Next Article in Journal
Synthesis and Application of Silver Nanoparticles for Caries Management: A Review
Previous Article in Journal
Roles of Prostaglandins and Hydrogen Sulfide in an Outflow Model of the Porcine Ocular Anterior Segment Ex Vivo
Previous Article in Special Issue
Oncogenic Potential of Replication Factor C Subunit 4: Correlations with Tumor Progression and Assessment of Potential Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Therapeutic, Clinicopathological, and Molecular Correlates of PRKACA Expression in Gastrointestinal Cancers

1
Department of Medical Laboratories, College of Applied Medical Sciences, Majmaah University, Al-Majmaah 11952, Saudi Arabia
2
Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
3
Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(10), 1263; https://doi.org/10.3390/ph17101263
Submission received: 8 August 2024 / Revised: 30 August 2024 / Accepted: 5 September 2024 / Published: 25 September 2024
(This article belongs to the Special Issue Small Molecules in Targeted Cancer Therapy and Diagnosis)

Abstract

:
Background/Objectives: PRKACA alterations have clear diagnostic and biological roles in the fibrolamellar variant of hepatocellular carcinoma and a potential predictive role in that cancer type. However, the roles of PRKACA have not been comprehensively examined in gastric and colorectal cancers (GC and CRC). This study, therefore, sought to investigate the roles of PRKACA expression in GC and CRC. Methods: The clinico-genomic data of 441 GC and 629 CRC cases were analyzed for therapeutic, clinicopathological, and biological correlates using appropriate bioinformatics and statistical tools. Furthermore, the deregulation of PRKACA expression in GC and CRC was investigated using correlative and regression analyses. Results: The results showed that PRKACA expression subsets were enriched for gene targets of chemotherapeutics, tyrosine kinase, and β-adrenergic inhibitors. Moreover, high PRKACA expression was associated with adverse clinicopathological and genomic features of GC and CRC. Gene Ontology Enrichment Analysis also showed that PRKACA-high subsets of the GI cancers were enriched for the biological and molecular functions that are associated with cell motility, invasion, and metastasis but not cell proliferation. Finally, multiple regression analyses identified multiple methylation loci, transcription factors, miRNA species, and PRKACA copy number changes that deregulated PRKACA expression in GC and CRC. Conclusions: This study has identified potential predictive and clinicopathological roles for PRKACA expression in GI cancers and has added to the growing body of knowledge on the deregulation of PRKACA in cancer.

1. Introduction

Gastrointestinal (GI) cancers remain important public health concerns despite the huge number of resources that have been expended in research efforts to decipher the clinical, molecular, and biological characteristics of these cancers [1,2,3]. Gastric and colorectal cancers (GC and CRC), as a group, comprise the commonest malignancies worldwide ahead of the female breast, lung, and prostate cancers, accounting for over 3.0 million (15.33% of all malignancies) new cases [4,5] and constitute the 2nd commonest cause of cancer deaths with 1,703,966 (16.76% of all malignancies) new deaths [4,5]. An improved understanding of the tumor biology of GI cancers will inform innovative and enhanced strategies for cancer management [6,7,8]. Hence, new research that illuminates GI carcinogenesis and progression is warranted.
This study investigated the potential therapeutic significance of PRKACA expression in GC and CRC, as well as its clinicopathological and molecular correlates. PRKACA encodes the catalytic subunit-α of PKA, a kinase enzyme that functions downstream of the β-receptors in the β-adrenergic signaling pathway [9,10,11,12]. PKA phosphorylates multiple downstream targets in the β-adrenergic signaling pathway, including transcription factors ATF, CREB, GATA1, STAT3, Src, and BARK [9,11,12]. However, the roles of PRKACA in cancer are incompletely understood, inasmuch as the cyclic adenosine monophosphate (cAMP)-PKA signaling has been shown to possess both tumor-suppressive and oncogenic roles in different tumor types and contexts [12]. A preclinical study demonstrated that crotonylation of PRKACA enhances the activity of PKA and thereby promotes colorectal cancer development via the PKA-FAK-AKT pathway, whereas PRKACA is secreted by prostate cancer cells [11]. In clinical cancer, PRKACA participates in a fusion event with DNAJB1 in up to a hundred percent of fibrolamellar hepatocellular carcinoma and has therefore been identified as a suitable diagnostic marker for and a driver of oncogenic activities in that cancer [13,14,15]. In addition, PRKACA was found to be upregulated in the serum and tumor of gastric and colorectal cancer patients but did not show any clinical correlates in a study that was markedly limited by sample sizes [16]. The therapeutic significance of PRKACA expression in cancer had previously been suggested by a breast cancer study, which showed that PRKACA mediates resistance to anti-HER2 therapy in breast cancer cell lines via inactivation of BAD and Bcl2-associated death promoter [17]. Indeed, elevated PRKACA expression was found in trastuzumab-resistant breast cancer patients, evidence that PRKACA is activated in trastuzumab-resistant breast cancer cases [16]. With respect to therapy, PRKACA could be a target for the treatment of cancer. For example, it has been suggested that the PAK signaling and PRKACA activities could be inhibited upstream using β2 receptor blockade with propranolol as a strategy for the treatment of invasive cancer [9]. In addition, Schalm et al. and Bauer et al. [14,18] demonstrated that PRKACA—in the DNAIJ-PRKACA fusion context—is a viable target for the treatment of the PRKACA-dependent fibrolamellar variant of hepatocellular carcinoma. However, the biological and therapeutic significances of PRKACA expression in clinical GC and CRC have not been thoroughly interrogated. Therefore, the aims of this study include (i) to interrogate the potential therapeutic significance of PRKACA in clinical GC and CRC, (ii) to investigate the clinicopathological and genomic correlates of PRKACA expression in GC and CRC, (iii) to investigate the biological significance of PRKACA expression in GC and CRC, (iv) to investigate the mechanisms of PRKACA deregulation in GI cancers.

2. Results

2.1. Enrichment of Gene Targets of Multiple Drug Agents in PRKACA-Low and -High Cancers

2.1.1. Gastric Cancer Cohort

Normalized PRKACA expression values were dichotomized using the mean PRKACA values for each cohort. There were 206 PRKACA-low (50.0%) and 206 PRKACA-high (50.0%) GC cases. GSEA followed by drug set annotations on Enrichr (https://maayanlab.cloud/Enrichr/enrich, accessed on 25 July 2024) revealed enrichment of genes that are targets of multiple drugs, including tyrosine kinase inhibitors, β-blockers, and conventional chemotherapeutic agents, among other drug classes, in both the high and low PRKACA subsets, and an adjusted p value of ≤0.05 and false discovery rate (FDR) of ≤0.05 (Figure 1). Gene targets enriched in the PRKACA-high subset include those for conventional chemotherapeutic agents such as paclitaxel, irinotecan, camptothecin, ifosfamide, and methotrexate; tyrosine kinase inhibitors sorafenib, dasatinib, axitinib, gefitinib; β-blockers such as atenolol, and metoprolol; and calcium channel blockers, such as dexverapamil, verapamil and diltiazem (Supplementary Materials Drug-targets_PRKACA_high_GC). PRKACA-low GC subset, on the other hand, shows significant enrichment of targets that are downregulated following treatment with chemotherapeutic agents such as irinotecan, camptothecin, and daunorubicin (Figure 1). Furthermore, the PRKACA-low subset was enriched for gene targets associated with tyrosine kinase inhibitors such as momelotinib, ponatinib, sunitinib, dasatinib, sorafenib, vandetanib, among many others (Supplementary Materials Drug-targets_PRKACA_low_GC). However, it was observed that one phenotype (PRKACA-high, for example) might be associated with both upregulation and downregulation of the gene set of the same drug, depending on the library that was interrogated.

