Abstract

Objective: Excessive oxidative stress is implicated in spleen injury. Platelet-rich plasma (PRP) and quercetin (QUR) have been shown to protect cells against oxidative stress. This study was designed to investigate their effect on dimethyl nitrosamine (DMN) induced spleen injury in male rats.
 Methods: Forty male Wistar rats were divided into four groups; Group (1): Negative control group (Con), Group (2): DMN group, DMN was given intraperitonealy at a dose of 4 mg/kg b. wt/day for four weeks for sub-chronic injury of spleen tissue, Group (3): DMN+PRP, rats were injected intraperitonealy with DMN at a dose of 4 mg/kg b. wt/day for four weeks then treated i. v. by single dose 50 μL of PRP, then left for a period of four weeks without any treatments, Group(4): DMN+QUR, rats received intraperitonealy DMN at a dose of 4 mg/kg b. wt/day for four weeks, then treated with quercetin orally at a dose of 50 mg/kg b. wt. in aqueous suspension daily using an intragastric tube for four weeks.
 Results: DMN inoculation resulted in significant elevations of oxidative stress, as evidenced by the increased malondialdehyde, hydrogen peroxide and xanthine oxidase levels associated with a significant decrease in Superoxide dismutase and catalase activities in the spleen tissue as compared to the normal control group. Moreover, DMN caused an up-regulation in the values of the splenic C-reactive protein (CRP), interleuckin-6 (IL-6), nuclear factor kappa B (NF-κB), leukotriene-C4 (LT-C4), P53 and Fas levels with a significant decline in anti-apoptotic protein B-cell lymphoma 2 level as compared to the normal control group. PRP and QUR significantly attenuated the DMN-evoked spleen oxidative stress and modulated the activities of antioxidant enzymes as compared to DMN group. In addition, treatment of DMN group with PRP or QUR resulted in an improvement in CRP, IL-6, NF-κB, LT-C4, P53 and Fas levels as compared to DMN group. Caspase-3 expression was positive in DMN group while no difference was present in control, PRP and Quercetin groups. However, the VEGF immunopositive reaction was found in DMN, PRP and Quercetin groups compared to control group. Histopathological results showed degeneration, haemorrhage, inflammatory cells and necrotoic areas in splenic tissue from DMN group compared to the treated groups where signs of recovery were observed in the whole splenic tissue.
 Conclusion: These data suggest that PRP and QUR protect rat spleen from DMN-induced oxidative stress, probably via their antioxidant activity, anti-inflammatory and anti-apoptotic effects. So, PRP and QUR are promising pharmacological agents for preventing the potential spleen injury of DMN following occupational or environmental exposures.

Highlights

  • Dimethylnitrosamine (DMN) could be formed during food processing, preservation, and/or preparation from precursor compounds already present in, or added to, the specific food items and causes acute and sub-chronic hepatotoxic and nephron-toxicity effects [1,2,3]

  • Various biological processes involving oxidative stress, apoptosis and inflammation related to DMN induction have been found [6], which can be controlled by various pathways

  • The protective effects of Platelet-rich plasma (PRP) or QUR against DMN-induced acute spleen injury in rats were investigated, and the results showed that they significantly improved DMN-induced acute spleen injury, which indicated that PRP or QUR may be potent candidates for the treatment of toxin-induced acute spleen injury

Read more

Summary

Introduction

Dimethylnitrosamine (DMN) could be formed during food processing, preservation, and/or preparation from precursor compounds already present in, or added to, the specific food items and causes acute and sub-chronic hepatotoxic and nephron-toxicity effects [1,2,3]. Various biological processes involving oxidative stress, apoptosis and inflammation related to DMN induction have been found [6], which can be controlled by various pathways. Reactive oxygen species (ROS) are detrimental to cells, primarily due to the damaging effect they exert on lipids, proteins and nucleic acids, which lead to structural and functional destruction [7, 8]. ROS give rise to liver destruction via lipid peroxidation, causing suppression of mitochondrial and peroxisomal b-oxidation enzymes, leading in turn to an accumulation of fatty acids in the hepatocytes, causing hepatic steatosis [9]. Various interventions have been recommended to counteract the effects of ROS by boosting the antioxidant defense systems

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