Unknown

Dataset Information

0

Complement therapeutics meets nanomedicine: overcoming human complement activation and leukocyte uptake of nanomedicines with soluble domains of CD55.


ABSTRACT: Complement activation plays an important role in pharmacokinetic and performance of intravenously administered nanomedicines. Significant efforts have been directed toward engineering of nanosurfaces with low complement activation, but due to promiscuity of complement factors and redundancy of pathways, it is still a major challenge. Cell membrane-anchored Decay Accelerating Factor (DAF, a.k.a. CD55) is an efficient membrane bound complement regulator that inhibits both classical and alternative C3 convertases by accelerating their spontaneous decay. Here we tested the effect of various short consensus repeats (SCRs, "sushi" domains) of human CD55 on nanoparticle-mediated complement activation in human sera and plasma. Structural modeling suggested that SCR-2, SCR-3 and SCR-4 are critical for binding to the alternative pathway C3bBb convertase, whereas SCR-1 is dispensable. Various domains were expressed in E.coli and purified by an affinity column. SCRs were added to lepirudin plasma or sera from different healthy subjects, to monitor nanoparticle-mediated complement activation as well as C3 opsonization. Using superparamagnetic iron oxide nanoworms (SPIO NWs), we found that SCR-2-3-4 was the most effective inhibitor (IC50 ~0.24??M for C3 opsonization in sera), followed by SCR-1-2-3-4 (IC50 ~0.6??M), whereas shorter domains (SCR-3, SCR-2-3, SCR-3-4) were ineffective. SCR-2-3-4 also inhibited C5a generation (IC50 ~0.16??M in sera). In addition to SPIO NWs, SCR-2-3-4 effectively inhibited C3 opsonisation and C5a production by clinically approved nanoparticles (Feraheme, LipoDox and Onivyde). SCR-2-3-4 inhibited both lectin and alternative pathway activation by nanoparticles. When added to lepirudin-anticoagulated blood from healthy donors, it significantly reduced the uptake of SPIO NWs by neutrophils and monocytes. These results suggest that soluble domains of membrane-bound complement inhibitors are potential candidates for preventing nanomedicine-mediated complement activation in human subjects.

SUBMITTER: Gifford G 

PROVIDER: S-EPMC6684249 | biostudies-literature | 2019 May

REPOSITORIES: biostudies-literature

altmetric image

Publications

Complement therapeutics meets nanomedicine: overcoming human complement activation and leukocyte uptake of nanomedicines with soluble domains of CD55.

Gifford Geoffrey G   Vu Vivian P VP   Banda Nirmal K NK   Holers V Michael VM   Wang Guankui G   Groman Ernest V EV   Backos Donald D   Scheinman Robert R   Moghimi S Moein SM   Simberg Dmitri D  

Journal of controlled release : official journal of the Controlled Release Society 20190408


Complement activation plays an important role in pharmacokinetic and performance of intravenously administered nanomedicines. Significant efforts have been directed toward engineering of nanosurfaces with low complement activation, but due to promiscuity of complement factors and redundancy of pathways, it is still a major challenge. Cell membrane-anchored Decay Accelerating Factor (DAF, a.k.a. CD55) is an efficient membrane bound complement regulator that inhibits both classical and alternative  ...[more]

Similar Datasets

| S-EPMC7282746 | biostudies-literature
| S-EPMC4505834 | biostudies-literature
| S-EPMC5224875 | biostudies-literature
| S-EPMC3466049 | biostudies-literature
| S-EPMC5805379 | biostudies-literature
| S-EPMC7528717 | biostudies-literature
| S-EPMC6248342 | biostudies-literature
| S-EPMC4658209 | biostudies-literature
| S-EPMC4976035 | biostudies-literature
| S-EPMC6238980 | biostudies-literature