Abstract

The origin and mechanism of vasocontraction observed after vascular exposure to acellular Hbs remain controversial. To help resolve the underlying mechanism, we characterized Hb-induced vasoactivities in terms of Hb purity, heme iron oxidation state, and ligand and pharmacodynamic properties. Isolated rat thoracic aortic rings with intact endothelium were suspended in oxygenated Krebs buffer, and isometric tension responses to various test Hb preparations were measured. In norepinephrine tone–enhanced aortic rings, both crude and purified Hbs exhibited similar dose-response characteristics; stroma-free Hb and HbA0, two Hb preparations with disparate purity, were equally potent in inducing vessel ring contraction. Purified Hb preparations significantly attenuated vasodilatory potency of both acetylcholine, an endothelium-dependent NO generator, and glyceryl trinitrate, an endothelium-independent NO generator. With the exception of nitrosylated Hb, ferrous Hbs, oxy Hb, and carbon monoxy Hb elicited contraction, whereas ferric derivatives, met Hb, and cyanomet Hb did not. In addition, NEM-Hb, an Hb with blocked cysteine residues, did not notably attenuate Hb vasoactivity. These results indicate that Hb itself is directly responsible for inducing contraction in the rat thoracic aortic rings. A primary mechanism for the Hb-induced vasoactivity appears to be heme iron inactivation of endothelium-derived NO. Nonheme interaction with endothelial NO does not appear to play a prominent role in this vascular model. In conclusion, Hb elicits dose-dependent contraction in isolated rat thoracic aorta with intact endothelium. Vasoactivity of Hbs, however, could greatly vary with heme iron oxidation state, nature of heme ligand, and model vessels used in the evaluation. (J Lab Clin Med 2000;135:180-7)

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