Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Bioinformatics and system biology approaches to determine the connection of SARS-CoV-2 infection and intrahepatic cholangiocarcinoma

  • Xinyi Zhou ,

    Contributed equally to this work with: Xinyi Zhou, Tengda Huang

    Roles Software

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Tengda Huang ,

    Contributed equally to this work with: Xinyi Zhou, Tengda Huang

    Roles Conceptualization, Formal analysis

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Hongyuan Pan,

    Roles Conceptualization, Data curation

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Ao Du,

    Roles Project administration, Resources

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Tian Wu,

    Roles Supervision, Validation

    Affiliation NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China

  • Jiang Lan,

    Roles Data curation, Investigation

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Yujia Song,

    Roles Conceptualization, Investigation

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Yue Lv,

    Roles Data curation, Project administration

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

  • Fang He,

    Roles Investigation, Methodology

    Affiliation Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China

  • Kefei Yuan

    Roles Conceptualization, Funding acquisition, Writing – original draft

    ykf13@163.com

    Affiliation Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China

Abstract

Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causal agent of coronavirus disease 2019 (COVID-19), has infected millions of individuals worldwide, which poses a severe threat to human health. COVID-19 is a systemic ailment affecting various tissues and organs, including the lungs and liver. Intrahepatic cholangiocarcinoma (ICC) is one of the most common liver cancer, and cancer patients are particularly at high risk of SARS-CoV-2 infection. Nonetheless, few studies have investigated the impact of COVID-19 on ICC patients.

Methods

With the methods of systems biology and bioinformatics, this study explored the link between COVID-19 and ICC, and searched for potential therapeutic drugs.

Results

This study identified a total of 70 common differentially expressed genes (DEGs) shared by both diseases, shedding light on their shared functionalities. Enrichment analysis pinpointed metabolism and immunity as the primary areas influenced by these common genes. Subsequently, through protein-protein interaction (PPI) network analysis, we identified SCD, ACSL5, ACAT2, HSD17B4, ALDOA, ACSS1, ACADSB, CYP51A1, PSAT1, and HKDC1 as hub genes. Additionally, 44 transcription factors (TFs) and 112 microRNAs (miRNAs) were forecasted to regulate the hub genes. Most importantly, several drug candidates (Periodate-oxidized adenosine, Desipramine, Quercetin, Perfluoroheptanoic acid, Tetrandrine, Pentadecafluorooctanoic acid, Benzo[a]pyrene, SARIN, Dorzolamide, 8-Bromo-cAMP) may prove effective in treating ICC and COVID-19.

Conclusion

This study is expected to provide valuable references and potential drugs for future research and treatment of COVID-19 and ICC.

1. Introduction

Coronavirus disease 2019 (COVID-19), first identified in December 2019 [1], is a recently discovered respiratory ailment caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While most COVID-19 patients experience mild to moderate symptoms, 5% suffer from acute respiratory distress syndrome (ARDS), multiple organ failure, or septic shock, and approximately 15% develop severe pneumonia [2]. New SARS-CoV-2 variants, such as Alpha, Beta, Delta, and Omicron, continue to emerge, leading to high case rates and significant global mortality. As of November 2023, the World Health Organization (WHO) has reported 772,052,752 COVID-19 cases, resulting in 6,985,278 deaths [3]. Previous research indicates that SARS-CoV-2 primarily infects human cells when its surface spike protein binds to the angiotensin-converting enzyme 2 (ACE2) receptor [4]. The spike protein is the protein for SARS-CoV-2 to recognize host cells and is also the main target of the human immune system [5].

Although the virus directly infects the lungs, its effect on the liver cannot be ignored. Patients with severe COVID-19 seem to have higher rates of liver dysfunction [6]. Patients with liver dysfunction are identified to have high risk of developing severe COVID-19 [7, 8]. Clinical data also demonstrate that patients with comorbidities fare worse than those without them [9, 10]. Besides, cancer patients undergoing chemotherapy or immunotherapy are more susceptible to COVID-19 infection [11]. Bioinformatics approaches have been employed to investigate the link between colorectal cancer and COVID-19 [12]. Liver cancer, ranking as the sixth most common and third deadliest malignancy globally [13], is closely associated with cirrhosis, Hepatitis B Virus (HBV) and Hepatitis C Virus (HCV) infections, and metabolic syndrome [14]. First-line (gemcitabine and cisplatin), second-line (FOLFOX), and adjuvant (capecitabine) systemic chemotherapy are currently the accepted standard of treatment of ICC [15]. In addition, ICC patients often experience severe liver dysfunction. Therefore, to better overcome COVID-19 and ICC in the future, it is imperative to explore and clarify the internal molecular mechanism between these two diseases.

