Elsevier

Autoimmunity Reviews

Volume 13, Issue 10, October 2014, Pages 1020-1025
Autoimmunity Reviews

Review
Reviews and prospectives of signaling pathway analysis in idiopathic pulmonary fibrosis

https://doi.org/10.1016/j.autrev.2014.08.028Get rights and content

Abstract

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing disease with disappointing survival rate, and uneffective therapeutic progress has been made in the last few years, forcing the urgent need to improve research to this disease. The commonly accepted pathogenic hypothesis of IPF is the trigger from continuous alveolar epithelium microinjuries and in the following series events, many signaling pathways were reported to lead to abnormal tissue repair and lung structure derangement in IPF, such as TGF-β, wnt, VEGF and PI3K–Akt signaling pathways. Traditional research of IPF related signaling pathway always focus on the independent function of pathway and disease signals, but the crosstalks and interactions among them were rarely valued. In this review, we summarize the signaling pathways which were reported to play important roles in the pathologic changes of IPF and the synergistic effect among those pathways. Next we discuss the application of genomics research and bioinformatics tools on IPF related pathway analysis, and give a systems biology perspective by integrating multi-level disease related data. The novel prospective of pathway analysis could tease out the complex pathway interaction profiles of IPF, and is powerful to detect IPF related biomarkers for early diagnose and potential therapeutic targets.

Introduction

IPF is one of the most pernicious and enigmatic form of lung fibrogenesis, the average survival time is about two to three years after diagnosis in affected patients [1]. Although some autoimmune diseases (e.g. rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis), genetic variants (e.g. MUC5B, VEGFA and TGF-b) and environment factors (e.g. viral infection, aging) have been reported to be risk factors of IPF, but the pathogenesis of this disorder is not fully understood [2].

Previous studies reported that IPF was characterized by continuous alveolar epithelium microinjuries and hyperplasia [3]. Injured epithelium could release growth factors, cytokines and matrix metalloproteinase, which caused the activation/proliferation of mesenchymal cell, deposition of extracellular matrix and the accumulation of fibroblasts. Next it may lead to basal membrane disruption, fibrin formation, abnormal wound repair and angiogenesis [4].

During those processes, several signaling pathways were activated to transmit important regulation signals to cell apoptosis, survival, migration or communication (see Table 1), such as transforming growth factor-beta (TGF-β) [5], wnt [6], vascular endothelial growth factor (VEGF) [7] and phosphoinositide 3-kinase (PI3K–Akt) [8], [9]. Therefore, many attentions had focused on the functions of disease related signaling pathways in IPF pathological research [9], [10]. Here, we summarized the signaling pathways which were reported to be key players in the pathogenesis of fibrosis, including the activation, hyperplasia and accumulation of myofibroblasts, and discussed the potential utility of signal molecules as future treatment targets in IPF.

However, along with the development of experiment technology and especially the implementation of many whole genome sequencing project, the number of disease related genomic data is rapidly increasing [11], [12], [13]. From the aspect of pathway research, the number of disease related signals is generating and the function of disease related signaling pathways is being comprehensive [14]. Whereas traditional pathway analysis has worked on the independent pathogenetic principles of signaling pathway and the independent function of related signal molecules, the crosstalk and interaction between pathways were rarely analyzed [15]. In this review, we discuss the novel integrative analysis prospective from systematic biological approach. In order to better clarify the associations between different signal molecules, and to comprehensively tease out the functional pathway profiles involved in the development and progression of IPF, the integrative analysis of genomic data is powerful and imperative. In the mean time, the comprehensive mapping of the signaling pathways in IPF would be powerful to detect novel early diagnosis marker and therapeutic targets, and give new research perspective on IPF research.