2.1.2. Colorectal Cancer Cohort

Normalized PRKACA expressions were dichotomized into PRKACA-low (N = 268, 50%) and PRKACA-high subsets (N = 268, 50%). Significant enrichment of gene targets of multiple drugs was observed in both PRKACA subsets in the CRC cohort. Similar to the GC cohort, PRKACA expression in the CRC cohort exhibited enrichment of genes that are associated with β-blockers, conventional chemotherapeutics, and tyrosine kinase inhibitors (Figure 1). Furthermore, as was observed in the GC cohort, the pattern of upregulation or downregulation of drug targets differs with the gene sets or libraries interrogated. For example, in the PRKACA-high subset, irinotecan gene sets showed both downregulated or upregulated targets depending on the library that was interrogated. This finding may have resulted from the differences in the biology of the cancer cell lines (e.g., breast cancer (MCF7) vs. leukemia (HL60) cell lines) that were utilized to generate the gene set libraries (Supplementary Material Drug-targets_PRKACA_high_CRC and Supplementary Material Drug-targets_PRKACA_low_CRC).

2.2. Clinicopathological and Molecular Features of PRKACA Expression

2.2.1. Gastric Cancer Cohort

The clinicopathological and molecular correlates of PRKACA expression were interrogated using the Chi-square test. The results showed that in the GC cohort, high PRKACA expression was significantly associated with the 60-years-and-below age group, higher nodal stage, and late disease stage but not with gender, histological tumor type, pathological tumor stage, or distant metastasis (Table 1). PRKACA expression did not show any association with a prognosis on Kaplan-Meier analysis (Log Rank X2 = 1.832, p = 0.176). Furthermore, high PRKACA expression was associated with the genome-stable and Epstein-Barr Virus subtypes of GC and with tumors having low aneuploidy and high global methylation scores (Table 2).

2.2.2. Colorectal Cancer Cohort

In the CRC cohort, high PRKACA expression was associated with younger age groups (particularly the 60-years-and-below age group), left-sided colon cancer, rectal cancer, and nodal stage, but not with gender, race/ethnicity, pathological tumor stage, metastasis, TNM stage, lymphovascular and vascular invasion, and histological type (Table 1). As with the GC cohort, PRKACA expression did not show any association with overall or progression-free survival (OS: Log Rank X2 = 0.039, p = 0.844; DFS: Log Rank X2 = 0.054, p = 0.816). Moreover, high PRKACA expression was associated with the chromosomal instability and microsatellite stable (MSS) subtypes of CRC and with tumors having low mutation count, low MSI Sensor score, and high Fraction Genome Altered and Aneuploidy scores, but not with MSI MANTIS score and tumor mutational burden (TMB) (Table 2).

2.3. Biological Significance of PRKACA Expression

2.3.1. Gastric Cancer Cohort

GSEA showed significant differential enrichment of HALLMARK_MYOGENESIS, HALLMARK_APICAL_JUNCTION, HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION, and HALLMARK_COAGULATION gene sets in the PRKACA-high subset of GC cohort (Supplementary Material GC_GSEA.PRKACA_high). GO Enrichment Analysis showed that the enriched genes identified have a biological function in Cell-Matrix Adhesion (GO:0007160), Regulation of Cell Migration (GO:0030334), Actomyosin Structure Organization (GO:0031032), Contractile Actin Filament Bundle Assembly (GO:0030038), Regulation of Cell Motility (GO:2000145), etc., functions which are involved in cell motility, migration and metastasis (Figure 2 and Supplementary Material GOEA_GC_PRKACA_high). On the other hand, the PRKACA-low GC subset showed differential enrichment of the HALLMARK_G2M_CHECKPOINT, HALLMARK_MTORC1_SIGNALING, HALLMARK_PROTEIN_SECRETION and HALLMARK_E2F_TARGETS gene sets (Supplementary Material GC_GSEA.PRKACA_low). GO Enrichment Analysis identified the core enrichment genes in these gene sets to have such biological functions as Mitotic Cell Cycle Phase Transition (GO:0044772), Mitotic Spindle Organization (GO:0007052), DNA-templated DNA Replication (GO:0006261), etc., all of which are associated with cell replication, proliferation and tumor growth (Figure 2 and Supplementary Material GOEA_GC_PRKACA_low). The results of the analyses suggest that PRKACA may be a pro-migration/invasion and anti-proliferation gene in gastric carcinogenesis and gastric cancer progression.

2.3.2. Colorectal Cancer Cohort

GSEA showed differential enrichment of the HALLMARK_MYOGENESIS, HALLMARK_APICAL_JUNCTION, HALLMARK_KRAS_SIGNALING_DN, HALLMARK_COAGULATION, HALLMARK_ANGIOGENESIS, HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION, HALLMARK_APICAL_SURFACE, HALLMARK_HEDGEHOG_SIGNALING, HALLMARK_NOTCH_SIGNALING, and HALLMARK_IL6_JAK_STAT3_SIGNALING gene sets in the PRKACA-positive CRC cohort (Supplementary Material CRC_GSEA.PRKACA_high). GO Enrichment Analysis showed that the enriched genes identified in the GSEA have biological functions, which included Cell-Matrix Adhesion (GO:0007160), Actomyosin Structure Organization (GO:0031032), Positive Regulation of Cell Migration (GO:0030335), Positive Regulation of Cell Motility (GO:2000147), etc. These biological functions are associated with cell motility, invasion, and metastasis, as noted above in the GC cohort (Figure 3 and Supplementary Material GOEA_CRC_PRKACA_high).
The PRKACA-low CRC subset did not show differential enrichment of any of the HALLMARK gene sets associated with cell proliferation.

2.4. Differential Core Enrichment Gene Sets between GC and CRC PRKACA-High Subsets

A total of 281 genes comprising core enrichment components from the five Hallmark gene sets were found for the GC PRKACA-high subset, while 503 genes were enriched in the CRC PRKACA-high subset (Supplementary Material Core.Enrichment_GeneSets). A comparison of the core enrichment gene sets in the GC and CRC PRKACA-high subsets showed that only two enriched genes, EGFR and LAMP2, overlapped between the GC and CRC subsets, whereas the GO enrichment analysis found that the enriched biological processes, molecular functions, and cellular components subserved by the enriched genes were similar between the GI cancers. This is evidence that PRKACA or the PKA signaling may regulate diverse genes/genetic pathways in different cancer types to accomplish the same biological processes and molecular functions.

2.5. Deregulation of PRKACA Expression in Gastrointestinal Cancers

2.5.1. Gastric Cancer Cohort

PRKACA deregulation was investigated using copy number, methylation, miRNA, and transcription factor expression. PRKACA copy number alterations were obtained from the copy number segment data of the TCGA GC cohorts using the segment mean threshold of 0.3 (for gain/amplification) and −0.3 (for deletion). Using this threshold, the study identified 31/412, 375/412, and 6/412 PRKACA deletions, neutral, and gain/amplification, respectively. There was a significant association between PRKACA copy number changes and expression (X2 = 23.609, p < 0.001; Figure 4). A total of 20 PRKACA promoter methylation loci and their beta values were retrieved from the gastric cancer methylation data. Bivariate correlation analysis demonstrated that 4/20 methylation loci were correlated with PRKACA expression (see Supplementary Material GC_PRKACA_Methylation_loci). Gene enrichment analysis with DEseq2 identified the top 40 miRNA species that were differentially enriched in the PRKACA-high and -low cases in the GC cohort. Bivariate analysis showed that the expression of 20/40 miRNA species showed correlations with PRKACA expression (see Supplementary Material GC_PRKACA-targeting miRNA). A total of 56 PRKACA-targeting transcription factors were retrieved from the transcription factor database TF2DNA_DB. Bivariate analysis showed that the expression of 27/56 transcription factors was correlated with PRKACA expression in the GC cohort (see Supplementary Material GC_PRKACA_TranscriptionFactors).
PRKACA copy number changes, methylation loci beta values, PRKACA-targeting transcription factors, and the differentially enriched miRNA values were incorporated into a multiple regression analysis. The results showed that multiple regulatory mechanisms independently predicted PRKACA expression in the GC cohort (Table 3, Figure 5).