This study utilized two datasets, GSE152418 and GSE119336, obtained from the Gene Expression Omnibus (GEO) database, to investigate the correlations between ICC and COVID-19. Differentially expressed genes (DEGs) were identified, and then 70 shared DEGs genes were found for both diseases. Pathway analysis was conducted using these mutual DEGs to gain insights into the underlying gene expression mechanisms. To gather hub genes, a protein-protein interaction (PPI) network was constructed using the 70 recognized DEGs. Next, the hub genes were used to elucidate the gene-regulatory network, predict potential drugs, and complete the gene-disease association network. A flowchart of the overall work is presented in Fig 1. The findings of this study will enhance our understanding of the interplay between COVID-19 and ICC, aid in drug selection, and facilitate the development of novel therapeutic strategies for combatting both diseases.

thumbnail
Fig 1. Schematic illustration of the overall general workflow of this study.

https://doi.org/10.1371/journal.pone.0300441.g001

2. Materials and methods

2.1 Collection of gene expression datasets

RNA-seq datasets from the NCBI [16], GEO (http://www.ncbi.nlm.nih.gov/geo) database were obtained to investigate common biological interrelationships between COVID-19 and ICC. The COVID-19 dataset, with GEO accession ID GSE152418, comprises transcriptional profiles from 17 healthy individuals and 17 COVID-19-infected individuals, sequenced using the Illumina NovaSeq 6000 platform (Homo sapiens) for RNA extraction [17]. The ICC dataset (GEO accession ID: GSE119336) contains 15 pairs of human ICC tumors and non-tumor liver tissues that were sequenced using an Illumina HiSeq 2000 (Homo sapiens) high-throughput sequencing donated by Zhang et al. [18].

2.2 Identification of differentially expressed genes and shared differentially expressed genes between ICC and COVID-19

DEGs are genes that exhibit significant differences in transcriptional levels among various test conditions [19]. The DEGs of GSE119336 and GSE152418 were identified from the expression values by R (version 4.2.1) software with the LIMMA package [20] and corrected by Benjamini-Hochberg to reduce the error detection rate (FDR). A cutoff criterion of FDR < 0.05 and |log2 Fold Change| > 1 was employed to identify significant DEGs in both datasets. The shared DEGs of GSE119336 and GSE152418 were acquired using the jvenn [21] (http://jvenn.toulouse.inra.fr/app/example.html), an online VENN graph mapping platform, to plot VENN analysis.

2.3 Gene ontology and pathway enrichment analysis

Gene enrichment analysis is a crucial analytical approach for categorizing genes into biological functions [22]. In order to understand the function of common DEGs, we performed gene ontology (GO) and pathway enrichment analyses connected with the mutual DEGs using Enrichr [23] (https://maayanlab.cloud/Enrichr/), a wide range of online gene set enrichment tool. The three types of GO database in the GO database are biological processes (BP), molecular functions (MF), and cellular components (CC). In pathway enrichment analysis, four databases were regarded, including the Kyoto Encyclopedia of Genes and Genomes (KEGG), Wikipathways, Reactome, and the Bioplanet. The P-value < 0.05 was used as a criterion to screen for reliable results.

2.4 Protein-protein interaction network analysis and hub gene extraction

The PPI was established using the STRING [24] (version 11.5) database (https://cn.string-db.org/), an online protein-protein association networks platform, and was then visualized and drawn as a network using Cytoscape [25] (version 3.9.1), an open source software platform for visualizing complex networks. A PPI network was constructed based on proteins encoded by common DEGs shared between COVID-19 and ICC, using a composite score threshold of 0.15. Hub genes, which demonstrate strong connections within potential modules [26], were predicted using the Cytoscape plug-in cytoHubba.

2.5 Gene-regulatory network analysis

To discover the transcriptional factors (TFs) and microRNAs (miRNAs) that regulate the hub genes post-transcriptionally, hub gene-TF interplay networks and hub gene-miRNA interaction networks have been dug by means of NetworkAnalyst [27] (version 3.0), a comprehensive visual analysis platform for gene expression profiling. The hub gene-TF interaction networks were built according to the JASPAR [28] database. Hub gene-miRNA interaction networks were constructed via the TarBase [29] (version 8.0) databases.

2.6 Gene-disease association analysis

In order to study the human genetic illnesses of shared genes between COVID-19 and ICC, DisGeNET [30] (https://www.disgenet.org/), a publicly accessible database containing information on approximately 24,000 diseases, 17,000 genes, and 117,000 genetic variations related to human illnesses, was used in our analysis. Similarly, NetworkAnalyst and Cytoscape were used to dig gene-disease relationships in order to find diseases associated with common DEGs.

2.7 Evaluation of candidate drugs

To anticipate protein-drug interactions and discover prospective pharmacological substances related to hub genes, we employed the Drug Signatures Database [31] (DSigDB, http://tanlab.ucdenver.edu/DSigDB), which contains 17,389 unique chemicals that span 19,531 genes and has 22,527 gene sets. Using the Enrichr web server and the DSigDB database, medicines targeting hub genes were identified between the COVID-19 and ICC datasets using a statistical threshold of P-value < 0.05.

3. Results

3.1 Recognition of differentially expressed genes and biological relationships between ICC and COVID-19

In order to evaluate the interactions and implications of ICC with COVID-19, the RNA-seq dataset was examined from the National Center for Biotechnology Information (NCBI). In the ICC dataset, 2,537 DEGs was found, including 1,095 were up-regulated and 1,442 were down-regulated (Fig 2A and S1 Table). Meanwhile, there are 1,267 up-regulated and 891 down-regulated genes showed in the COVID-19 dataset (Fig 2B and S2 Table). Table 1 is a list of the condensed data of DEGs. With the use of the cross-comparative analysis, we were able to find 70 DEGs that were shared by the ICC and COVID-19 datasets (Fig 2C and S3 Table). These outcomes revealed that COVID-19 and ICC had certain molecular similarities.

thumbnail
Fig 2. Volcano plots and Venn diagram depicts the shared DEGs among COVID-19 and ICC.

