Abstract

Simple SummaryLung cancer is the leading cause of cancer death among both men and women, partly due to limited therapy responses. New avenues of knowledge are indicating that lung cancer cells do not form a tumor in isolation but rather obtain essential support from their surrounding host tissue rich in altered fibroblasts. Notably, there is growing evidence that tumor progression and even the current limited responses to therapies could be prevented by rescuing the normal behavior of fibroblasts, which are critical housekeepers of normal tissue function. For this purpose, it is key to improve our understanding of the molecular mechanisms driving the pathologic alterations of fibroblasts in cancer. This work provides a comprehensive review of the main molecular mechanisms involved in fibroblast transformation based on epigenetic reprogramming, and summarizes emerging therapeutic approaches to prevent or overcome the pathologic effects of tumor-associated fibroblasts. Lung cancer is the leading cause of cancer-related death worldwide. The desmoplastic stroma of lung cancer and other solid tumors is rich in tumor-associated fibroblasts (TAFs) exhibiting an activated/myofibroblast-like phenotype. There is growing awareness that TAFs support key steps of tumor progression and are epigenetically reprogrammed compared to healthy fibroblasts. Although the mechanisms underlying such epigenetic reprogramming are incompletely understood, there is increasing evidence that they involve interactions with either cancer cells, pro-fibrotic cytokines such as TGF-β, the stiffening of the surrounding extracellular matrix, smoking cigarette particles and other environmental cues. These aberrant interactions elicit a global DNA hypomethylation and a selective transcriptional repression through hypermethylation of the TGF-β transcription factor SMAD3 in lung TAFs. Likewise, similar DNA methylation changes have been reported in TAFs from other cancer types, as well as histone core modifications and altered microRNA expression. In this review we summarize the evidence of the epigenetic reprogramming of TAFs, how this reprogramming contributes to the acquisition and maintenance of a tumor-promoting phenotype, and how it provides novel venues for therapeutic intervention, with a special focus on lung TAFs.

Highlights

  • The most frequent lung cancer subtype, and it is subdivided into adenocarcinoma (ADC, which typically arises in distal pulmonary sites and is common in non-smokers), squamous cell carcinoma (SCC, which is frequently found in proximal airways and is strongly associated with smoking), and other less frequent subtypes [1]

  • We subsequently showed that the larger epigenetic repression of the important pro-fibrotic transcription factor SMAD3 in SCC-tumorassociated fibroblasts (TAFs) compared to ADC-TAFs elicited a compensatory increase in the activity of its closely related homolog SMAD2, which is not pro-fibrotic, and how this epigenetic control of the SMAD3/SMAD2 balance is a key process underlying the lack of therapeutic effects of nintedanib in SCC in vitro and in vivo [19]

  • There is growing evidence that TAFs are epigenetically reprogrammed, as illustrated by the observation of global DNA hypomethylation concomitantly with hypermethylation of selective genes such as SMAD3, histone core modifications driven by N-methyltransferases, and the dysregulation of numerous miRNAs

Read more

Summary

Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide, and smoking and aging are major risk factors [1,2]. The most frequent lung cancer subtype, and it is subdivided into adenocarcinoma (ADC, which typically arises in distal pulmonary sites and is common in non-smokers), squamous cell carcinoma (SCC, which is frequently found in proximal airways and is strongly associated with smoking), and other less frequent subtypes [1]. All these subtypes are epithelial in origin, it is clear that the interplay between carcinoma cells and their surrounding desmoplastic stroma is essential for cancer establishment and progression [3–5]. We summarize recent evidence of major reported epigenetic changes in TAFs

DNA Methylation Changes in TAFs
Histone Core Modifications in TAFs
Non-Coding RNA Alterations in TAFs
Epigenetics of TAF Activation
Histone Core Modifications and DNMTs in Fibroblast Activation
MiRNAs in Fibroblast Activation
Emerging Mechanisms Underlying the Epigenetic Reprogramming of Lung TAFs
Crosstalk with Cancer Cells
Extracellular and Intracellular Mechanical Cues
Smoking and Other Environmental Factors
Hypoxia
Potential Therapeutic Implications
Limitations of Antifibrotic Drugs in Lung Cancer
Drugs That Alter Histone Marks in Fibroblasts
Drugs That Modify DNA Methylation Marks in Fibroblasts and Mesenchymal Cells
Drugs of Nuclear Vitamin D Receptor
Findings
Conclusions
Full Text
Published version (Free)

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