2.5.2. Colorectal Cancer Cohort

In the CRC cohort, there were 10/480, 455/480, and 15/480 PRKACA copy number deletions, neutral, and gain/amplification, respectively. Furthermore, there were significant associations between PRKACA copy number changes and expression (X2 = 5.039, p = 0.007; Figure 4). The bivariate analysis identified 11/20 methylation loci whose beta values exhibited a significant correlation with PRKACA expression in the CRC cohort (see Supplementary Material CRC_PRKACA_Methylation_loci). Only one methylation locus, cg17818798, exhibited a shared PRKACA methylation-expression correlation between the CRC and GC cohorts. The expression of 25/40 miRNA species identified in the miRNA gene enrichment analysis of the CRC cohort showed correlations with PRKACA expression on bivariate analysis (see Supplementary Material CRC_PRKACA-targeting miRNA). The expression of seven (7) miRNA species, including hsa-let-7c, hsa-mir-1-1, hsa-mir-1-2, hsa-mir-133b, hsa-mir-143, hsa-mir-7641-1, and hsa-mir-99a were found to be commonly correlated with PRKACA expression in the GC and CRC cohorts. Of the 56 PRKACA-targeting transcription factors retrieved from the TF2DNA_DB, the expression levels of 26 showed a significant correlation with PRKACA expression (see Supplementary Material CRC_PRKACA_TranscriptionFactors). The expression levels of 16/56 PRKACA-targeting transcription factors retrieved from the TF2DNA_DB were found to be commonly correlated with PRKACA expression in both the GC and the CRC cohorts. These included HOXA1, ZNF451, TFEB, TSHZ3, ZNF644, ZNF557, DHX34, HEY2, ZNF235, KLF15, HAND2, E2F7, TGIF1, ZBTB7A, MAFK, and CHAMP1.
Multiple linear regression analysis showed that a combination of regulatory mechanisms, including PRKACA copy number alterations, transcription factor expression levels, promoter methylation status, and miRNA expression levels, independently predicted PRKACA expression in the CRC cohort (Table 4; Figure 5). The methylation locus cg17818798 and ZNF451 and ZNF557 expression were the predictors of PRKACA expression common to both the GC and CRC cohorts.

3. Discussion

PRKACA, the α-catalytic subunit of PKA, is an essential component of PKA signaling that catalyzes the downstream targets of PKA [9,12]. This study demonstrated that PRKACA expression in GC and CRC displays enrichment of genes that are associated with multiple conventional chemotherapeutic agents, FDA-approved tyrosine kinase inhibitors, and β-adrenergic blockers. The study findings support the role of PRKACA as a potential biomarker for multiple tyrosine kinase inhibitors, chemotherapeutic drugs, and β-adrenergic blockers. The findings also lend credence to the Cole and Sood study [9], which suggested that the inhibition of β-adrenergic signaling could possibly be utilized for the treatment of invasive cancer. This is even more so with the demonstration that high PRKACA expression was associated with the tumor hallmark of invasion or migration, rather than proliferation, in the GC and CRC cohorts. Moreover, the enrichment of gene targets of multiple drugs in PRKACA-high cases highlights the concept of an integrated, multitargeted approach to cancer therapy [19], wherein a single biomarker could at once be predictive of response to multiple drugs with varying mechanisms of action, on the basis of the association of that biomarker with a global gene expression network that incorporates the gene targets for those drugs [20]. The advantages of such an integrated approach to cancer therapy would include the augmentation of the therapeutic options for the oncologist and the reduction of the risks of drug toxicity and resistance without the violation of the tenets of personalized medicine [21,22]. To reiterate, the pro-migration/invasion characteristics of PRKACA, as found in this study, support a proposal for targeting PRKACA directly [14,18] and/or the β-adrenergic pathway [9] for treating tumor invasion.
The tumor-promoting activities of the PRKACA and PKA signaling have been described for classic and fibrolamellar variants of hepatocellular carcinoma, and breast, prostate, ovarian, and non-small cell lung cancers, glioblastoma, astrocytoma, and leukemia [11,12,13,14,15]. The association of PRKACA expression with some adverse clinicopathological features of gastrointestinal cancers, such as advanced age, tumor site, tumor histotype, nodal stage, and TNM stage, are in keeping with the tumor-promoting activities in the above-mentioned studies. Furthermore, this study showed that PRKACA expression is associated with poor prognosis molecular features and subtypes of gastrointestinal cancers. To the best of our knowledge, these findings have not been previously elucidated in any gastrointestinal cancer cohorts.
Interestingly, GSEA and Gene Ontology Enrichment Analysis demonstrated that while PRKACA-high tumors exhibited differential enrichment of genes that regulate cell motility, migration, and metastasis in the GC and CRC cohorts, there was enrichment of genes that regulate cell proliferation and growth in the GC, but not the CRC, PRKACA-low subset. The differential enrichment of actin filament organization, actin polymerization, actomyosin structure, focal adhesion structure, and membrane organelle organization are in keeping with the study by McKenzie et al. [23], which found that the PKA signaling is locally and rapidly activated by mechanical stretch in an actomyosin contractility-dependent manner, thus establishing the activated PKA as an effector of cellular mechanotransduction and a regulator of mechanically guided cell migration. Our findings also concur with the study by Tonucci et al. [24], which demonstrated that phosphorylation of CIP4 by PKA promotes the formation of functional invadopodia and, thus, confirms PKA phosphorylation of CIP4 as a regulator of the metastatic phenotype in cancer cells. Furthermore, the gene enrichment analysis results are in agreement with the Cheng et al. study [25], which demonstrated that PKA targets focal adhesion kinase for the promotion of cancer cell metastasis by cAMP. Moreover, Duan et al. [26] demonstrated that the phosphorylation of TPI at serine 58 by PRKACA enhances its enzymatic activity and glycolysis and, thus, its promotion of cancer growth and metastasis. Overall, our findings suggest that the primary function of PRKACA in cancer may be pro-migration/invasion rather than cell proliferation in GI cancers and may explain the seemingly paradoxical roles that have been described for PRKACA in different cancers [12]. For example, whilst PKA signaling has been demonstrated to regulate actomyosin contractility and cell migration, as well as membrane deformation, actin polymerization, cancer cell invasion, and metastasis, through phosphorylation of CIP4 and FAK [23,24,25,26], activating PRKACA mutations have been associated with smaller tumor sizes of adrenocortical adenomas [27,28,29,30]. PRKACA mutations in these adenomas have been demonstrated to have a limited impact on cell proliferation and tumor growth [30].
The concept of a proliferation-invasion dichotomy in cancer cells is not new in oncology [31,32,33,34]. In many tumor types, individual cancer cells exist either as proliferating or invading cells within the tumor body, using various molecular mechanisms to switch between proliferative and invasive phenotypes [31,32,33,34]. Conversely, genes and genetic pathways that possess pro-metastasis, but not proliferation or even anti-proliferation, functions in tumorigenesis and tumor progression are not uncommonly activated in cancer. The TGFB pathway is a ready example of a pathway that has seemingly paradoxical roles in cancer. The TGFB pathway, an anti-cell proliferation pathway, is known to induce the epithelial-mesenchymal transition and cell migration in cancer cells [35]. Furthermore, the cell cycle regulator p16 inhibits tumor cell proliferation through impedance of the G1-S phase progression [36,37]. However, p16 has been demonstrated to be upregulated or overexpressed in the frontiers of invading tumor cell nests and in tumor buds, evidence that it is essential for tumor cell invasion and metastasis [38].
The most common deregulatory mechanisms of PRKACA expression include structural variation or gene fusion [13,14,15], somatic mutations [10,27,28,29,30], and copy number alterations [27,39]. Whilst no structural variation or somatic mutations of PRKACA were sought in this study, PRKACA copy number alterations were demonstrably a significant mechanism of PRKACA expression regulation in our gastrointestinal cancer cohorts. However, the predominant regulatory mechanisms that better predicted PRKACA expression in our gastric and colorectal cancer cohorts are epigenetic, transcriptional, and microRNA regulatory mechanisms. These aforementioned mechanisms are the most common gene regulatory mechanisms in cancers generally [40].
In conclusion, this study has demonstrated that a subset of GC and CRC exhibit elevated expression of PRKACA, which is associated with the overrepresentation of gene targets of multiple FDA-approved tyrosine kinase inhibitors and β-adrenergic signaling inhibitors. PRKACA-high tumors were also associated with adverse clinicopathological and molecular characteristics of GI cancers. Furthermore, the study showed that tumors with high PRKACA expression were enriched for biological and molecular functions associated with cell motility, cell invasion, and metastasis. The deregulatory mechanisms of epigenetics, miRNA, and copy number alteration mechanisms were found to predict PRKACA expression in GC and CRC.

