Malaria remains one of the most important infectious diseases in the world today. In 2013 the WHO estimated that 195 million individuals were infected with malaria. Annually, malaria causes 0.5-1.2 million deaths, mostly children. To date, although promising, the best malaria vaccine trials have demonstrated limited efficacy, and while combination therapy and insecticide-treated bed nets have clearly reduced the global burden of malaria, much more needs to be done. The pathogenesis of malaria is intimately linked to the innate immune response which: 1) limits parasite reproduction, 2) is required for the proper processing of antigen in order to achieve acquired immunity and, 3) can be the cause of severe acidosis, life-threatening anemia and cerebral disease that causes death in patients. This latter aspect of the innate immune response represents a real threat to human patients. Malaria parasites elicit inflammatory responses by engaging germ-line encoded innate immune receptors expressed on host leukocytes and tissue cells. Recently we have found that malaria also elicits a type I interferon (IFN) response in patient blood leukocytes. Two parasite products, DNA and hemozoin, disproportionately affect the innate immune response. Hemozoin is very important both because it helps traffic parasite DNA through the host phagolysosomal compartment (where TLR9 is engaged), and then into the cytosol because of its effect on phagolysosomal membrane stability. Furthermore, plasmodial DNA is a potent trigger for IFN production. We hypothesize that malarial infection triggers the cytosolic DNA sensor cyclic-GMP-AMP-synthase (cGAS) and induces production of 2'3'-cGAMP, a second messenger for STimulator of Interferon Genes (STING), leading to type I IFN production. Type I IFNs induce the expression of hundreds of IFN-responsive genes and the strong IFN gene signature in the blood of malaria patients may contribute to the pathogenesis during malarial infection by reprogramming the innate immune effector function of phagocytes and other innate immune cells. Our proposed studies will better define the mechanisms by which malaria drives IFN by determining if cGAS is involved in human disease, and may lead to novel therapeutic strategies.

Public Health Relevance

Every year, malaria causes somewhere between a half million and 1.2 million deaths worldwide, mostly in children; thus, identifying effective strategie to prevent and treat malaria is a global priority. Malaria is caused by the pathogen Plasmodium and a better understanding of how our immune system responds to Plasmodium infection is critical to develop better treatments and vaccines. Our proposed studies will examine how human immune cells recognize and respond to Plasmodium infection and identify potential drug targets to combat this devastating disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21AI124171-02
Application #
9264979
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Pesce, John T
Project Start
2016-04-21
Project End
2018-03-31
Budget Start
2017-04-01
Budget End
2018-03-31
Support Year
2
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Massachusetts Medical School Worcester
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
01655
Gallego-Marin, Carolina; Schrum, Jacob E; Andrade, Warrison A et al. (2018) Cyclic GMP-AMP Synthase Is the Cytosolic Sensor of Plasmodium falciparum Genomic DNA and Activates Type I IFN in Malaria. J Immunol 200:768-774
Mera-Ramírez, Angélica; Castillo, Andrés; Orobio, Yenifer et al. (2017) Screening of TNF?, IL-10 and TLR4 single nucleotide polymorphisms in individuals with asymptomatic and chronic cutaneous leishmaniasis in Colombia: a pilot study. BMC Infect Dis 17:177
Andrade, Warrison A; Agarwal, Sarika; Mo, Shunyan et al. (2016) Type I Interferon Induction by Neisseria gonorrhoeae: Dual Requirement of Cyclic GMP-AMP Synthase and Toll-like Receptor 4. Cell Rep 15:2438-48
Andrade, Warrison A; Firon, Arnaud; Schmidt, Tobias et al. (2016) Group B Streptococcus Degrades Cyclic-di-AMP to Modulate STING-Dependent Type I Interferon Production. Cell Host Microbe 20:49-59