Haptoglobin preserves vascular nitric oxide signaling during hemolysis

CA Schaer, JW Deuel, D Schildknecht… - American journal of …, 2016 - atsjournals.org
CA Schaer, JW Deuel, D Schildknecht, L Mahmoudi, I Garcia-Rubio, C Owczarek, S Schauer…
American journal of respiratory and critical care medicine, 2016atsjournals.org
Rationale: Hemolysis occurs not only in conditions such as sickle cell disease and malaria
but also during transfusion of stored blood, extracorporeal circulation, and sepsis. Cell-free
Hb depletes nitric oxide (NO) in the vasculature, causing vasoconstriction and eventually
cardiovascular complications. We hypothesize that Hb-binding proteins may preserve
vascular NO signaling during hemolysis. Objectives: Characterization of an archetypical
function by which Hb scavenger proteins could preserve NO signaling during hemolysis …
Rationale: Hemolysis occurs not only in conditions such as sickle cell disease and malaria but also during transfusion of stored blood, extracorporeal circulation, and sepsis. Cell-free Hb depletes nitric oxide (NO) in the vasculature, causing vasoconstriction and eventually cardiovascular complications. We hypothesize that Hb-binding proteins may preserve vascular NO signaling during hemolysis.
Objectives: Characterization of an archetypical function by which Hb scavenger proteins could preserve NO signaling during hemolysis.
Methods: We investigated NO reaction kinetics, effects on arterial NO signaling, and tissue distribution of cell-free Hb and its scavenger protein complexes.
Measurements and Main Results: Extravascular translocation of cell-free Hb into interstitial spaces, including the vascular smooth muscle cell layer of rat and pig coronary arteries, promotes vascular NO resistance. This critical disease process is blocked by haptoglobin. Haptoglobin does not change NO dioxygenation rates of Hb; rather, the large size of the Hb:haptoglobin complex prevents Hb extravasation, which uncouples NO/Hb interaction and vasoconstriction. Size-selective compartmentalization of Hb functions as a substitute for red blood cells after hemolysis and preserves NO signaling in the vasculature. We found that evolutionarily and structurally unrelated Hb-binding proteins, such as PIT54 found in avian species, functionally converged with haptoglobin to protect NO signaling by sequestering cell-free Hb in large protein complexes.
Conclusions: Sequential compartmentalization of Hb by erythrocytes and scavenger protein complexes is an archetypical mechanism, which may have supported coevolution of hemolysis and normal vascular function. Therapeutic supplementation of Hb scavengers may restore vascular NO signaling and attenuate disease complications in patients with hemolysis.
ATS Journals