Volcano plots of (A) COVID-19 and (B) ICC, with genes with |log2Fold Change| > 1 and FDR < 0.05. (C) The Venn diagram depicts the shared DEGs among COVID-19 and ICC.

https://doi.org/10.1371/journal.pone.0300441.g002

thumbnail
Table 1. Overview of datasets with their geo-features and their quantitative measurements in this analysis.

https://doi.org/10.1371/journal.pone.0300441.t001

3.2 Gene enrichment analyses of shared DEGs

Our study used gene ontology and pathway enrichment analysis to learn more about these typical DEGs’ roles and signaling pathways. Gene functional similarity is frequently assessed using the GO enrichment analysis [32]. A modeling technique called pathway analysis is used to show how crucial molecular or biological processes interact and illustrate the reciprocal impacts of various diseases [33]. In order to uncover highly enriched functional GO keywords and pathways, we ran a functional-enrichment test on common DEGs using the Enrichr program.

70 common DEGs were enriched in 334 terms, including 253 biological processes, 65 molecular functions, and 16 cellular components (S4 Table). Then we summarized the top 10 terms according to P-value in each category in Table 2 and visualized in Fig 3A–3C. It can be found that many of these terms are related to metabolism and immunity, such as lipid biosynthetic process (GO:0008610) and negative regulation of dendritic cell apoptotic process (GO:2000669), which have a strong association with COVID-19 and ICC.

thumbnail
Fig 3. Analysis of common DEGs between COVID-19 and ICC using ontology and pathway enrichment.

Ontological analysis: (A) Biological processes, (B) Molecular function, and (C) Cellular components. Pathway enrichment analysis: (D) KEGG, (E) Wikipathways, (F) Reactome, and the (G) Bioplanet.

https://doi.org/10.1371/journal.pone.0300441.g003

thumbnail
Table 2. Ontological analysis of common DEGs between COVID-19 and ICC.

https://doi.org/10.1371/journal.pone.0300441.t002

We found 22 reliable pathways in Kyoto Encyclopedia of Genes and Genomes (KEGG), 32 reliable pathways in Wikipathways, 62 reliable pathways in Reatcome, and 77 reliable pathways in Bioplanet (S5 Table). The top 10 reliable pathways found in each database are listed in Table 3, and the bar graphs of pathway enrichment analysis are shown in Fig 3D–3G. In these pathways, more about the metabolic pathways were discovered, such as glyoxylate and dicarboxylate metabolism in KEGG, fatty acid biosynthesis in Wikipathways, metabolism of steroids in Reactome, and bile acid and bile salt metabolism in BioPlanet, which indicated that COVID-19 and ICC have common effects on these pathways.

thumbnail
Table 3. Pathway enrichment analysis of common DEGs between COVID-19 and ICC.

https://doi.org/10.1371/journal.pone.0300441.t003

3.3 Protein-protein interaction networks analysis and identification of hub genes

To better understand biological signals, response mechanisms of energy substance metabolism, and functional links between proteins in disease states, this study obtained the PPI network via STRING. Subsequently, PPI was visualized in Cytoscape to forecast interaction between common protein-coding DEGs. The PPI network of common DEGs consists of 65 nodes and 177 edges (Fig 4 and S6 Table). According to PPI network analysis integrating Cytohubba plugin in Cytoscape, we ranked the most interconnected nodes top 10 DEGs (14.28%) as hub genes. The hub genes are as follows: SCD, ACSL5, ACAT2, HSD17B4, ALDOA, ACSS1, ACADSB, CYP51A1, PSAT1, and HKDC1. With the aid of the Cytohubba plugin, we also built a network of submodules to better comprehend their closeness and close connection, including 35 nodes and 114 edges (Fig 5). In the following analysis, we will focus on these 10 hub genes. These hub genes show potential biomarkers that can provide new therapeutic strategies for COVID-19 and ICC.

thumbnail
Fig 4. PPI network of common DEGs between COVID-19 and ICC.

The circular nodes in the figure stand in for DEGs, while the edges indicate node interactions. The PPI network consists of 177 edges and 65 nodes. String was used to create the PPI network, and Cytoscape was used to display it.

https://doi.org/10.1371/journal.pone.0300441.g004

thumbnail
Fig 5. Determination of hub genes from the PPI network by using the Cytohubba plugin in Cytoscape.

Hub genes were obtained using the Cytohubba plugin. Here, the red nodes indicate the highlighted top 10 hub genes and their interactions with other molecules. The network consists of 35 nodes and 144 edges.

https://doi.org/10.1371/journal.pone.0300441.g005

3.4 Construction of regulatory networks at transcriptional level

To better understand the regulatory hub genes and detect the key alterations at the transcriptional level, we used network analysis to search for TFs and miRNAs of regulatory hub genes. The TFs-hub genes interactions are shown in Fig 6, and the information of interaction is presented in S7 Table. In the network, 44 TFs have been found. And ALDOA, CYP51A1, ACSL5, SCD, and CREB1 were more highly expressed among hub genes as genes have a higher degree in the network of TF-hub gene interactions. S1 Fig and S8 Table depict the relationships of miRNA-hub genes. By the similar method, multiple discovered hub genes were projected to be regulated by 112 miRNAs, such as SCD, ALDOA, PSAT1, and CYP51A1. In-depth study of these genes has common implications for treating COVID-19 and ICC.

thumbnail
Fig 6. Interaction network of hub-gene-TFs.

