Caenorhabditis elegans is a powerful model system for genetics and developmental biology, and for identifying biochemical targets to develop drugs active against parasitic nematodes. Eukaryotes, including nematodes and humans, share many similar metabolic pathways, which makes specific targeting challenging. Our recent studies suggest that C. elegans and other nematodes, unlike other animals, may use a plant-like pathway as the primary biosynthetic route to the major phospholipid component phosphatidylcholine. In this metabolic pathway, a pair of phosphoethanolamine methyltransferases (PMT) catalyze the sequential methylation of phosphoethanolamine to phosphocholine, which can then be incorporated into phosphatidylcholine. Importantly, both of the PMT are required for normal growth and development of C. elegans, as RNAi knockout of their expression leads to severe developmental phenotypes including arrested growth and death. This suggests that inhibition of PMT activity may be a possible strategy for controlling nematode growth. Because the PMT are not found in mammals and are highly conserved across multiple parasitic nematodes and protozoans, these enzymes are promising targets for inhibitor development;however, very little is known about how the PMT function at the molecular level. This proposal aims to explore and characterize this new metabolic pathway in C. elegans as a possible target for the development of anti-parasitic compounds. The experiments outlined in this proposal aim to address two basic and unanswered questions - what is the molecular and chemical basis for phosphobase methylation and what is the biological role of this critical metabolic pathway in C. elegans? The planned research sets out to understand the molecular basis for how the PMT function by determining the X-ray crystal structures of these enzymes and by analyzing the kinetic and thermodynamic features that define catalysis and phosphobase substrate specificity (Aims 1 and 2). To complement in vitro studies of the PMT, we will also test the metabolic contribution of these proteins to phosphatidylcholine synthesis and determine their expression patterns (Aim 3). These in vivo experiments will expand our understanding of the essential function these proteins play in the growth and development of nematodes. Because the PMT are highly conserved across nematode parasites of humans, animals, and plants, as well as in protozoan parasites, understanding how these enzymes function and identifying inhibitors targeting them will contribute to developing new anti-parasitic compounds of potential medical, veterinary, and agricultural value.

Public Health Relevance

to public health. Parasitic nematodes are a major cause of human health problems with an estimated 3 billion people infected worldwide by these organisms. Identifying biochemical targets that differ between the parasite and host species is essential for finding effective new anti-parasitic drugs. Using the free-living nematode Caenorhabditis elegans as a model system, my group has identified a new biochemical pathway essential for worm growth and development that is not found in mammals. This proposal aims to understand the molecular function of the enzymes in the phosphobase methylation pathway and to aid in the development of inhibitors as potential anti-parasitic compounds.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI097119-02
Application #
8418701
Study Section
Macromolecular Structure and Function E Study Section (MSFE)
Program Officer
Rogers, Martin J
Project Start
2012-02-15
Project End
2016-01-31
Budget Start
2013-02-01
Budget End
2014-01-31
Support Year
2
Fiscal Year
2013
Total Cost
$214,320
Indirect Cost
$73,320
Name
Washington University
Department
Biology
Type
Schools of Arts and Sciences
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Vallone, Alessandra; D'Alessandro, Sarah; Brogi, Simone et al. (2018) Antimalarial agents against both sexual and asexual parasites stages: structure-activity relationships and biological studies of the Malaria Box compound 1-[5-(4-bromo-2-chlorophenyl)furan-2-yl]-N-[(piperidin-4-yl)methyl]methanamine (MMV019918) and analog Eur J Med Chem 150:698-718
Lee, Soon Goo; Jez, Joseph M (2017) Conformational changes in the di-domain structure of Arabidopsis phosphoethanolamine methyltransferase leads to active-site formation. J Biol Chem 292:21690-21702
Cascella, Barbara; Lee, Soon Goo; Singh, Sukrit et al. (2017) The small molecule JIB-04 disrupts O2binding in the Fe-dependent histone demethylase KDM4A/JMJD2A. Chem Commun (Camb) 53:2174-2177
Korasick, David A; Jez, Joseph M; Strader, Lucia C (2015) Refining the nuclear auxin response pathway through structural biology. Curr Opin Plant Biol 27:22-8
Saen-Oon, Suwipa; Lee, Soon Goo; Jez, Joseph M et al. (2014) An alternative mechanism for the methylation of phosphoethanolamine catalyzed by Plasmodium falciparum phosphoethanolamine methyltransferase. J Biol Chem 289:33815-25
Lee, Soon Goo; Jez, Joseph M (2014) Nematode phospholipid metabolism: an example of closing the genome-structure-function circle. Trends Parasitol 30:241-50
Lee, Soon Goo; Jez, Joseph M (2013) Evolution of structure and mechanistic divergence in di-domain methyltransferases from nematode phosphocholine biosynthesis. Structure 21:1778-87
Lee, Soon Goo; Alpert, Tara D; Jez, Joseph M (2012) Crystal structure of phosphoethanolamine methyltransferase from Plasmodium falciparum in complex with amodiaquine. Bioorg Med Chem Lett 22:4990-3
Lee, Soon Goo; Kim, Youngchang; Alpert, Tara D et al. (2012) Structure and reaction mechanism of phosphoethanolamine methyltransferase from the malaria parasite Plasmodium falciparum: an antiparasitic drug target. J Biol Chem 287:1426-34