Brain endothelial STING1 activation by Plasmodium-sequestered heme promotes cerebral malaria via type I IFN response

TF Pais, H Ali, J Moreira da Silva… - Proceedings of the …, 2022 - National Acad Sciences
TF Pais, H Ali, J Moreira da Silva, N Duarte, R Neres, C Chhatbar, RC Acúrcio, RC Guedes…
Proceedings of the National Academy of Sciences, 2022National Acad Sciences
Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused
by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which
is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The
molecular triggers and sensors eliciting brain type I IFN cellular responses during CM
remain largely unknown. We herein identified the stimulator of interferon response cGAMP
interactor 1 (STING1) as the key innate immune sensor that induces Ifnβ1 transcription in the …
Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The molecular triggers and sensors eliciting brain type I IFN cellular responses during CM remain largely unknown. We herein identified the stimulator of interferon response cGAMP interactor 1 (STING1) as the key innate immune sensor that induces Ifnβ1 transcription in the brain of mice infected with Plasmodium berghei ANKA (Pba). This STING1/IFNβ-mediated response increases brain CXCL10 governing the extent of brain leukocyte infiltration and blood–brain barrier (BBB) breakdown, and determining CM lethality. The critical role of brain endothelial cells (BECs) in fueling type I IFN–driven brain inflammation was demonstrated in brain endothelial–specific IFNβ-reporter and STING1-deficient Pba-infected mice, which were significantly protected from CM lethality. Moreover, extracellular particles (EPs) released from Pba-infected erythrocytes activated the STING1-dependent type I IFN response in BECs, a response requiring intracellular acidification. Fractionation of the EPs enabled us to identify a defined fraction carrying hemoglobin degradation remnants that activates STING1/IFNβ in the brain endothelium, a process correlated with heme content. Notably, stimulation of STING1-deficient BECs with heme, docking experiments, and in vitro binding assays unveiled that heme is a putative STING1 ligand. This work shows that heme resultant from the parasite heterotrophic activity operates as an alarmin, triggering brain endothelial inflammatory responses via the STING1/IFNβ/CXCL10 axis crucial to CM pathogenesis and lethality.
National Acad Sciences