Abstract

Early assessment of adverse drug effects in humans is critical to avoid long-lasting harm. However, current approaches for early detection of adverse effects still lack predictive and organ-specific biomarkers to evaluate undesired responses in humans. Microphysiological systems (MPSs) are in vitro representations of human tissues and provide organ-specific translational insights for physiological processes. In this study, a brain MPS was utilized to assess molecular signatures of neurotoxic and non-neurotoxic compounds using targeted and untargeted molecular approaches. The brain MPS comprising of human embryonic stem (ES) cell-derived neural progenitor cells seeded on three-dimensional (3D), chemically defined, polyethylene glycol hydrogels was treated with the neurotoxic drug, bortezomib and the non-neurotoxic drug, tamoxifen over 14-days. Possible toxic effects were monitored with human N-acetylaspartic acid (NAA) kinetics, which correlates the neuronal function/health and DJ-1/PARK7, an oxidative stress biomarker. Changes in NAA levels were observed as early as 2-days post-bortezomib treatment, while onset detection of oxidative stress (DJ-1) was delayed until 4-days post-treatment. Separately, the untargeted extracellular metabolomics approach revealed distinct fingerprints 2-days post-bortezomib treatment as perturbations in cysteine and glycerophospholipid metabolic pathways. These results suggest accumulation of reactive oxygen species associated with oxidative stress, and disruption of membrane structure and integrity. The NAA response was strongly correlated with changes in a subset of the detected metabolites at the same time point 2-days post-treatment. Moreover, these metabolite changes correlated strongly with DJ-1 levels measured at the later time point (4-days post-treatment). This suggests that early cellular metabolic dysfunction leads to later DJ-1 leakage and cell death, and that early measurement of this subset of metabolites could predict the later occurrence of cell death. While the approach demonstrated here provides an individual case study for proof of concept, we suggest that this approach can be extended for preclinical toxicity screening and biomarker discovery studies.

Highlights

  • Given that central nervous system (CNS) toxicity is a leading cause of toxicity-related clinical trial failures (Cook et al, 2014; Walker et al, 2018), the predictive capabilities of current preclinical toxicity testing methods remain inadequate

  • The brain Microphysiological systems (MPSs) comprises a 3D Polyethylene Glycol (PEG) hydrogel with Neural progenitor cells (NPCs) that have differentiated into βIII-tubulin+ and glial fibrillary acidic protein (GFAP)+ cells and self-assembled to form 3D neural constructs

  • For 0.01 μM dose of bortezomib on day 18 (4-day treatment), NAA levels were significantly reduced compared to control, and remained so at day 22 and beyond

Read more

Summary

Introduction

Given that central nervous system (CNS) toxicity is a leading cause of toxicity-related clinical trial failures (Cook et al, 2014; Walker et al, 2018), the predictive capabilities of current preclinical toxicity testing methods remain inadequate. The twodimensional (2D) mono-cultures of typical in vitro CNS models fail to recapitulate the physiological complexity of CNS tissues, limiting their ability to predict adverse responses at the tissue or organ level (in vivo) from the effects observed at the molecular or cellular level (in vitro) (Langhans, 2018). MPSs encompass a range of cellular- and tissuelevel models in three-dimensional (3D) culture platforms meant to recapitulate more physiologically-relevant functions of human organs and tissues compared to traditional 2D culture systems. MPSs are more cost-effective than animal models and can be used for numerous pharmaceutical development applications including drug absorption, distribution, metabolism, excretion, and toxicity (ADMET), evaluating efficacy and investigating pharmacodynamic mechanisms. Drug absorption and metabolism processes have been studied in an integrated gut-liver platform that enables observation of organ-organ crosstalk (Chen et al, 2017; Tsamandouras et al, 2017). Higher degree multi-MPS systems have been applied to assess systemic drug effects on human physiology (Maschmeyer et al, 2015; Oleaga et al, 2016; Zhang et al, 2017; Edington et al, 2018)

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