The cohesive regulatory interaction network of hub-gene-TFs obtained from the Network Analyst and described by Cytoscape. Herein, the green nodes are TFs, and the yellow nodes are hub genes.

https://doi.org/10.1371/journal.pone.0300441.g006

3.5 Gene-disease association analysis

If different diseases have one or more similar genes, then we consider these diseases to be related to each other [34]. A total of 263 diseases were found to be associated with common genes and screened for significant diseases associated with at least two common genes (Fig 7). In our network, many diseases related to liver and cancer have been found, such as cholestasis, elevated hepatic transaminases, fatty liver, liver cirrhosis, liver dysfunction, mammary neoplasms, neoplasm invasiveness, neoplasm metastasis, non-small cell lung carcinoma, and prostatic neoplasms. Besides, the gene-disease association analysis also found some psychiatric disorders, including epilepsy, hyperreflexia, schizophrenia, and cognitive delay. These results portend the common association of COVID-19 and ICC with these diseases.

thumbnail
Fig 7. The gene-disease association network represents diseases associated with common genes.

The diseases are depicted by the square node and gene symbols are defined by the circle node.

https://doi.org/10.1371/journal.pone.0300441.g007

3.6 Identification of candidate drugs

To discover potential drugs for COVID-19 and ICC, we analyzed the protein-drug interactions of hub genes [35]. Potential therapeutic drugs were identified from the DSigDB database based on transcription characteristics using enrichment, and the top 10 candidate compounds (Periodate-oxidized adenosine, Desipramine, Quercetin, Perfluoroheptanoic acid, Tetrandrine, Pentadecafluorooctanoic acid, Benzo[a]pyrene, SARIN, Dorzolamide, 8-Bromo-cAMP) were identified based on P-value. The top ten enriched drugs in the DSigDB database are shown in Table 4, which indicated potential therapeutic effects on both COVID-19 and ICC.

4. Discussion

The 2019 SARS-CoV-2 global pandemic has riveted the world’s attention. With more and more variants of the virus, the transmission rate and morbidity rate of COVID-19 gradually increased. Although COVID‐19 primarily affects the respiratory system, liver dysfunction is also common in COVID-19 patients, such as elevated liver transaminases and elevations of cholestatic liver enzymes [36]. ICC, which is the second most common liver cancer, may be linked to COVID-19. To verify this idea, we use bioinformatics methods to find the relationship between these two diseases, and dig out some potential drugs This study could establish a link between COVID-19 and ICC and suggest possible treatment options for ICC patients infected with COVID-19.

Lipid biosynthetic process (GO:0008610), glyoxylate and dicarboxylate metabolism pathway (KO:00630), and fatty acid degradation pathway (KO:00071) are found in GO terms and KEGG pathways. Research detected that COVID-19 patients was dysregulated metabolites involved in lipid metabolism [37]. Another study also found that SARS-CoV-2 infection elevated the expression of the RE1-silencing transcription factor (REST), which regulated the transcriptional expression of secreted metabolic factors such as myeloperoxidase, apelin, and myostatin, causing disruptions in glucose and lipid metabolism [38]. Moreover, recent studies have found that altered lipid metabolism is a new hallmark of cancer [39]. A study found that KDM5C, a histone H3K4-specific demethylase, can repress FASN-mediated lipid metabolism to exert tumor suppressor activity in ICC [40]. Consistent with the results of GO and pathway analysis, there are also many genes related to metabolism in hub genes. Stearoyl-CoA desaturase (SCD) was reported to plays a key role in lipid biosynthesis pathways involved in tumorigenesis, and so pharmacological inhibitors have been developed such as MF-438, CAY10566 and A939572 [41], but has few research in ICC. In addition, both ACSL5 and HSD17B4 were found to be associated with fatty acid synthesis, which may indicate the impact of COVID-19 and ICC on lipid metabolism. These results suggested that COVID-19 and ICC may jointly affect lipid metabolic function of human body. However, whether lipid metabolism can be a therapeutic target for these two diseases needs further study.

Regulation of dendritic cell apoptotic process (GO:2000668), negative regulation of leukocyte apoptotic process (GO:2000107), positive regulation of GTPase activity (GO:0043547), and positive regulation of lymphocyte migration (GO:2000403) are related to immunity, which suggested that both ICC and COVID-19 have a huge impact on the immune system. SARS-CoV-2 has been demonstrated to alter normal immune responses, resulting in a weakened immune system and uncontrolled inflammatory reactions in COVID-19 severe and critical patients [42], which is the major cause of ARDS. Plasma levels of IL-2, IL-7, IL-10, granulocyte colony-stimulating factor (G-CSF), IP-10, MCP1, macrophage inflammatory protein 1α (MIP1α), and tumor necrosis factor (TNF) have been observed in patients with severe COVID-19 were higher than in healthy adults [43]. On the other hand, cancer is usually associated with immune escape by suppressing the immune system. A study found that tumor-derived exosomal miR-183-5p up-regulates PD-L1-expressing macrophages to foster immune suppression and disease progression in ICC through the miR-183-5p/PTEN/AKT/PD-L1 pathway [44]. Additionally, in this study, complement and coagulation cascades related pathways are found in top 10 pathway in each database. It has been shown that SARS-CoV-2 may activate the complement system’s classical and lectin pathways [45], and lectin pathways components were found deposited in lung tissue of COVID-19 patients [46], which is consistent with the results of our pathway analysis. Meanwhile, the complement system may be involved in liver dysfunction in viral-induced acute liver failure cases [47]. The aforementioned hub protein SCD also plays a role in immune function. A recent study found that suppression of SCD reduces humoral immune response to immunization and weakens immune defense against respiratory influenza infection [48]. But SCD1 expressed in cancer cells and immune cells causes immune resistance conditions, and its inhibition augments antitumor T cells and therapeutic effects of anti-PD-1 antibody [49]. Not only that, hub gene PSAT1 can also enhance immunosuppressive through PERK-ATF4-PSAT1 axis in tumor [50, 51]. CREB1, a TF with the highest correlation score in our TF-hub gene interaction analysis, was reported to promote T cell cytotoxicity [52]. In conclusion, both COVID-19 and ICC can elicit immune system responses. COVID-19 usually causes elevated inflammatory immune response, while ICC causes immune suppression. But the combined effect of these two immune responses on human body is unknown.