4. Materials and Methods

4.1. Study Cohorts

This study retrospectively analyzed the clinicopathological and genomic data of the cancer genome atlas (TCGA) colon, rectal, and gastric cancer cohorts [41,42,43,44]. All the data were retrieved from the Genome Data Commons (GDC) and cBioPortal for Cancer Genomics databases. Transcript quantification was accomplished with RNASeq (mRNA) and miRNASeq (miRNA). Methylation beta values were generated using a methylation array on the Illumina Human Methylation 450 platform, while the masked copy number segment data were generated using the Affymetrix SNP 6.0 genotyping array.

4.2. Data Processing

The clinico-genomic data of the GI cancer cohorts were retrieved from the GDC and CBioPortal databases using Linux-based scripts and codes that were written in the Windows-based Ubuntu 20.04 environment. Linux-based codes and scripts were also utilized to prepare the gene expression datasets in accordance with the Molecular Signature Database (MSigDB) [45,46] and DESeq2 Gene Enrichment Analyses requirements (https://cloud.genepattern.org/, accessed on 25 July 2024) [47], whereas Excel spreadsheet was utilized in the generation of the phenotype and derivative gene set files (see below), following which these were converted to cls and grp files, respectively. The gastric cancer cohort comprised 441 primary cases with clinicopathological (including prognostic data), RNASeq, chromosomal copy number segment, methylation, and somatic mutation data. The following data were available for this cohort: clinicopathological (between 380 and 441 of 441 cases for each clinicopathological indices; Table 1); mRNA expression (415/441 cases); chromosomal copy number segment (441/441 cases); methylation (between 393 and 440 of 441 cases for individual methylation loci); microRNA expression (441/441 cases) data.
The colorectal cancer cohort included 629 primary cases with the following amount of data: clinicopathological (between 545 and 629 of 629 cases for each clinicopathological indices; Table 1); mRNA expression (534/629 cases); chromosomal copy number segment (512/629 cases); methylation (between 331 and 524 of 629 cases for individual methylation loci); microRNA expression (506/629 cases) data.
The PRKACA expression data from these cohorts were converted into normally distributed data by using the method described by Templeton [48] before their utilization for statistical analyses.

4.3. Study Approach

First, GSEA and drug-target annotation with Drug Signature Database (DSigDB) gene set libraries in the Enrichr environment (https://maayanlab.cloud/Enrichr/enrich, accessed on 25 July 2024) [45,46,49,50,51] were utilized to infer the potential for PRKACA expression to predict susceptibility or resistance to drug agents. Then, the relationship between the clinicopathological indices and PRKACA expression in each of the GI cancers was sought in the cohorts using the appropriate statistical tests. In addition, the biological significance of PRKACA expression in GI carcinogenesis was confirmed using GSEA with the GC and CRC gene expression datasets and the MSigDB Hallmark gene sets [45,46]. The list of genes in the core enrichment of the significant gene sets was applied to gene ontology analysis in the Enrichr environment [45,46,49,50] to verify the biological, molecular, and functional significance of the enriched genes. Moreover, the mechanisms of altered PRKACA expression were sought in each of the GI cancer cohorts using the copy number segment, structural variation, methylation (beta values), transcription factor, and miRNA expression data. PRKACA-targeting transcription factors were retrieved from the transcription factor database TF2DNA_DB (https://www.fiserlab.org/tf2dna_db/search_genes.html, accessed on 25 July 2024) [52], and their expression values were correlated with PRKACA expression in either cancer cohorts using bivariate analyses. Significantly correlated transcription factors were then selected for regression analyses. Differential miRNA enrichment was sought between PRKACA-low and PRKACA-high cases using the online DESeq2 software v.3 on the GenePattern computing environment [47]. Gene enrichment analyses by DESeq2 were used to identify the top 40 miRNA species that differentially expressed in the PRKACA-low and -high subsets in the GC and CRC cohorts. Significantly enriched miRNAs, which also showed correlations with PRKACA expression by bivariate analyses, were then incorporated into a regression analysis, together with the methylation, PRKACA-targeting transcription factors, structural variation, and PRKACA copy number indices, to infer their roles in the deregulation of PRKACA expression.

4.4. Statistical Analyses

The enrichment analysis in the GSEA and DESeq2 software was performed using the software’s default parameters. GSEA was performed as a phenotype permutation. Gene ontology enrichment and drug-target association annotations were performed in the Enrichr environment using thresholds nominal p-value ≤ 0.05 and FDR ≤ 0.05. The clinicopathological and genomic data of the cancer cohorts were input into SPSS version 29. The Chi-square (or Fisher) test was used to probe for significant associations between categorical variables, while bivariate correlative analysis was utilized to test the correlations between continuous variables. Multivariate analysis of PRKACA expression correlation was investigated with multiple linear regression and binary logistic regression analyses. The one-way ANOVA test was used to measure the mean differences of continuous variables between discrete groups. The prognostic significance of PRKACA expression was defined using Kaplan–Meier and Cox regression analyses. A p-value of <0.05 was used as the threshold for significant tests, while the Benjamini–Hochberg correction was used to correct for multiple testing at an FDR of <0.05.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph17101263/s1, Supplementary Material Drug-targets_PRKACA_high_GC, Supplementary Material Drug-targets_PRKACA_low_GC, Supplementary Material Drug-targets_PRKACA_high_CRC, Supplementary Material Drug-targets_PRKACA_low_CRC, Supplementary Material GC_GSEA.PRKACA_high, Supplementary Material GOEA_GC_PRKACA_high, Supplementary Material GC_GSEA.PRKACA_low, Supplementary Material GOEA_GC_PRKACA_low, Supplementary Material CRC_GSEA.PRKACA_high, Supplementary Material GOEA_CRC_PRKACA_high, Supplementary Material Core.Enrichment_GeneSets, Supplementary Material GC_PRKACA_Methylation_loci, Supplementary Material GC_PRKACA-targeting miRNA, Supplementary Material GC_PRKACA_TranscriptionFactors, Supplementary Material CRC_PRKACA_Methylation_loci, Supplementary Material CRC_PRKACA-targeting miRNA, Supplementary Material CRC_PRKACA_TranscriptionFactors.

