections in leukemia, Sepsis Luciano Azevedo. IntechOpen; 2012.

  22. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646ā€“74.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  23. Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C, Stuart JM. The cancer genome atlas pan-cancer analysis project. Nat Genet. 2013;45(10):1113ā€“1120.

  24. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  25. Li J, Tibshirani R. Finding consistent patterns: a nonparametric approach for identifying differential expression in RNA-Seq data. Stat Methods Med Res. 2013;22(5):519ā€“36.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

We are thankful for the computing resources supported by the Center for Bioinformatics Research (CBR) by Ministry of Education ATU program and National Chiao Tung University, and the Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B) of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan.

About this supplement

This article has been published as part of BMC Bioinformatics Volume 22 Supplement 10 2021: Selected articles from the 19th Asia Pacific Bioinformatics Conference (APBC 2021): bioinformatics.The full contents of the supplement are available at https://bmcbioinformatics.biomedcentral.com/articles/supplements/volume-22-supplement-10.

Funding

Publication costs are funded by the Ministry of Science and Technology (MOST 109-2634-F-009-021) in Taiwan, the Artificial Intelligence to Precision Health: Integrating Dynamic Physiological Signals and EMR to build a Medical Digital Twins Platform (MOST 109-2321-B-009-007), and the National Health Research Institutes (NHRI-EX109-10504PI). The funding bodies played no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

SHH analysed the omics data and wrote the manuscript; YSL designed the experiments; YCL and JYL provided contribution and discussion on biological meaning; YHC prepared the computing environments for R language and program code; JMY designed and supervised the study, provided the intellectual contribution, discussion and edited the manuscript. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Jinn-Moon Yang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent to publish

Not applicable.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Fig. S1

. The correlations of the ranking of all genes were compared between CoMI, T-test, FC, and SAM in five cancer types. Fig. S2. The odds ratios of tissue-specific genes selected by CoMI and SAM in five cancer types, including LIHC, GBM, BRCA, and COAD. Fig. S3. Boxplot for the results of the ranking of tissue-specific genes identified by CoMI and T-test in five cancer types. Fig. S4. Boxplot for the results of the ranking of tissue-specific genes identified by CoMI and SAM in five cancer types. Fig. S5. Comparisons between CoMI and COPA for tissue-specific properties in five cancer types. Fig. S6. Kaplanā€“Meier plots representing patients stratified by the auto-select best cutoff of top-ranked 10 genes identified by CoMI in GBM.Ā Fig. S7.Ā The distributions of CoMI values with different SMI and SDist values. Fig. S8.Ā The relationship between SMI, SDist, and tissue-specific genes.

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, visithttp://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://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

Huang, SH., Lo, YS., Luo, YC. et al. CoMI: consensus mutual information for tissue-specific gene signatures. BMC Bioinformatics 22 (Suppl 10), 624 (2021). https://doi.org/10.1186/s12859-022-04682-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12859-022-04682-2

Keywords