There was a report that a patient diagnosed with advanced Hodgkin’s lymphoma, who was not being treated for lymphoma, contracted COVID-19 and four months after ending treatment for COVID-19, was re-examined by PET-CT and found that most of his tumors had disappeared, with levels of biomarkers associated with the tumor dropping by more than 90% [53]. Interestingly, the associations between COVID-19 and cancer were also identified in our study. The hub gene in our analysis ACSL5, ALDOA, and HKDC1 are directly associated with liver cancer [5456]. Besides, gene-disease network analysis found some cancer related diseases, including mammary neoplasms, non-small cell lung carcinoma, and prostatic neoplasms. At the same time, neoplasm invasiveness and neoplasm metastasis are also showing in the result, which suggested that the ICC patient with COVID-19 may have a risk of developing other types of tumors and metastases. Similarly, in TF-gene network, SREBF1 was found to enhance the viability and motility in cancer [57]. The above evidences suggest that COVID-19 may have an effect on tumor migration and metastasis in ICC patients, but the detailed effect and mechanism require further investigation.

Regarding drug prediction, several chemical substances have shown promise as potential treatments for COVID-19, including quercetin and tetrandrine [58, 59]. Notably, these drugs also possess anti-cancer properties. Quercetin can influence pathways such as PI3K/Akt/mTOR, Wnt/β-catenin, and MAPK/ERK1/2 to induce apoptosis in cancer cells [60]. Tetrandrine, another candidate, has anti-angiogenic properties [61]. Therefore, it is plausible that ICC patients infected with COVID-19 could benefit from these drugs. Besides, the top 2 candidates, Periodate-oxidized adenosine and desipramine, have been reported to have some anticancer effects [62, 63], despite not being originally intended for that purpose.

However, bioinformatics, which is based on the advancement of modern computer technology and the simplicity of biological experimental techniques, cannot replace clinical testing [64]. Additionally, the selected datasets in this study include different groups of people with two different diseases, rather than the same population with both ICC and COVID-19, which may lead to some differences between the results of our analysis and the actual results. To ensure the credibility of these findings, it is essential to conduct in vivo, in vitro, and clinical studies to validate the results of the bioinformatics analysis. Furthermore, this study suggests that therapeutic approaches for ICC and COVID-19 comorbidity can be further explored from the perspective of lipid metabolism and immunology. And whether potential candidate drugs can treat ICC and covid-19 at the same time is also a topic worthy of further research.

5. Conclusions

To help gain insight into the connection between ICC and COVID-19, we utilized transcriptomic data analysis to identify differentially expressed genes shared in both diseases. A total of 70 common DEGs and 10 hub genes revealed certain similarities between ICC and COVID-19 in terms of pathogenic processes. Further, we identified 44 TFs and 112 miRNAs by building a transcriptional regulatory network targeting hub genes. Notably, drug prediction results indicate quercetin and tetrandrine as potential agents for the treatment of ICC and COVID-19. Although our study has certain limitations, these results can provide ideas and directions for subsequent research on the two diseases, such as target screening, targeted therapy, and drug development. Overall, this study could shed new light on the treatment and drug development of ICC and COVID-19.

Supporting information

S1 Table. Differentially expressed genes in GSE119336.

https://doi.org/10.1371/journal.pone.0300441.s001

(XLSX)

S2 Table. Differentially expressed genes in GSE152418.

https://doi.org/10.1371/journal.pone.0300441.s002

(XLSX)

S3 Table. Shared DEGs between GSE119336 and GSE152418.

https://doi.org/10.1371/journal.pone.0300441.s003

(XLSX)

S5 Table. KEGG, Wikipathways, Reactome, and Bioplanet of common DEGs.

https://doi.org/10.1371/journal.pone.0300441.s005

(XLSX)

S1 Fig. The interconnected regulatory interaction network of hub-gene-miRNAs.

Herein, the blue nodes indicate miRNAs and the red nodes are hub genes.

https://doi.org/10.1371/journal.pone.0300441.s009

(TIF)

Acknowledgments

We thank all patients who participated in this study and donated samples and the database GEO for providing their platform.

