Abstract

BackgroundEpigenetics has been investigated in cancer initiation, and development, especially, since the appearance of epigenomics. Epigenetics may be defined as the mechanisms that lead to heritable changes in gene function and without affecting the sequence of genome. These mechanisms explain how individuals with the same genotype produce phenotypic differences in response to environmental stimuli. Recently, with the accumulation of high-throughput next-generation sequencing (NGS) data, a key goal of systems biology is to construct networks for different cellular levels to explore whole cellular mechanisms. At present, there is no satisfactory method to construct an integrated genetic and epigenetic cellular network (IGECN), which combines NGS omics data with gene regulatory networks (GRNs), microRNAs (miRNAs) regulatory networks, protein-protein interaction networks (PPINs), and epigenetic regulatory networks of methylation using high-throughput NGS data.ResultsWe investigated different kinds of NGS omics data to develop a systems biology method to construct an integrated cellular network based on three coupling models that describe genetic regulatory networks, protein–protein interaction networks, microRNA (miRNA) regulatory networks, and methylation regulation. The proposed method was applied to construct IGECNs of gastric cancer and the human immune response to human immunodeficiency virus (HIV) infection, to elucidate human defense response mechanisms. We successfully constructed an IGECN and validated it by using evidence from literature search. The integration of NGS omics data related to transcription regulation, protein-protein interactions, and miRNA and methylation regulation has more predictive power than independent datasets. We found that dysregulation of MIR7 contributes to the initiation and progression of inflammation-induced gastric cancer; dysregulation of MIR9 contributes to HIV-1 infection to hijack CD4+ T cells through dysfunction of the immune and hormone pathways; dysregulation of MIR139-5p, MIRLET7i, and MIR10a contributes to the HIV-1 integration/replication stage; dysregulation of MIR101, MIR141, and MIR152 contributes to the HIV-1 virus assembly and budding mechanisms; dysregulation of MIR302a contributes to not only microvesicle-mediated transfer of miRNAs but also dysfunction of NF-κB signaling pathway in hepatocarcinogenesis.ConclusionThe coupling dynamic systems of the whole IGECN can allow us to investigate genetic and epigenetic cellular mechanisms via omics data and big database mining, and are useful for further experiments in the field of systems and synthetic biology.

Highlights

  • Epigenetics has been investigated in cancer initiation, and development, especially, since the appearance of epigenomics

  • Dysregulation of MIR7 contributes to the initiation and progression of inflammation-induced gastric cancer; dysregulation of MIR9 contributes to human immunodeficiency virus (HIV)-1 infection to hijack CD4+ T cells through dysfunction of the immune and hormone pathways; dysregulation of MIR139-5p, MIRLET7i, and MIR10a contributes to the HIV-1 integration/replication stage through DNA hypermethylation and immune system dysfunction; dysregulation of MIR101, MIR141, and MIR152 contributes to the HIV-1 virus assembly/budding stage through DNA hypermethylation, ubiquitin transfer, and endoplasmic reticulum-associated degradation; dysregulation of MIR302a contributes to microvesicle-mediated transfer of miRNAs and dysfunction of NF-κB signaling pathway in hepatocarcinogenesis

  • At early infection stage (Fig. 5), we identified that the expression changes of MIR9 (p-value < 0.448) and general transcription factor IIi (GTF2I) (p-value < 0.96) contribute to the expression change of small ubiquitin-like modifier 3 (SUMO3) via respectively regulating zinc finger protein 131 (ZNF131) (p-value < 9.3☓10-14) and DEAD (Asp-Glu-Ala-Asp) box helicase 3, X-linked (DDX3X) (p-value < 1☓10-16)

Read more

Summary

Introduction

Epigenetics has been investigated in cancer initiation, and development, especially, since the appearance of epigenomics. Epigenetics may be defined as the mechanisms that lead to heritable changes in gene function and without affecting the sequence of genome. With advances in molecular biology technologies for whole genome sequencing, expression profiling, and highthroughput experiments, large amounts of biological data covering various biological levels have emerged [1,2,3] These kinds of ‘omics’ data include genetic sequences (genomics), microarray-based genome-wide expression profiles (transcriptomics), protein abundance data (proteomics), and microRNA (miRNA) and methylation data, and they provide an unprecedented view of cellular components and their cellular mechanisms in biological systems [4]. An integrated cellular network of transcription regulation and PPIs was introduced by Wang and Chen [9, 10] They use gene-expression data at multiple time points to prune and combine candidate gene regulatory and signaling networks obtained from genomescale data. To explore cellular mechanism, we need more genomic data, such as epigenetic regulation data

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call