Abstract

The liver is one of the most sexually dimorphic organs. The hepatic metabolic pathways that are subject to sexual dimorphism include xenobiotic, amino acid and lipid metabolism. Non-alcoholic fatty liver disease and hepatocellular carcinoma are among diseases with sex-dependent prevalence, progression and outcome. Although male and female livers differ in their abilities to metabolize foreign compounds, including drugs, sex-dependent treatment and pharmacological dynamics are rarely applied in all relevant cases. Therefore, it is important to consider hepatic sexual dimorphism when developing new treatment strategies and to understand the underlying mechanisms in model systems. We isolated primary hepatocytes from male and female C57BL6/N mice and examined the sex-dependent transcriptome, proteome and extracellular metabolome parameters in the course of culturing them for 96 h. The sex-specific gene expression of the general xenobiotic pathway altered and the female-specific expression of Cyp2b13 and Cyp2b9 was significantly reduced during culture. Sex-dependent differences of several signaling pathways increased, including genes related to serotonin and melatonin degradation. Furthermore, the ratios of male and female gene expression were inversed for other pathways, such as amino acid degradation, beta-oxidation, androgen signaling and hepatic steatosis. Because the primary hepatocytes were cultivated without the influence of known regulators of sexual dimorphism, these results suggest currently unknown modulatory mechanisms of sexual dimorphism in vitro. The large sex-dependent differences in the regulation and dynamics of drug metabolism observed during cultivation can have an immense influence on the evaluation of pharmacodynamic processes when conducting initial preclinical trials to investigate potential new drugs.

Highlights

  • Over the past decade, medical research has focused on the development of patient-oriented therapies

  • There is a clear separation between the expression of female and male proteins (Fig. 1a) and genes (Fig. 1b), the differences between sexes became more diffuse in the course of cultivation

  • During further primary hepatocyte culture, the gene expression pattern almost returned to the initial level in the vertical direction, while it shifted in the horizontal direction (Fig. 1b)

Read more

Summary

Introduction

Medical research has focused on the development of patient-oriented therapies. Different approaches, such as personalized, stratified and precision medicine, based on biomedical investigations with large amounts of data, have been discussed (Erikainen and Chan 2019). The primary regulators of hepatic sexual dimorphism are testosterone and estradiol signaling as well as the sex-dependent expression of growth hormone (GH) The release of GH leads to the phosphorylation, activation and deoxyribonucleic acid (DNA) binding of signal transducer and activator of transcription 5b (STAT5b), which exhibits the same pulsatile activity pattern in males (Waxman and O’Connor 2006; Conforto et al 2012). There is a cross-talk between GH and steroid hormone regulation, which allowed the construction of mathematical models of female and male human hepatocytes (Cvitanović Tomaš et al 2018; Cvitanović et al 2017), underlining the need to treat both sexes separately

Methods
Results
Discussion
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