Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors

Introduction: C3 is central for all complement activation pathways, thus an attractive therapeutic target. Many C3-targeted agents are under extensive development with one already approved for clinical use. However, most, if not all, C3 inhibitors are human or non-human primate C3-specific, making...

Full description

Bibliographic Details
Main Authors: Jin Chen, Lingjun Zhang, Maojing Yang, Elizabeth D. Hughes, Zachary T. Freeman, Thomas L. Saunders, Feng Lin
Format: Article
Language:English
Published: Karger Publishers 2023-11-01
Series:Journal of Innate Immunity
Online Access:https://beta.karger.com/Article/FullText/534963
_version_ 1797373836298027008
author Jin Chen
Lingjun Zhang
Maojing Yang
Elizabeth D. Hughes
Zachary T. Freeman
Thomas L. Saunders
Feng Lin
author_facet Jin Chen
Lingjun Zhang
Maojing Yang
Elizabeth D. Hughes
Zachary T. Freeman
Thomas L. Saunders
Feng Lin
author_sort Jin Chen
collection DOAJ
description Introduction: C3 is central for all complement activation pathways, thus an attractive therapeutic target. Many C3-targeted agents are under extensive development with one already approved for clinical use. However, most, if not all, C3 inhibitors are human or non-human primate C3-specific, making evaluating their efficacies in vivo before a clinical trial extremely difficult and costly. Methods: We first studied the compatibility of human C3 in the rat complement system, then developed a C3 humanized rat using the CRISPR/Cas9 technology. We thoroughly characterized the resultant human C3 humanized rats and tested the treatment efficacy of an established primate-specific C3 inhibitor in a model of complement-mediated hemolysis in the C3 humanized rats. Results: We found that supplementing human C3 protein into the C3-deficient rat blood restored its complement activity, which was inhibited by rat Factor H or compstatin, , suggesting that human C3 is compatible to the rat complement system. The newly developed C3 humanized rats appeared healthy and expressed human but not rat C3 without detectable spontaneous C3 activation. More importantly, complement-mediated hemolysis in the C3 humanized rats was also inhibited by compstatin both in vitro and in vivo. Conclusion: The successfully developed C3 humanized rats provided a much-desired rodent model to evaluate novel C3 inhibitors in vivo as potential drugs.
first_indexed 2024-03-08T18:56:11Z
format Article
id doaj.art-c531a0589394406ca02d9b8770b9cade
institution Directory Open Access Journal
issn 1662-8128
language English
last_indexed 2024-03-08T18:56:11Z
publishDate 2023-11-01
publisher Karger Publishers
record_format Article
series Journal of Innate Immunity
spelling doaj.art-c531a0589394406ca02d9b8770b9cade2023-12-28T13:27:42ZengKarger PublishersJournal of Innate Immunity1662-81282023-11-011110.1159/000534963534963Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitorsJin ChenLingjun ZhangMaojing YangElizabeth D. HughesZachary T. FreemanThomas L. SaundersFeng LinIntroduction: C3 is central for all complement activation pathways, thus an attractive therapeutic target. Many C3-targeted agents are under extensive development with one already approved for clinical use. However, most, if not all, C3 inhibitors are human or non-human primate C3-specific, making evaluating their efficacies in vivo before a clinical trial extremely difficult and costly. Methods: We first studied the compatibility of human C3 in the rat complement system, then developed a C3 humanized rat using the CRISPR/Cas9 technology. We thoroughly characterized the resultant human C3 humanized rats and tested the treatment efficacy of an established primate-specific C3 inhibitor in a model of complement-mediated hemolysis in the C3 humanized rats. Results: We found that supplementing human C3 protein into the C3-deficient rat blood restored its complement activity, which was inhibited by rat Factor H or compstatin, , suggesting that human C3 is compatible to the rat complement system. The newly developed C3 humanized rats appeared healthy and expressed human but not rat C3 without detectable spontaneous C3 activation. More importantly, complement-mediated hemolysis in the C3 humanized rats was also inhibited by compstatin both in vitro and in vivo. Conclusion: The successfully developed C3 humanized rats provided a much-desired rodent model to evaluate novel C3 inhibitors in vivo as potential drugs.https://beta.karger.com/Article/FullText/534963
spellingShingle Jin Chen
Lingjun Zhang
Maojing Yang
Elizabeth D. Hughes
Zachary T. Freeman
Thomas L. Saunders
Feng Lin
Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors
Journal of Innate Immunity
title Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors
title_full Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors
title_fullStr Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors
title_full_unstemmed Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors
title_short Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors
title_sort development of a c3 humanized rat as a new model for evaluating novel c3 inhibitors
url https://beta.karger.com/Article/FullText/534963
work_keys_str_mv AT jinchen developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors
AT lingjunzhang developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors
AT maojingyang developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors
AT elizabethdhughes developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors
AT zacharytfreeman developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors
AT thomaslsaunders developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors
AT fenglin developmentofac3humanizedratasanewmodelforevaluatingnovelc3inhibitors