Abstract

Doxorubicin is widely used in the treatment of different cancers, and its side effects can be severe in many tissues, including the intestines. Symptoms such as diarrhoea and abdominal pain caused by intestinal inflammation lead to the interruption of chemotherapy. Nevertheless, the molecular mechanisms associated with doxorubicin intestinal toxicity have been poorly explored. This study aims to investigate such mechanisms by exposing 3D small intestine and colon organoids to doxorubicin and to evaluate transcriptomic responses in relation to viability and apoptosis as physiological endpoints. The in vitro concentrations and dosing regimens of doxorubicin were selected based on physiologically based pharmacokinetic model simulations of treatment regimens recommended for cancer patients. Cytotoxicity and cell morphology were evaluated as well as gene expression and biological pathways affected by doxorubicin. In both types of organoids, cell cycle, the p53 signalling pathway, and oxidative stress were the most affected pathways. However, significant differences between colon and SI organoids were evident, particularly in essential metabolic pathways. Short time-series expression miner was used to further explore temporal changes in gene profiles, which identified distinct tissue responses. Finally, in silico proteomics revealed important proteins involved in doxorubicin metabolism and cellular processes that were in line with the transcriptomic responses, including cell cycle and senescence, transport of molecules, and mitochondria impairment. This study provides new insight into doxorubicin-induced effects on the gene expression levels in the intestines. Currently, we are exploring the potential use of these data in establishing quantitative systems toxicology models for the prediction of drug-induced gastrointestinal toxicity.

Highlights

  • Doxorubicin (DOX) is a chemotherapeutical drug that belongs to the class of anthracyclines, being first isolated from Streptomyces peucetius var. caesius in 1967 [1]

  • In silico proteomics revealed important proteins involved in doxorubicin metabolism and cellular processes that were in line with the transcriptomic responses, including cell cycle and senescence, transport of molecules, and mitochondria impairment

  • The main goal of this study was to investigate molecular mechanisms of toxicity of DOX in 3D human organoid models of both colon and small intestine. These new and promising cell culture models have shown to be suitable for the investigation of diseases, targeted therapies, drug development and screening overcoming the limitations of the 2D systems [11,12]

Read more

Summary

Introduction

Doxorubicin (DOX) is a chemotherapeutical drug that belongs to the class of anthracyclines, being first isolated from Streptomyces peucetius var. caesius in 1967 [1]. DOX has application in the treatment of a wide range of cancers, such as solid tumours, acute myeloblastic and lymphoblastic leukaemia, breast, ovarian, prostate, gastric carcinomas, osteosarcomas, and soft tissue sarcomas [2]. Despite being one of the most standardized and recommended drugs to treat such malignancies in the last 40 years, DOX is often associated with severe side effects. DOX is associated with rather serious effects, including cardiotoxicity, leading to cardiomyopathy and subsequently congestive heart failure, hepatotoxicity, erythema, and disruption of the intestinal epithelium [2,3]. Disruption and subsequent inflammation of the intestinal epithelium induce symptoms such as diarrhoea, vomiting, abdominal pain and nausea. Cancer treatments can be compromised, dramatically impairing patient’s survival and quality of life

Objectives
Methods
Results
Conclusion
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