Section snippets

Activated signaling pathways involved in the pathology changes of IPF

In the early pathological alteration of IPF, excessive accumulation of fibroblast and extracellular matrix play key roles in the formation of fibrotic lesion, followed by the formation of a scar [16], [17]. Fibroblastic foci are regions located immediately adjacent to regions of hyperplastic or apoptotic alveolar epithelial cells. Epithelial microinjuries trigger abnormal epithelial–mesenchymal interactions and pathogenesis of idiopathic pulmonary fibrosis. Current phenomenon indicated that the

Pathway analysis of pulmonary fibrosis

Despite the known pathway data involved in the pathology of idiopathic pulmonary fibrosis which were obtained from recently publications, huge amounts of high-throughput information are produced in post-genomic era and the pool of omics data is rapidly growing. Integrated analysis of multiple types of omics data, such as metabolomics, proteomics, transcriptomics, and genomics, yields more information on the biological process than analysis of single type of signaling pathway. Rheumatologists

Integrative analysis of pathways is urgent for IPF

It is noteworthy that integrated analysis of functional genomics and cellular/molecular immunology is the general trend of polygenic diseases, especially on disease related pathway analysis [82], [83], [84]. Despite screening the entire human genome in patients, much more information is needed to tease out the functional profile of genes involved in the development and progression of IPF. During disease related pathway analysis, considering multilevel data from Gene ontology analysis,

Conclusion

With the increase of the experiment and biology data, massive amounts of disease susceptibility genes and mutation sites are explored, and also, IPF related signaling pathways are relatively richer. Systematically combine the wealth of biological resources from multiple levels, and integrated analysis those omics data with signaling pathways, would contribute to the construction of a more comprehensive IPF related signaling pathway database. From the IPF signaling pathway resource, researchers

Take-home messages

  • Many signaling pathways play key roles in the pathogenesis of IPF.

  • They regulate the proliferation, differentiation, apoptosis and migration of cells, which contained the epithelium, myofibroblasts and fibroblasts.

  • Crosstalks and interactions among different signaling pathways are being emphasized.

  • Integrating biological data from functional genomics and cellular/molecular immunology give a wealth of signaling pathway database.

  • Systematically analyzing signaling pathways with the massive and

Acknowledgements

This study was supported by grants from the Key Program of National Science and Technology Major Project of China (No. 2012ZX09103301-026) and the National Natural Science Foundation of China (No. 81030058).

References (87)

  • M. Chilosi et al.

    Aberrant Wnt/beta-catenin pathway activation in idiopathic pulmonary fibrosis

    Am J Pathol

    (05-01-2003)
  • F. Caraci et al.

    TGF-beta1 targets the GSK-3beta/beta-catenin pathway via ERK activation in the transition of human lung fibroblasts into myofibroblasts

    Pharmacol Res

    (04-01-2008)
  • D.D. Schlaepfer et al.

    Signaling through focal adhesion kinase

    Prog Biophys Mol Biol

    (01-19-1999)
  • X. Zhao et al.

    Focal adhesion kinase and its signaling pathways in cell migration and angiogenesis

    Adv Drug Deliv Rev

    (07-18-2011)
  • Q. Ding et al.

    FAK-related nonkinase is a multifunctional negative regulator of pulmonary fibrosis

    Am J Pathol

    (05-01-2013)
  • J. Tang et al.

    Vascular endothelial growth factor promotes cardiac stem cell migration via the PI3K/Akt pathway

    Exp Cell Res

    (12-10-2009)
  • T. Kobayashi et al.

    Smad3 mediates TGF-beta1 induction of VEGF production in lung fibroblasts

    Biochem Biophys Res Commun

    (02-11-2005)
  • R.L. Clifford et al.

    Novel regulation of vascular endothelial growth factor-A (VEGF-A) by transforming growth factor (beta)1: requirement for Smads, (beta)-CATENIN, AND GSK3(beta)

    J Biol Chem

    (12-19-2008)
  • J.M. Tang et al.

    Phosphorylated Akt overexpression and loss of PTEN expression in non-small cell lung cancer confers poor prognosis

    Lung Cancer-J Iaslc

    (02-01-2006)
  • H. Xia et al.

    Pathologic caveolin-1 regulation of PTEN in idiopathic pulmonary fibrosis

    Am J Pathol

    (06-01-2010)
  • R.S. Nho et al.

    PTEN regulates fibroblast elimination during collagen matrix contraction

    J Biol Chem

    (11-03-2006)
  • R.S. Nho et al.

    Pathological alteration of FoxO3a activity promotes idiopathic pulmonary fibrosis fibroblast proliferation on type i collagen matrix

    Am J Pathol

    (11-01-2011)
  • K. Shi et al.

    Pathogenesis pathways of idiopathic pulmonary fibrosis in bleomycin-induced lung injury model in mice

    Respir Physiol Neurobiol

    (01-01-2014)
  • J. Eichner et al.

