Abstract

The main function of red blood cells (RBCs) is the transport of respiratory gases along the vascular tree. To fulfill their task, RBCs are able to elastically deform in response to mechanical forces and, pass through the narrow vessels of the microcirculation. Decreased RBC deformability was observed in pathological conditions linked to increased oxidative stress or decreased nitric oxide (NO) bioavailability, like hypertension. Treatments with oxidants and with NO were shown to affect RBC deformability ex vivo, but the mechanisms underpinning these effects are unknown. In this study we investigate whether changes in intracellular redox status/oxidative stress or nitrosation reactions induced by reactive oxygen species (ROS) or NO may affect RBC deformability. In a case-control study comparing RBCs from healthy and hypertensive participants, we found that RBC deformability was decreased, and levels of ROS were increased in RBCs from hypertensive patients as compared to RBCs from aged-matched healthy controls, while NO levels in RBCs were not significantly different. To study the effects of oxidants on RBC redox state and deformability, RBCs from healthy volunteers were treated with increasing concentrations of tert-butylhydroperoxide (t-BuOOH). We found that high concentrations of t-BuOOH (≥ 1 mM) significantly decreased the GSH/GSSG ratio in RBCs, decreased RBC deformability and increased blood bulk viscosity. Moreover, RBCs from Nrf2 knockout (KO) mice, a strain genetically deficient in a number of antioxidant/reducing enzymes, were more susceptible to t-BuOOH-induced impairment in RBC deformability as compared to wild type (WT) mice. To study the role of NO in RBC deformability we treated RBC suspensions from human volunteers with NO donors and nitrosothiols and analyzed deformability of RBCs from mice lacking the endothelial NO synthase (eNOS). We found that NO donors induced S-nitrosation of the cytoskeletal protein spectrin, but did not affect human RBC deformability or blood bulk viscosity; moreover, under unstressed conditions RBCs from eNOS KO mice showed fully preserved RBC deformability as compared to WT mice. Pre-treatment of human RBCs with nitrosothiols rescued t-BuOOH-mediated loss of RBC deformability. Taken together, these findings suggest that NO does not affect RBC deformability per se, but preserves RBC deformability in conditions of oxidative stress.

Highlights

  • The biochemical, biophysical, and mechanical properties of RBCs, as well as their structural characteristics are optimized for their function

  • RBCs were considered for a long time as simple “bags” packed with hemoglobin circulating in the blood for the only purposes of gas exchange and maintenance of acid/base equilibria; evidence is accumulating that RBC function is far more complex and highly regulated

  • In contrast to the common knowledge that nitric oxide (NO) directly affects RBC deformability, this study shows that neither eNOS-dependent NO formation nor NO donors affected RBC deformability per se; instead, we found that treatment with nitrosothiols contributes to preserve their resilience to intracellular oxidative modifications and loss of membrane flexibility

Read more

Summary

Introduction

The biochemical, biophysical, and mechanical properties of RBCs, as well as their structural characteristics are optimized for their function. RBCs carry a very high (supersaturated) concentration of hemoglobin (equivalent to 10 mM heme), which is kept in the reduced Fe2+/oxygen binding state by a battery of antioxidant and reducing enzymes (Kuhn et al, 2017). The distribution and biophysical characteristics of the cytoskeletal proteins (mainly spectrin) are responsible for their typical biconcave “donut-like” shape. RBC shape and deformability allow the cells to dynamically adapt to changes in hydrodynamic forces along the vascular tree, and to squeeze through capillaries smaller than their own diameter at rest (Kuhn et al, 2017). Reactive oxygen species (ROS)-mediated damage of RBC membrane components is thought to increase erythrocyte membrane rigidity and fragility, resulting in intravascular hemolysis, release of hemoglobin into the plasma, and systemic NO scavenging. In spite of the clinical significance of these phenomena the biological chemistry and biochemistry of the processes that control physiological RBC deformability in health and disease, and the underlying signaling pathways remain poorly characterized

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