Author Contributions

Conceptualization, A.A. and A.A.O.; methodology, G.A.; software, A.A.; validation, A.A.O. and G.A.; formal analysis, A.A. and G.A.; investigation, A.A.O.; resources, A.A.; data curation, G.A.; writing—original draft preparation, A.A.; writing—review and editing, G.A. and A.A.O.; visualization, A.A.O. and G.A.; supervision, A.A.; project administration, A.A.; funding acquisition, A.A.O., A.A., and G.A. All authors have read and agreed to the published version of the manuscript.

Funding

This study is supported via funding from Prince Sattam bin Abdulaziz University project number (PSAU/2023/R/1445).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All the genomic and clinicopathological data utilized for this study are freely available in the cBioPortal for Cancer Genomics website (https://www.cbioportal.org/, accessed on 25 June 2024), and the Genome Data Commons repository (https://portal.gdc.cancer.gov/analysis_page, accessed on 25 June 2024).

Acknowledgments

The authors wish to express their gratitude to the Cancer Genome Atlas and the cBioPortal for Cancer Genomics for making the data used in this study publicly available.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Jardim, S.R.; de Souza, L.M.P.; de Souza, H.S.P. The Rise of Gastrointestinal Cancers as a Global Phenomenon: Unhealthy Behavior or Progress? Int. J. Environ. Res. Public Health 2023, 20, 3640. [Google Scholar] [CrossRef] [PubMed]
  2. Li, M.M.; Yuan, J.; Guan, X.Y.; Ma, N.F.; Liu, M. Molecular subclassification of gastrointestinal cancers based on cancer stem cell traits. Exp. Hematol. Oncol. 2021, 10, 53. [Google Scholar] [CrossRef]
  3. Minciuna, C.-E.; Tanase, M.; Manuc, T.E.; Tudor, S.; Herlea, V.; Dragomir, M.P.; Calin, G.A.; Vasilescu, C. The seen and the unseen: Molecular classification and image based-analysis of gastrointestinal cancers. CSBJ 2022, 20, 5065–5075. [Google Scholar] [CrossRef] [PubMed]
  4. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
  5. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  6. Piña-Sánchez, P.; Chávez-González, A.; Ruiz-Tachiquín, M.; Vadillo, E.; Monroy-García, A.; Montesinos, J.J.; Grajales, R.; Gutiérrez de la Barrera, M.; Mayani, H. Cancer Biology, Epidemiology, and Treatment in the 21st Century: Current Status and Future Challenges From a Biomedical Perspective. Cancer Control 2021, 28, 10732748211038735. [Google Scholar] [CrossRef]
  7. Chehelgerdi, M.; Chehelgerdi, M.; Allela, O.Q.B.; Pecho, R.D.C.; Jayasankar, N.; Rao, D.P.; Thamaraikani, T.; Vasanthan, M.; Viktor, P.; Lakshmaiya, N.; et al. Progressing nanotechnology to improve targeted cancer treatment: Overcoming hurdles in its clinical implementation. Mol. Cancer 2023, 22, 169. [Google Scholar] [CrossRef]
  8. Shimozaki, K.; Nakayama, I.; Hirota, T.; Yamaguchi, K. Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era. Cells 2023, 12, 1049. [Google Scholar] [CrossRef] [PubMed]
  9. Cole, S.W.; Sood, A.K. Molecular Pathways: Beta-Adrenergic Signaling in Cancer. Clin. Cancer Res. 2012, 18, 1201–1206. [Google Scholar] [CrossRef]
  10. Stratakis, C.A. Cyclic AMP-dependent protein kinase catalytic subunit A (PRKACA): The expected, the unexpected, and what might be next. J. Pathol. 2018, 244, 257–259. [Google Scholar] [CrossRef]
  11. Hou, J.Y.; Gao, L.J.; Shen, J.; Zhou, L.; Shi, J.Y.; Sun, T.; Hao, S.L.; Wang, D.P.; Cao, J.M. Crotonylation of PRKACA enhances PKA activity and promotes colorectal cancer development via the PKA-FAK-AKT pathway. Genes Dis. 2022, 10, 332–335. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, H.; Kong, Q.; Wang, J.; Jiang, Y.; Hua, H. Complex roles of cAMP–PKA–CREB signaling in cancer. Exp. Hematol. Oncol. 2020, 9, 32. [Google Scholar] [CrossRef] [PubMed]
  13. Neumayer, C.; Ng, D.; Jiang, C.S.; Qureshi, A.; Lalazar, G.; Vaughan, R.; Simon, S.M. Oncogenic Addiction of Fibrolamellar Hepatocellular Carcinoma to the Fusion Kinase DNAJB1-PRKACA. Clin. Cancer Res. 2023, 29, 271–278. [Google Scholar] [CrossRef] [PubMed]
  14. Bauer, J.; Köhler, N.; Maringer, Y.; Bucher, P.; Bilich, T.; Zwick, M.; Dicks, S.; Nelde, A.; Dubbelaar, M.; Scheid, J.; et al. The oncogenic fusion protein DNAJB1-PRKACA can be specifically targeted by peptide-based immunotherapy in fibrolamellar hepatocellular carcinoma. Nat. Commun. 2022, 13, 6401. [Google Scholar] [CrossRef] [PubMed]
  15. Kastenhuber, E.R.; Lalazar, G.; Houlihan, S.L.; Tschaharganeh, D.F.; Baslan, T.; Chen, C.C.; Requena, D.; Tian, S.; Bosbach, B.; Wilkinson, J.E.; et al. DNAJB1-PRKACA fusion kinase interacts with β-catenin and the liver regenerative response to drive fibrolamellar hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2017, 114, 13076–13084. [Google Scholar] [CrossRef] [PubMed]
  16. Wu, Y.; Li, Y.; Liu, J.; Chen, M.; Li, W.; Chen, Y.; Xu, M. The diagnostic value of extracellular protein kinase A (ECPKA) in serum for gastric and colorectal cancer. Transl. Cancer Res. 2020, 9, 3870–3878. [Google Scholar] [CrossRef]
  17. Moody, S.E.; Schinzel, A.C.; Singh, S.; Chen, M.; Li, W.; Chen, Y.; Xu, M. PRKACA mediates resistance to HER2-targeted therapy in breast cancer cells and restores anti-apoptotic signaling. Oncogene 2015, 34, 2061–2071. [Google Scholar] [CrossRef]
  18. Schalm, S.S.; O’Hearn, E.; Wilson, K.; LaBranche, T.P.; Silva, G.; Zhang, Z.; DiPietro, L.; Bifulco, N.; Woessner, R.; Stransky, N.; et al. Evaluation of Protein Kinase cAMP-Activated Catalytic Subunit Alpha as a Therapeutic Target for Fibrolamellar Carcinoma. Gastro Hep Adv. 2023, 2, 307–321. [Google Scholar] [CrossRef]
  19. Tang, J.; Aittokallio, T. Network pharmacology strategies toward multi-target anticancer therapies: From computational models to experimental design principles. Curr. Pharm. Des. 2014, 20, 23–36. [Google Scholar] [CrossRef]
  20. Ferlier, T.; Coulouarn, C. Regulation of Gene Expression in Cancer—An Overview. Cells 2022, 11, 4058. [Google Scholar] [CrossRef]
  21. Bayat-Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef] [PubMed]
