Protein kinase C-dependent signaling controls the midgut epithelial barrier to malaria parasite infection in anopheline mosquitoes

PLoS One. 2013 Oct 11;8(10):e76535. doi: 10.1371/journal.pone.0076535. eCollection 2013.

Abstract

Anopheline mosquitoes are the primary vectors of parasites in the genus Plasmodium, the causative agents of malaria. Malaria parasites undergo a series of complex transformations upon ingestion by the mosquito host. During this process, the physical barrier of the midgut epithelium, along with innate immune defenses, functionally restrict parasite development. Although these defenses have been studied for some time, the regulatory factors that control them are poorly understood. The protein kinase C (PKC) gene family consists of serine/threonine kinases that serve as central signaling molecules and regulators of a broad spectrum of cellular processes including epithelial barrier function and immunity. Indeed, PKCs are highly conserved, ranging from 7 isoforms in Drosophila to 16 isoforms in mammals, yet none have been identified in mosquitoes. Despite conservation of the PKC gene family and their potential as targets for transmission-blocking strategies for malaria, no direct connections between PKCs, the mosquito immune response or epithelial barrier integrity are known. Here, we identify and characterize six PKC gene family members--PKCδ, PKCε, PKCζ, PKD, PKN, and an indeterminate conventional PKC--in Anopheles gambiae and Anopheles stephensi. Sequence and phylogenetic analyses of the anopheline PKCs support most subfamily assignments. All six PKCs are expressed in the midgut epithelia of A. gambiae and A. stephensi post-blood feeding, indicating availability for signaling in a tissue that is critical for malaria parasite development. Although inhibition of PKC enzymatic activity decreased NF-κB-regulated anti-microbial peptide expression in mosquito cells in vitro, PKC inhibition had no effect on expression of a panel of immune genes in the midgut epithelium in vivo. PKC inhibition did, however, significantly increase midgut barrier integrity and decrease development of P. falciparum oocysts in A. stephensi, suggesting that PKC-dependent signaling is a negative regulator of epithelial barrier function and a potential new target for transmission-blocking strategies.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Anopheles / drug effects
  • Anopheles / enzymology*
  • Anopheles / parasitology*
  • Bayes Theorem
  • Digestive System / drug effects
  • Digestive System / parasitology*
  • Enzyme Activation / drug effects
  • Epithelium / drug effects
  • Epithelium / parasitology*
  • Lipopolysaccharides / pharmacology
  • Malaria / parasitology*
  • Multigene Family
  • NF-kappa B / genetics
  • Parasites / drug effects
  • Parasites / physiology
  • Phylogeny
  • Plasmodium falciparum / drug effects
  • Plasmodium falciparum / physiology
  • Promoter Regions, Genetic / genetics
  • Protein Kinase C / antagonists & inhibitors
  • Protein Kinase C / chemistry
  • Protein Kinase C / genetics
  • Protein Kinase C / metabolism*
  • Receptors, Pattern Recognition / metabolism
  • Signal Transduction* / drug effects
  • Signal Transduction* / genetics

Substances

  • Lipopolysaccharides
  • NF-kappa B
  • Receptors, Pattern Recognition
  • Protein Kinase C