References

  1. 1. Fauci AS, Lane HC, Redfield RR. Covid-19—navigating the uncharted. Mass Medical Soc; 2020. p. 1268–9.
  2. 2. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8(4):420–2. Epub 2020/02/23. pmid:32085846; PubMed Central PMCID: PMC7164771.
  3. 3. Weekly Epidemiological Update on COVID‐19 [updated 2023]. Available from: https://www.who.int/publications/m/item/weekly-epidemiologicalupdate-on-covid-19—14-december-2022.
  4. 4. Huang T, JIANG N, Song Y, Pan H, Yu B, Li X, et al. Bioinformatics and system biology approach to identify the influences of SARS-CoV2 on metabolic unhealthy obese patients. Frontiers in Molecular Biosciences. 10:1274463. pmid:37877121
  5. 5. Gaziano L, Giambartolomei C, Pereira AC, Gaulton A, Posner DC, Swanson SA, et al. Actionable druggable genome-wide Mendelian randomization identifies repurposing opportunities for COVID-19. Nat Med. 2021;27(4):668–76. Epub 2021/04/11. pmid:33837377; PubMed Central PMCID: PMC7612986.
  6. 6. Zhang C, Shi L, Wang FS. Liver injury in COVID-19: management and challenges. Lancet Gastroenterol Hepatol. 2020;5(5):428–30. Epub 2020/03/08. pmid:32145190; PubMed Central PMCID: PMC7129165.
  7. 7. Huang T, Zheng D, Song Y, Pan H, Qiu G, Xiang Y, et al. Demonstration of the impact of COVID-19 on metabolic associated fatty liver disease by bioinformatics and system biology approach. Medicine. 2023;102(35):e34570. pmid:37657050
  8. 8. Huang T, Yu B, Zhou X, Pan H, Du A, Bai J, et al. Exploration of the link between COVID‐19 and alcoholic hepatitis from the perspective of bioinformatics and systems biology. MedComm–Future Medicine. 2023;2(2):e42.
  9. 9. Guan W-j, Liang W-h, Zhao Y, Liang H-r, Chen Z-s, Li Y-m, et al. Comorbidity and its impact on 1590 patients with COVID-19 in China: a nationwide analysis. European Respiratory Journal. 2020;55(5).
  10. 10. Ejaz H, Alsrhani A, Zafar A, Javed H, Junaid K, Abdalla AE, et al. COVID-19 and comorbidities: Deleterious impact on infected patients. Journal of infection and public health. 2020;13(12):1833–9. pmid:32788073
  11. 11. Liang W, Guan W, Chen R, Wang W, Li J, Xu K, et al. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020;21(3):335–7. Epub 2020/02/19. pmid:32066541; PubMed Central PMCID: PMC7159000.
  12. 12. Song Y, Huang T, Pan H, Du A, Wu T, Lan J, et al. The influence of COVID-19 on colorectal cancer was investigated using bioinformatics and systems biology techniques. Frontiers in Medicine. 2023;10. pmid:37457582
  13. 13. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49. Epub 2021/02/05. pmid:33538338.
  14. 14. Bridgewater J, Galle PR, Khan SA, Llovet JM, Park JW, Patel T, et al. Guidelines for the diagnosis and management of intrahepatic cholangiocarcinoma. J Hepatol. 2014;60(6):1268–89. Epub 2014/04/01. pmid:24681130.
  15. 15. Kelley RK, Bridgewater J, Gores GJ, Zhu AX. Systemic therapies for intrahepatic cholangiocarcinoma. J Hepatol. 2020;72(2):353–63. Epub 2020/01/20. pmid:31954497.
  16. 16. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets—update. Nucleic Acids Res. 2013;41(Database issue):D991–5. Epub 2012/11/30. pmid:23193258; PubMed Central PMCID: PMC3531084.
  17. 17. Arunachalam PS, Wimmers F, Mok CKP, Perera R, Scott M, Hagan T, et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science. 2020;369(6508):1210–20. Epub 2020/08/14. pmid:32788292; PubMed Central PMCID: PMC7665312.
  18. 18. Zhang C, Zhang B, Meng D, Ge C. Comprehensive analysis of DNA methylation and gene expression profiles in cholangiocarcinoma. Cancer cell international. 2019;19:352. Epub 2020/01/01. pmid:31889904; PubMed Central PMCID: PMC6933876.
  19. 19. Costa-Silva J, Domingues D, Lopes FM. RNA-Seq differential expression analysis: An extended review and a software tool. PLoS One. 2017;12(12):e0190152. Epub 2017/12/22. pmid:29267363; PubMed Central PMCID: PMC5739479.
  20. 20. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47. Epub 2015/01/22. pmid:25605792; PubMed Central PMCID: PMC4402510.
  21. 21. Bardou P, Mariette J, Escudie F, Djemiel C, Klopp C. jvenn: an interactive Venn diagram viewer. BMC Bioinformatics. 2014;15(1):293. Epub 2014/09/02. pmid:25176396; PubMed Central PMCID: PMC4261873.
  22. 22. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50. Epub 2005/10/04. pmid:16199517; PubMed Central PMCID: PMC1239896.
  23. 23. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128. Epub 2013/04/17. pmid:23586463; PubMed Central PMCID: PMC3637064.
  24. 24. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–D13. Epub 2018/11/27. pmid:30476243; PubMed Central PMCID: PMC6323986.
  25. 25. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. Epub 2003/11/05. pmid:14597658; PubMed Central PMCID: PMC403769.
