[HTML][HTML] Complement C3 deficiency attenuates chronic hypoxia-induced pulmonary hypertension in mice

EM Bauer, H Zheng, S Comhair, S Erzurum, TR Billiar… - PloS one, 2011 - journals.plos.org
EM Bauer, H Zheng, S Comhair, S Erzurum, TR Billiar, PM Bauer
PloS one, 2011journals.plos.org
Background Evidence suggests a role of both innate and adaptive immunity in the
development of pulmonary arterial hypertension. The complement system is a key sentry of
the innate immune system and bridges innate and adaptive immunity. To date there are no
studies addressing a role for the complement system in pulmonary arterial hypertension.
Methodology/Principal Findings Immunofluorescent staining revealed significant C3d
deposition in lung sections from IPAH patients and C57Bl6/J wild-type mice exposed to …
Background
Evidence suggests a role of both innate and adaptive immunity in the development of pulmonary arterial hypertension. The complement system is a key sentry of the innate immune system and bridges innate and adaptive immunity. To date there are no studies addressing a role for the complement system in pulmonary arterial hypertension.
Methodology/Principal Findings
Immunofluorescent staining revealed significant C3d deposition in lung sections from IPAH patients and C57Bl6/J wild-type mice exposed to three weeks of chronic hypoxia to induce pulmonary hypertension. Right ventricular systolic pressure and right ventricular hypertrophy were increased in hypoxic vs. normoxic wild-type mice, which were attenuated in C3−/− hypoxic mice. Likewise, pulmonary vascular remodeling was attenuated in the C3−/− mice compared to wild-type mice as determined by the number of muscularized peripheral arterioles and morphometric analysis of vessel wall thickness. The loss of C3 attenuated the increase in interleukin-6 and intracellular adhesion molecule-1 expression in response to chronic hypoxia, but not endothelin-1 levels. In wild-type mice, but not C3−/− mice, chronic hypoxia led to platelet activation as assessed by bleeding time, and flow cytometry of platelets to determine cell surface P-selectin expression. In addition, tissue factor expression and fibrin deposition were increased in the lungs of WT mice in response to chronic hypoxia. These pro-thrombotic effects of hypoxia were abrogated in C3−/− mice.
Conclusions
Herein, we provide compelling genetic evidence that the complement system plays a pathophysiologic role in the development of PAH in mice, promoting pulmonary vascular remodeling and a pro-thrombotic phenotype. In addition we demonstrate C3d deposition in IPAH patients suggesting that complement activation plays a role in the development of PAH in humans.
PLOS