  22. He, L.; Kulesskiy, E.; Saarela, J.; Turunen, L.; Wennerberg, K.; Aittokallio, T.; Tang, J. Methods for High-throughput Drug Combination Screening and Synergy Scoring. Methods Mol. Biol. 2018, 1711, 351–398. [Google Scholar] [CrossRef]
  23. McKenzie, A.J.; Svec, K.V.; Williams, T.F.; Howe, A.K. Protein kinase A activity is regulated by actomyosin contractility during cell migration and is required for durotaxis. Mol. Biol. Cell 2020, 31, 45–58. [Google Scholar] [CrossRef]
  24. Tonucci, F.M.; Almada, E.; Borini-Etichetti, C.; Pariani, A.; Hidalgo, F.; Rico, M.J.; Girardini, J.; Favre, C.; Goldenring, J.R. Identification of a CIP4 PKA phosphorylation site involved in the regulation of cancer cell invasiveness and metastasis. Cancer Lett. 2019, 461, 65–77. [Google Scholar] [CrossRef]
  25. Cheng, Y.; Gao, X.H.; Li, X.J.; Cao, Q.H.; Zhao, D.D.; Zhou, J.R.; Wu, H.X.; Wang, Y.; You, L.J. Depression promotes prostate cancer invasion and metastasis via a sympathetic-cAMP–FAK signaling pathway. Oncogene 2018, 37, 2953–2966. [Google Scholar] [CrossRef] [PubMed]
  26. Duan, Y.; Li, J.; Wang, F.; Wei, J.; Yang, Z.; Sun, M.; Liu, J.; Wen, M.; Huang, W.; Chen, Z.; et al. Protein modifications throughout the lung cancer proteome unravel the cancer-specific regulation of glycolysis. Cell Rep. 2021, 37, 110137. [Google Scholar] [CrossRef] [PubMed]
  27. Berthon, A.S.; Szarek, E.; Stratakis, C.A. PRKACA: The catalytic subunit of protein kinase A and adrenocortical tumors. Front. Cell Dev. Biol. 2015, 3, 26. [Google Scholar] [CrossRef] [PubMed]
  28. Di Dalmazi, G.; Kisker, C.; Calebiro, D.; Mannelli, M.; Canu, L.; Arnaldi, G.; Quinkler, M.; Rayes, N.; Tabarin, A.; Jullié, M.L.; et al. Novel somatic mutations in the catalytic subunit of the protein kinase A as a cause of adrenal Cushing’s syndrome: A European multicentric study. J. Clin. Endocrinol. Metab. 2014, 99, E2093–E2100. [Google Scholar] [CrossRef]
  29. Goh, G.; Scholl, U.I.; Healy, J.M.; Choi, M.; Prasad, M.L.; Nelson-Williams, C.; Kuntsman, J.W.; Korah, R.; Suttorp, A.C.; Dietrich, D.; et al. Recurrent activating mutation in PRKACA in cortisol-producing adrenal tumors. Nat. Genet. 2014, 46, 613–617. [Google Scholar] [CrossRef]
  30. Sato, Y.; Maekawa, S.; Ishii, R.; Sanada, M.; Morikawa, T.; Shiraishi, Y.; Yoshida, K.; Nagata, Y.; Sato-Otsubo, A.; Yoshizato, T.; et al. Recurrent somatic mutations underlie corticotropin-independent Cushing’s syndrome. Science 2014, 344, 917–920. [Google Scholar] [CrossRef]
  31. Hatzikirou, H.; Basanta, D.; Simon, M.; Schaller, K.; Deutsch, A. ‘Go or Grow’: The key to the emergence of invasion in tumour progression? Math. Med. Biol. A J. IMA 2012, 29, 49–65. [Google Scholar] [CrossRef] [PubMed]
  32. Hoek, K.S.; Eichhoff, O.M.; Schlegel, N.C.; Döbbeling, U.; Kobert, N.; Schaerer, L.; Hemmi, S.; Dummer, R. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res. 2008, 68, 650–656. [Google Scholar] [CrossRef] [PubMed]
  33. Gil-Henn, H.; Patsialou, A.; Wang, Y.; Warren, M.S.; Condeelis, J.S.; Koleske, A.J. Arg/Abl2 promotes invasion and attenuates proliferation of breast cancer in vivo. Oncogene 2013, 32, 2622–2630. [Google Scholar] [CrossRef] [PubMed]
  34. Patsialou, A.; Wang, Y.; Pignatelli, J.; Chen, X.; Entenberg, D.; Oktay, M.; Condeelis, J.S. Autocrine CSF1R signaling mediates switching between invasion and proliferation downstream of TGFβ in claudin-low breast tumor cells. Oncogene 2015, 34, 2721–2731. [Google Scholar] [CrossRef]
  35. Yang, Y.; Ye, W.L.; Zhang, R.N.; He, X.S.; Wang, J.R.; Liu, Y.X.; Wang, Y.; Yang, X.M.; Zhang, Y.J.; Gan, W.J. The Role of TGF-β Signaling Pathways in Cancer and Its Potential as a Therapeutic Target. Evid. Based Complement. Altern. Med. 2021, 2021, 6675208. [Google Scholar] [CrossRef]
  36. Li, J.; Poi, M.J.; Tsai, M.D. Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry 2011, 50, 5566–5582. [Google Scholar] [CrossRef]
  37. Li, S.; Zhu, Y.; Liang, Z.; Wang, X.; Meng, S.; Xu, X.; Xu, X.; Wu, J.; Ji, A.; Hu, Z.; et al. Up-regulation of p16 by miR-877-3p inhibits proliferation of bladder cancer. Oncotarget 2016, 7, 51773–51783. [Google Scholar] [CrossRef]
  38. Jung, A.; Schrauder, M.; Oswald, U.; Knoll, C.; Sellberg, P.; Palmqvist, R.; Niedobitek, G.; Brabletz, T.; Kirchner, T. The invasion front of human colorectal adenocarcinomas shows co-localization of nuclear beta-catenin, cyclin D1, and p16INK4A and is a region of low proliferation. Am. J. Pathol. 2001, 159, 1613–1617. [Google Scholar] [CrossRef]
  39. Itoh, T.; Omori, Y.; Seino, M.; Hirose, K.; Date, F.; Ono, Y.; Mizukami, Y.; Aoki, S.; Ishida, M.; Mizuma, M.; et al. Gene Rearrangement and Expression of PRKACA and PRKACB Govern Morphobiology of Pancreatobiliary Oncocytic Neoplasms. Mod. Pathol. 2024, 3, 100358. [Google Scholar] [CrossRef]
  40. Saviana, M.; Le, P.; Micalo, L.; Del Valle-Morales, D.; Romano, G.; Acunzo, M.; Li, H.; Nana-Sinkam, P. Crosstalk between miRNAs and DNA Methylation in Cancer. Genes 2023, 14, 1075. [Google Scholar] [CrossRef]
  41. Liu, Y.; Sethi, N.S.; Hinoue, T.; Schneider, B.G.; Cherniack, A.D.; Sanchez-Vega, F.; Seoane, J.A.; Farshidfar, F.; Bowlby, R.; Islam, M.; et al. Comparative Molecular Analysis of Gastrointestinal Adenocarcinomas. Cancer Cell 2018, 33, 721–735.e8. [Google Scholar] [CrossRef] [PubMed]
  42. Ebili, H.O.; Agboola, A.O.; Rakha, E. MSI-WES: A simple approach for microsatellite instability testing using whole exome sequencing. Future Oncol. 2021, 17, 3595–3606. [Google Scholar] [CrossRef]
  43. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [PubMed]
  44. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed]
  45. Subramanian, A.; Tamayo, P.; Mootha, V.K.; Mukherjee, S.; Ebert, B.L.; Gillette, M.A.; Paulovich, A.; Pomeroy, S.L.; Golub, T.R.; Lander, E.S.; et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 2005, 102, 15545–15550. [Google Scholar] [CrossRef]
  46. Liberzon, A.; Birger, C.; Thorvaldsdóttir, H.; Ghandi, M.; Mesirov, J.P.; Tamayo, P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015, 1, 417–425. [Google Scholar] [CrossRef] [PubMed]