  26. 26. Liu Y, Gu HY, Zhu J, Niu YM, Zhang C, Guo GL. Identification of Hub Genes and Key Pathways Associated With Bipolar Disorder Based on Weighted Gene Co-expression Network Analysis. Front Physiol. 2019;10:1081. Epub 2019/09/05. pmid:31481902; PubMed Central PMCID: PMC6710482.
  27. 27. Zhou G, Soufan O, Ewald J, Hancock REW, Basu N, Xia J. NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res. 2019;47(W1):W234–W41. Epub 2019/04/02. pmid:30931480; PubMed Central PMCID: PMC6602507.
  28. 28. Sandelin A, Alkema W, Engstrom P, Wasserman WW, Lenhard B. JASPAR: an open-access database for eukaryotic transcription factor binding profiles. Nucleic Acids Research. 2004;32(suppl_1):D91–D4. WOS:000188079000018. pmid:14681366
  29. 29. Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, et al. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA–gene interactions. Nucleic acids research. 2018;46(D1):D239–D45. pmid:29156006
  30. 30. Pinero J, Ramirez-Anguita JM, Sauch-Pitarch J, Ronzano F, Centeno E, Sanz F, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48(D1):D845–D55. Epub 2019/11/05. pmid:31680165; PubMed Central PMCID: PMC7145631.
  31. 31. Yoo M, Shin J, Kim J, Ryall KA, Lee K, Lee S, et al. DSigDB: drug signatures database for gene set analysis. Bioinformatics. 2015;31(18):3069–71. Epub 2015/05/21. pmid:25990557; PubMed Central PMCID: PMC4668778.
  32. 32. Liu W, Liu J, Rajapakse JC. Gene Ontology Enrichment Improves Performances of Functional Similarity of Genes. Sci Rep. 2018;8(1):12100. Epub 2018/08/16. pmid:30108262; PubMed Central PMCID: PMC6092333.
  33. 33. Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–D51. Epub 2020/10/31. pmid:33125081; PubMed Central PMCID: PMC7779016.
  34. 34. Al-Mustanjid M, Mahmud SMH, Royel MRI, Rahman MH, Islam T, Rahman MR, et al. Detection of molecular signatures and pathways shared in inflammatory bowel disease and colorectal cancer: A bioinformatics and systems biology approach. Genomics. 2020;112(5):3416–26. Epub 2020/06/15. pmid:32535071.
  35. 35. Mahmud SMH, Al-Mustanjid M, Akter F, Rahman MS, Ahmed K, Rahman MH, et al. Bioinformatics and system biology approach to identify the influences of SARS-CoV-2 infections to idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease patients. Brief Bioinform. 2021;22(5):bbab115. Epub 2021/04/14. pmid:33847347; PubMed Central PMCID: PMC8083324.
  36. 36. Nardo AD, Schneeweiss-Gleixner M, Bakail M, Dixon ED, Lax SF, Trauner M. Pathophysiological mechanisms of liver injury in COVID-19. Liver international: official journal of the International Association for the Study of the Liver. 2021;41(1):20–32. Epub 2020/11/16. pmid:33190346; PubMed Central PMCID: PMC7753756.
  37. 37. Shen B, Yi X, Sun Y, Bi X, Du J, Zhang C, et al. Proteomic and Metabolomic Characterization of COVID-19 Patient Sera. Cell. 2020;182(1):59–72 e15. Epub 2020/06/04. pmid:32492406; PubMed Central PMCID: PMC7254001.
  38. 38. He X, Liu C, Peng J, Li Z, Li F, Wang J, et al. COVID-19 induces new-onset insulin resistance and lipid metabolic dysregulation via regulation of secreted metabolic factors. Signal transduction and targeted therapy. 2021;6(1):427. Epub 2021/12/18. pmid:34916489; PubMed Central PMCID: PMC8674414.
  39. 39. Vriens K, Christen S, Parik S, Broekaert D, Yoshinaga K, Talebi A, et al. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature. 2019;566(7744):403–6. Epub 2019/02/08. pmid:30728499; PubMed Central PMCID: PMC6390935.
  40. 40. Zhang B, Zhou BH, Xiao M, Li H, Guo L, Wang MX, et al. KDM5C Represses FASN-Mediated Lipid Metabolism to Exert Tumor Suppressor Activity in Intrahepatic Cholangiocarcinoma. Front Oncol. 2020;10:1025. Epub 2020/07/28. pmid:32714863; PubMed Central PMCID: PMC7344276.
  41. 41. Raggi C, Taddei ML, Rae C, Braconi C, Marra F. Metabolic reprogramming in cholangiocarcinoma. J Hepatol. 2022;77(3):849–64. Epub 2022/05/21. pmid:35594992.
  42. 42. Tay MZ, Poh CM, Renia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nature reviews Immunology. 2020;20(6):363–74. Epub 2020/04/30. pmid:32346093; PubMed Central PMCID: PMC7187672.
  43. 43. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. Epub 2020/01/28. pmid:31986264; PubMed Central PMCID: PMC7159299.
  44. 44. Luo C, Xin H, Zhou Z, Hu Z, Sun R, Yao N, et al. Tumor-derived exosomes induce immunosuppressive macrophages to foster intrahepatic cholangiocarcinoma progression. Hepatology. 2022;76(4):982–99. Epub 2022/02/03. pmid:35106794.
  45. 45. Matricardi PM, Dal Negro RW, Nisini R. The first, holistic immunological model of COVID-19: Implications for prevention, diagnosis, and public health measures. Pediatr Allergy Immunol. 2020;31(5):454–70. Epub 2020/05/03. pmid:32359201; PubMed Central PMCID: PMC7267459.