    Integrated enrichment analysis and pathway-centered visualization of metabolomics, proteomics, transcriptomics, and genomics data by using the InCroMAP software

    J Chromatogr B Analyt Technol Biomed Life Sci

    (2014)
  • R. Priori et al.

    Metabolomics in rheumatic diseases: the potential of an emerging methodology for improved patient diagnosis, prognosis, and treatment efficacy

    Autoimmun Rev

    (08-01-2013)
  • L. Zhu et al.

    Tollip, an intracellular trafficking protein, is a novel modulator of the transforming growth factor-beta signaling pathway

    J Biol Chem

    (11-16-2012)
  • E.D. Litwack et al.

    Identification and characterization of two novel brain-derived immunoglobulin superfamily members with a unique structural organization

    Mol Cell Neurosci

    (02-01-2004)
  • M. Mura et al.

    Predicting survival in newly diagnosed idiopathic pulmonary fibrosis: a 3-year prospective study

    Eur Respir J

    (07-01-2012)
  • M. Gharaee-Kermani et al.

    New insights into the pathogenesis and treatment of idiopathic pulmonary fibrosis: a potential role for stem cells in the lung parenchyma and implications for therapy

    Pharm Res

    (05-01-2007)
  • M. Selman et al.

    Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy

    Ann Intern Med

    (01-16-2001)
  • M. Selman et al.

    Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-talk disorder

    Respir Res

    (01-20-2002)
  • I.E. Fernandez et al.

    The impact of TGF-beta on lung fibrosis: from targeting to biomarkers

    Proc Am Thorac Soc

    (07-01-2012)
  • K.M. Antoniou et al.

    Expression analysis of Akt and MAPK signaling pathways in lung tissue of patients with idiopathic pulmonary fibrosis (IPF)

    J Recept Signal Transduct Res

    (08-01-2010)
  • W.D. Hardie et al.

    Signaling pathways in the epithelial origins of pulmonary fibrosis

    Cell Cycle

    (07-15-2010)
  • D.J. Kass et al.

    Evolving genomic approaches to idiopathic pulmonary fibrosis: moving beyond genes

    Clin Transl Sci

    (10-01-2011)
  • T. Nance et al.

    Transcriptome analysis reveals differential splicing events in IPF lung tissue

    PLoS One

    (01-20-2014)
  • K.S. Lee et al.

    Inhibition of VEGF blocks TGF-beta1 production through a PI3K/Akt signalling pathway

    Eur Respir J

    (03-01-2008)
  • E.B. Meltzer et al.

    Idiopathic pulmonary fibrosis

    Orphanet J Rare Dis

    (01-20-2008)
  • R. Abe et al.

    Peripheral blood fibrocytes: differentiation pathway and migration to wound sites

    J Immunol

    (06-15-2001)
  • B.C. Willis et al.

    TGF-beta-induced EMT: mechanisms and implications for fibrotic lung disease

    Am J Physiol Lung Cell Mol Physiol

    (09-01-2007)
  • Z. Wu et al.

    Detection of epithelial to mesenchymal transition in airways of a bleomycin induced pulmonary fibrosis model derived from an alpha-smooth muscle actin-Cre transgenic mouse

    Respir Res

    (01-20-2007)
  • K.K. Kim et al.

    Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix

    Proc Natl Acad Sci U S A

    (08-29-2006)
  • H. Kasai et al.

    TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT)

    Respir Res

    (01-20-2005)
  • Cited by (69)

    • Thymol protects against bleomycin-induced pulmonary fibrosis via abrogation of oxidative stress, inflammation, and modulation of miR-29a/TGF-β and PI3K/Akt signaling in mice

      2023, Life Sciences
      Citation Excerpt :

      Importantly, THY previously targeted this pathway to prevent the cellular proliferation and invasion of bladder cancer and colorectal adenocarcinoma [54,55]. In addition to the prominent role of PI3K/Akt pathway in the fibrosis process, particularly in IPF, other molecular signaling components such as TGF-β, Wnt/β-catenin, and vascular endothelial growth factor (VEGF) were found to be involved in fibrosis [56]. Remarkably, THY effectively targeted those critical pathways in different disease models.

    View all citing articles on Scopus
    View full text