  47. Reich, M.; Liefeld, T.; Gould, J.; Lerner, J.; Tamayo, P.; Mesirov, J. GenePattern 2.0. Nat. Genet. 2006, 38, 500–501. [Google Scholar] [CrossRef]
  48. Templeton, G.F. A Two-Step Approach for Transforming Continuous Variables to Normal: Implications and Recommendations for IS Research. Commun. Assoc. Inf. Syst. 2011, 28, 41–58. [Google Scholar] [CrossRef]
  49. Yoo, M.; Shin, J.; Kim, J.; Ryall, K.A.; Lee, K.; Lee, S.; Jeon, M.; Kang, J.; Tan, A.C. DSigDB: Drug Signatures Database for Gene Set Analysis. Bioinformatics 2015, 31, 3069–3071. [Google Scholar] [CrossRef] [PubMed]
  50. Kuleshov, M.V.; Jones, M.R.; Rouillard, A.D.; Fernandez, N.F.; Duan, Q.; Wang, Z.; Koplev, S.; Jenkins, S.L.; Jagodnik, K.M.; Lachmann, A.; et al. Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016, 44, W90–W97. [Google Scholar] [CrossRef]
  51. Xie, Z.; Bailey, A.; Kuleshov, M.V.; Clarke, D.J.B.; Evangelista, J.E.; Jenkins, S.L.; Lachmann, A.; Wojciechowicz, M.L.; Kropiwnicki, E.; Jagodnik, K.M.; et al. Gene set knowledge discovery with Enrichr. Curr. Protoc. 2021, 1, e90. [Google Scholar] [CrossRef] [PubMed]
  52. Pujato, M.; Kieken, F.; Skiles, A.A.; Tapinos, N.; Fiser, A. Prediction of DNA binding motifs from 3D models of transcription factors; identifying TLX3 regulated genes. Nucleic Acids Res. 2014, 42, 13500–13512. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clustergram, bar chart, and Manhattan plot showing enrichment of gene targets of multiple therapeutic agents in the two PRKACA subsets of GC (Upper panel) and CRC (Lower panel). The full names of the ontology terms are listed in Supplementary Materials Drug-targets_PRKACA_high_GC and Drug-targets_PRKACA_low_GC.
Figure 1. Clustergram, bar chart, and Manhattan plot showing enrichment of gene targets of multiple therapeutic agents in the two PRKACA subsets of GC (Upper panel) and CRC (Lower panel). The full names of the ontology terms are listed in Supplementary Materials Drug-targets_PRKACA_high_GC and Drug-targets_PRKACA_low_GC.
Pharmaceuticals 17 01263 g001
Figure 2. Gene ontology enrichment in the PRKACA subsets of GC. The upper panel displays cluster gram, volcano plot, and a bar chart showing the enrichment of biological functions involved in cell migration/invasion in the PRKACA-high GC cohort. The lower panel shows charts that display the enrichment of biological processes associated with cell proliferation in the PRKACA-low GC cohort. Details of the gene ontology terms can be found in the Supplementary Materials GC_GSEA.PRKACA_high and GC_GSEA.PRKACA_low.
Figure 2. Gene ontology enrichment in the PRKACA subsets of GC. The upper panel displays cluster gram, volcano plot, and a bar chart showing the enrichment of biological functions involved in cell migration/invasion in the PRKACA-high GC cohort. The lower panel shows charts that display the enrichment of biological processes associated with cell proliferation in the PRKACA-low GC cohort. Details of the gene ontology terms can be found in the Supplementary Materials GC_GSEA.PRKACA_high and GC_GSEA.PRKACA_low.
Pharmaceuticals 17 01263 g002
Figure 3. Gene Ontology Enrichment Analysis of PRKACA-high subset of the CRC cohort. Clustergram, volcano plot, and bar chart showing enrichment of biological processes involved in cell proliferation. Details of the ontology terms and gene names displayed on the images are available in the Supplementary Material CRC_GSEA.PRKACA_high.
Figure 3. Gene Ontology Enrichment Analysis of PRKACA-high subset of the CRC cohort. Clustergram, volcano plot, and bar chart showing enrichment of biological processes involved in cell proliferation. Details of the ontology terms and gene names displayed on the images are available in the Supplementary Material CRC_GSEA.PRKACA_high.
Pharmaceuticals 17 01263 g003
Figure 4. Box plots showing significant correlations between PRKACA expression and copy number alterations in the GC (Upper) and CRC (Lower) cohorts.
Figure 4. Box plots showing significant correlations between PRKACA expression and copy number alterations in the GC (Upper) and CRC (Lower) cohorts.
Pharmaceuticals 17 01263 g004
Figure 5. Scatterplots showing significant relationships between PRKACA expression and the regressors (PRKACA methylation, PRKACA copy number alterations, PRKACA-targeting transcription factors, and miRNA species) in the GC (Upper) and CRC (Lower) cohorts.
Figure 5. Scatterplots showing significant relationships between PRKACA expression and the regressors (PRKACA methylation, PRKACA copy number alterations, PRKACA-targeting transcription factors, and miRNA species) in the GC (Upper) and CRC (Lower) cohorts.
Pharmaceuticals 17 01263 g005
Table 1. Clinicopathological correlates of PRKACA expression in gastric and colorectal cancers.
Table 1. Clinicopathological correlates of PRKACA expression in gastric and colorectal cancers.
PRKACA Expression
Clinicopathological FeaturesLow PRKACAHigh PRKACATotalX2 *p ValueAdjusted p Value
Gastric cancer cohort
Age Group30–60 yrs5279 131 7.6300.0060.016
61–90 yrs150126 276
Total 202 205 407
GenderMale1371282650.8570.3550.441
Female6978147
Total206206412
Race/EthnicityWhite1261332592.6090.1120.179
Native Americans101
Black10212
Asians315687
Total168191359
Pathological tumor stagepT1139220.7530.3860.441
pT2384987
pT38596181
pT46351114
Total199205404
Pathological nodal stageN046771239.7750.0020.008
N15455109
N2483179
N3463682
Total194199393
Pathological metastasis stageM01831813640.2800.5960.596
M1 and above151227
Total198193391
TNM stageEarly Stage458112613.226<0.0010.002
Late Stage152123275
Total197204401
Histological type of gastric cancerDiffuse type Adenocarcinoma103133.8920.0490.098
Intestinal type Adenocarcinoma196203399
Total206206412
Colorectal cancer cohort
Age Group31–60 yrs701031739.2950.0020.007
61–90 yrs198165363
Total268268536
Gender Male1411422830.0070.9310.931
Female127126253
Total268268536
Race/EthnicityWhite1161672833.8760.0490.106
Asian/Native American7613
Black342963
Total157202359
Pathological tumor stagepTis and pT1510150.1200.7290.790