  46. 46. Gao T, Hu M, Zhang X, Li H, Zhu L, Liu H, et al. Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. MedRxiv. 2020.
  47. 47. Melgaco JG, Veloso CE, Pacheco-Moreira LF, Vitral CL, Pinto MA. Complement System as a Target for Therapies to Control Liver Regeneration/Damage in Acute Liver Failure Induced by Viral Hepatitis. Journal of immunology research. 2018;2018:3917032. Epub 2018/11/08. pmid:30402508; PubMed Central PMCID: PMC6196788.
  48. 48. Zhou X, Zhu X, Li C, Li Y, Ye Z, Shapiro VS, et al. Stearoyl-CoA Desaturase-Mediated Monounsaturated Fatty Acid Availability Supports Humoral Immunity. Cell Rep. 2021;34(1):108601. Epub 2021/01/07. pmid:33406440; PubMed Central PMCID: PMC7839063.
  49. 49. Katoh Y, Yaguchi T, Kubo A, Iwata T, Morii K, Kato D, et al. Inhibition of stearoyl-CoA desaturase 1 (SCD1) enhances the antitumor T cell response through regulating beta-catenin signaling in cancer cells and ER stress in T cells and synergizes with anti-PD-1 antibody. Journal for immunotherapy of cancer. 2022;10(7). Epub 2022/07/07. pmid:35793868; PubMed Central PMCID: PMC9260842.
  50. 50. Raines LN, Zhao H, Wang Y, Chen HY, Gallart-Ayala H, Hsueh PC, et al. PERK is a critical metabolic hub for immunosuppressive function in macrophages. Nature immunology. 2022;23(3):431–45. Epub 2022/03/02. pmid:35228694; PubMed Central PMCID: PMC9112288.
  51. 51. Pratap UP, Vadlamudi RK. PERK promotes immunosuppressive M2 macrophage phenotype by metabolic reprogramming and epigenetic modifications through the PERK-ATF4-PSAT1 axis. Immunometabolism (Cobham (Surrey, England)). 2022;4(3):e00007. Epub 2022/08/16. pmid:35966637; PubMed Central PMCID: PMC9359065.
  52. 52. Maekawa Y, Minato Y, Ishifune C, Kurihara T, Kitamura A, Kojima H, et al. Notch2 integrates signaling by the transcription factors RBP-J and CREB1 to promote T cell cytotoxicity. Nature immunology. 2008;9(10):1140–7. Epub 2008/08/30. pmid:18724371.
  53. 53. Challenor S, Tucker D. SARS-CoV-2-induced remission of Hodgkin lymphoma. British Journal of Haematology. 2021;192(3):415–. WOS:000604040200001. pmid:33386647
  54. 54. Quan J, Bode AM, Luo X. ACSL family: The regulatory mechanisms and therapeutic implications in cancer. European journal of pharmacology. 2021;909:174397. Epub 2021/08/02. pmid:34332918.
  55. 55. Niu Y, Lin Z, Wan A, Sun L, Yan S, Liang H, et al. Loss-of-Function Genetic Screening Identifies Aldolase A as an Essential Driver for Liver Cancer Cell Growth Under Hypoxia. Hepatology. 2021;74(3):1461–79. Epub 2021/04/05. pmid:33813748; PubMed Central PMCID: PMC8518375.
  56. 56. Dong L, Lu D, Chen R, Lin Y, Zhu H, Zhang Z, et al. Proteogenomic characterization identifies clinically relevant subgroups of intrahepatic cholangiocarcinoma. Cancer cell. 2022;40(1):70–87 e15. Epub 2022/01/01. pmid:34971568.
  57. 57. Li LY, Yang Q, Jiang YY, Yang W, Jiang Y, Li X, et al. Interplay and cooperation between SREBF1 and master transcription factors regulate lipid metabolism and tumor-promoting pathways in squamous cancer. Nature Communications. 2021;12(1):1–17. ARTN 4362. WOS:000677489800015. pmid:34272396
  58. 58. Derosa G, Maffioli P, D’Angelo A, Di Pierro F. A role for quercetin in coronavirus disease 2019 (COVID-19). Phytotherapy research: PTR. 2021;35(3):1230–6. Epub 2020/10/10. pmid:33034398; PubMed Central PMCID: PMC7675685.
  59. 59. Heister PM, Poston RN. Pharmacological hypothesis: TPC2 antagonist tetrandrine as a potential therapeutic agent for COVID-19. Pharmacology research & perspectives. 2020;8(5):e00653. Epub 2020/09/16. pmid:32930523; PubMed Central PMCID: PMC7503088.
  60. 60. Reyes-Farias M, Carrasco-Pozo C. The Anti-Cancer Effect of Quercetin: Molecular Implications in Cancer Metabolism. International journal of molecular sciences. 2019;20(13). Epub 2019/07/03. pmid:31261749; PubMed Central PMCID: PMC6651418.
  61. 61. N B, K RC. Tetrandrine and cancer—An overview on the molecular approach. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2018;97:624–32. Epub 2017/11/05. pmid:29101806.
  62. 62. Cory JG, Mansell MM, George CB, Wilkinson DS. Inhibition of nucleic acid synthesis in Ehrlich tumor cells by periodate-oxidized adenosine and adenylic acid. Archives of Biochemistry and Biophysics. 1974;160(2):495–503. pmid:4364771
  63. 63. Chang H-C, Huang C-C, Huang C-J, Cheng J-S, Liu S-I, Tsai J-Y, et al. Desipramine-induced apoptosis in human PC3 prostate cancer cells: activation of JNK kinase and caspase-3 pathways and a protective role of [Ca2+] i elevation. Toxicology. 2008;250(1):9–14. pmid:18606486
  64. 64. Akalın PK. Introduction to bioinformatics. Molecular nutrition & food research. 2006;50(7):610–9. pmid:16810733