Abstract

Decreased inflammatory status has been reported in subjects with mild unconjugated hyperbilirubinemia. However, mechanisms of the anti-inflammatory actions of bilirubin (BR) are not fully understood. The aim of this study is to assess the role of BR in systemic inflammation using hyperbilirubinemic Gunn rats as well as their normobilirubinemic littermates and further in primary hepatocytes. The rats were treated with lipopolysaccharide (LPS, 6 mg/kg intraperitoneally) for 12 h, their blood and liver were collected for analyses of inflammatory and hepatic injury markers. Primary hepatocytes were treated with BR and TNF-α. LPS-treated Gunn rats had a significantly decreased inflammatory response, as evidenced by the anti-inflammatory profile of white blood cell subsets, and lower hepatic and systemic expressions of IL-6, TNF-α, IL-1β, and IL-10. Hepatic mRNA expression of LPS-binding protein was upregulated in Gunn rats before and after LPS treatment. In addition, liver injury markers were lower in Gunn rats as compared to in LPS-treated controls. The exposure of primary hepatocytes to TNF-α with BR led to a milder decrease in phosphorylation of the NF-κB p65 subunit compared to in cells without BR. In conclusion, hyperbilirubinemia in Gunn rats is associated with an attenuated systemic inflammatory response and decreased liver damage upon exposure to LPS.

Highlights

  • Bilirubin (BR), the end product of the heme degradation pathway in the intravascular compartment, is an important endogenous antioxidant, and it plays a crucial role in protection against oxidative stress as has been demonstrated in numerous in vitro, in vivo, and clinical studies

  • NF-κB is activated by many signals including bacterial LPS, which binds to the LPS-binding protein (LBP), and interacts with Toll-like receptor 4 (TLR4)/cluster of differentiation 14 (CD14) receptors [13]

  • Higher white blood cell (WBC) counts were observed after LPS treatment in control rats as compared to in hyperbilirubinemic Gunn animals ((12.39 ± 5.26) × 109/L vs. (8.70 ± 1.94) × 109/L, p = 0.05)

Read more

Summary

Introduction

Bilirubin (BR), the end product of the heme degradation pathway in the intravascular compartment, is an important endogenous antioxidant, and it plays a crucial role in protection against oxidative stress as has been demonstrated in numerous in vitro, in vivo, and clinical studies (for review, see [1]). These, predominantly suppressing activities, are aimed against: the complement system [4], damage-associated molecular patterns (DAMPs) signaling [5], Toll-like receptors (TLRs), such as TLR4 (a bacterial lipopolysaccharide (LPS) receptor) [6], macrophage activities [7] as well as B cell-mediated antibody production [8], and differentiation of T cells, including regulatory T cells (Tregs) [9], all with wide-spread potential clinical consequences towards autoimmune diseases [10] and transplant medicine [5,9]. Taking into consideration the reported inhibitory effects of BR on protein phosphorylation [15] as well as its general immune system-suppressing activities [10], we hypothesize that BR might interfere with phosphorylation of NF-κB p65 subunit, and prevent translocation of NF-κB into the nucleus

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