pT2553388
pT3172196368
pT4362965
Total268268536
Pathological nodal stageN01701343047.1600.0070.018
N15576131
N2435699
Total268266534
Pathological metastasis stageM02021923940.2720.6020.711
M1353873
Total237230467
TNM stageEarly Stage1531292823.9560.04670.106
Late Stage115137252
Total268266534
Histological type of colorectal cancerAdenocarcinoma NOS2252404653.4520.0630.117
Mucinous Adenocarcinoma402666
Total265266531
Colonic tumor site 1Right colon1649225623.553<0.001<0.001
Left colon92128220
Total256220476
Primary tumor site Colon268177445108.404<0.001<0.001
Rectum09090
Total268267535
Vascular Invasion (VI)VI absent1801743541.7460.1860.269
VI present4862110
Total228236464
Lymphovascular Invasion LVI absent1531462990.5130.4740.616
LVI present8795182
Total240241481
Perineural Invasion (PNI)PNI absent75951701.8840.1700.269
PNI present203959
Total95134229
* Pearson Chi-square test for 2 × 2 tables, and Linear-by-linear association test for >2 × 2 tables; TNM = Tumor, Node and Metastasis.
Table 2. Molecular correlates of PRKACA expression in gastric and colorectal cancers.
Table 2. Molecular correlates of PRKACA expression in gastric and colorectal cancers.
PRKACA Expression
Molecular CorrelatesLow PRKACAHigh PRKACATotalX2 *p ValueAdjusted p Value
Gastric cancer cohort
Molecular subtypesCIN1201022227.2310.0070.014
MSI423173
GS183250
EBV111930
POLE257
Total 193 189 382
MSS vs. MSIMSS1401382781.1920.2750.275
MSI423173
Total182169351
Aneuploidy scoreLow Aneuploidy Score881101985.5630.0180.024
High Aneuploidy Score11287199
Total200197397
Global methylation scoreLow Global methylation score1287420228.455<0.001<0.001
High Global methylation score77131208
Total205205410
Colorectal cancer cohort
Molecular subtypesCIN11217829021.901<0.001<0.001
MSI422163
GS391655
POLE347
Total196219415
MSS vs. MSIMSS15419835211.2600.0010.002
MSI422163
Total196219415
Mutation CountLow Mutation count931202136.1680.0130.017
High Mutation count136110246
Total229230459
Fraction of Genome AlteredLow Fraction Genome Altered15111226310.5920.0010.002
High Fraction Genome Altered102136238
Total253248501
MANTIS ScoreLow MANTIS Score1091162250.7240.3950.395
High MANTIS Score135123258
Total244239483
MSI SensorLow MSI Sensor Score10615125719.578<0.001<0.001
High MSI Sensor Score15096246
Total256247503
Aneuploidy ScoreLow Aneuploidy Score120892098.5370.0030.005
High Aneuploidy Score92121213
Total212210422
Tumor Mutational Burden Low TMB831081913.7010.0540.061
High TMB115102217
Total198210408
* Pearson Chi-square test for 2 × 2 tables, and Linear-by-linear association test for >2 × 2 tables; CIN = chromosomal instability, MSI = microsatellite instability, EBV = Epstein-Barr Virus, GS = genome stable, POLE = polymerase e, MSS = microsatellite stable, MANTIS = Microsatellite Analysis for Normal Tumor InStability (computational tool for assessing the extent of MSI in cancers), MSI Sensor = an alternative computational tool for assessing MSI in cancers.
Table 3. Deregulation of PRKACA expression in gastric cancer *.
Table 3. Deregulation of PRKACA expression in gastric cancer *.
Gastric Cancer
RR2Adjusted R2S.E. of Estimate
0.7420.5500.526366.808
Coefficients
Unstandardized Coefficientstp
BS. E.
(Constant)2453.432240.52910.200<0.001
hsa-mir-4900.5860.1304.514<0.001
PRKACA CNV450.82569.9366.446<0.001
ZNF644 Expression−60.54115.418−3.927<0.001
ZNF557 Expression79.89624.8403.2160.001
cg178187983007.832745.0724.037<0.001
cg26613742−2233.574373.632−5.978<0.001
cg015965201031.285338.2173.0490.002
HEY2 Expression71.02018.8693.764<0.001
CDC5L Expression−8.8724.218−2.1030.036
TFEB Expression21.1757.5972.7870.006
cg19621460447.432160.5992.7860.006
hsa-mir-7641-113.5964.8052.8300.005
SP8 Expression−61.20021.970−2.7860.006
ZNF101 Expression45.03016.5022.7290.007
ZKSCAN3 Expression128.24746.1632.7780.006
ZNF451 Expression−97.30534.028−2.8600.005
cg01010868−29,345.39712879.446−2.2780.023
ANOVA
dfFp
Regression1722.777<0.001
Residual317
Total334
* Multiple linear regression analysis.
Table 4. Deregulation of PRKACA expression in colorectal cancer *.
Table 4. Deregulation of PRKACA expression in colorectal cancer *.
RR2Adjusted R2S.E. of Estimate
0.7730.5980.561259.799
Coefficients
Unstandardized Coefficientstp
BS. E.
(Constant)−520.078360.444−1.4430.150
ZBTB7A Expression31.0654.1777.437<0.001
hsa-mir-1430.0010.0002.4610.015
cg15814923−6980.8182832.849−2.4640.014
hsa-mir-577−0.5050.196−2.5780.011
TSHZ3 Expression92.83319.1044.859<0.001
DHX34 Expression18.6026.6492.7980.006
cg17818798−996.038497.031−2.0040.046
cg19586199−610.229186.442−3.2730.001
ZNF451 Expression−124.10127.985−4.434<0.001
ZNF442 Expression324.096163.8621.9780.049
ZKSCAN8 Expression68.66315.4814.435<0.001
MAFK Expression−15.6864.159−3.772<0.001
HOXA1 Expression80.33623.0283.4890.001
ZNF516 Expression−111.06223.760−4.674<0.001
cg171195682626.810942.3952.7870.006
hsa-mir-216a20.7267.7692.6680.008
ZNF140 Expression−37.66420.633−1.8250.069
ZNF557 Expression89.68934.6132.5910.010
hsa-mir-1-2−1.2780.643−1.9860.048
cg20110535846.904368.8662.2960.023
hsa-mir-5092−78.54631.141−2.5220.012
ZNF235 Expression−162.61478.187−2.0800.039
ANOVA
dfFp
Regression2216.538<0.001
Residual245
Total267
* Multiple linear regression analysis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Othaim, A.A.; Alasiri, G.; Alfahed, A. Therapeutic, Clinicopathological, and Molecular Correlates of PRKACA Expression in Gastrointestinal Cancers. Pharmaceuticals 2024, 17, 1263. https://doi.org/10.3390/ph17101263

AMA Style

Othaim AA, Alasiri G, Alfahed A. Therapeutic, Clinicopathological, and Molecular Correlates of PRKACA Expression in Gastrointestinal Cancers. Pharmaceuticals. 2024; 17(10):1263. https://doi.org/10.3390/ph17101263

Chicago/Turabian Style

Othaim, Ayoub Al, Glowi Alasiri, and Abdulaziz Alfahed. 2024. "Therapeutic, Clinicopathological, and Molecular Correlates of PRKACA Expression in Gastrointestinal Cancers" Pharmaceuticals 17, no. 10: 1263. https://doi.org/10.3390/ph17101263

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

Article Metrics

Back to TopTop