First, optimal production of TgCyp18 may under normal circumstanc

First, optimal production of selleck products TgCyp18 may under normal circumstances work on CCR5 and/or other receptor(s) to recruit immune cells that produce cytokines. This possibility seems obvious in view of our previous results that showed that

TgCyp18 controlled the in vitro migration of macrophages and spleen cells in a CCR5-dependent manner [14]. In contrast, TgCyp18 may initiate cytokine production and macrophage proliferation in a CCR5-independent manner [13, 14]. Second, it is possible that stimulation of host cells with TgCyp18 via CCR5 and/or other receptor(s) could trigger expression of chemokine receptors and its ligands for cell migration. Increased CCL5 levels in the livers of the wild-type mice C59 wnt purchase infected with RH-OE parasites indicates that parasite migration to this organ occurred in a TgCyp18- and CCR5-dependent manner. Furthermore, parasite migration, which occurred in a CCR5-independent and TgCyp18-dependent way, can be explained by the higher levels of CCL2 and CXCL10 in the liver and CCL5 in the ascites fluid of CCR5−/− mice infected with RH-OE. Thus, the present results suggest that TgCyp18 has the ability to enhance host-cell migration via CCL5 and parasite

dissemination by CCL2 and CXCL10 in a CCR5-independent manner. Conclusion We determined that TgCyp18 plays a crucial role in the migration of CD11b+ cells to the site of T. gondii infection, and that the mechanisms responsible could be both dependent on and independent of CCR5 expression levels. Enhanced migration of host cells will mediate T. gondii transport to organs, especially the buy MK-8776 liver. We have shown that there are several options available to T. gondii for completing its infection cycle, one of which is CCR5-dependent, Pyruvate dehydrogenase others of which involve TgCyp18-mediated production of chemokines in a CCR5-independent manner. Additional work will be required to clarify the precise role that TgCyp18 plays in parasite-infected host cells and in parasite migration in the host.

Acknowledgments The authors are grateful to Drs. J. C. Boothroyd, (Stanford University), K. A. Joiner (Yale University), and D. S. Roos (University of Pennsylvania) for supplying the DNA constructs used to develop recombinant T. gondii. The authors would also like to thank Youko Matsushita, Megumi Noda, Yoshie Imura and Myagmarsuren Punsantsogvoo for their help with the experiments. Hany M. Ibrahim was supported by the Egyptian Ministry of High Education and Scientific Research. This research was supported by the Japan Society for the Promotion of Science through the Funding Program for Next Generation World-Leading Researchers (NEXT Program), initiated by the Council for Science and Technology Policy (2011/LS003). Electronic supplementary material Additional file 1: Figure S1: Absolute number of immune cells in the ascites fluid of mice. WT and CCR5-/- (KO) mice were infected intraperitoneally with T. gondii tachyzoites.

Comments are closed.