Abstract

Beyond transient control of the infection, additional benefits of mass drug administration of praziquantel in endemic communities have been suggested in communities but not mechanistically investigated experimentally. The present study sought to evaluate the additional and hitherto unreported benefits of repeated mass drug administration of praziquantel. We used a tractable mouse model of Schistosoma mansoni infection to assess the effects of repeated infection-treatment cycles on the host susceptibility to reinfection. Parasitaemia was assessed by quantification of Schistosoma egg burden in liver tissues and morbidity was followed up by histological observation of liver lesions by microscopy and using biochemical measurement of liver transaminases. Immune responses were further determined by serum probing of schistosoma-specific antibodies, cytokines and quantification of liver cellular and soluble mediator responses by flow cytometry and ELISA, respectively. At similar ages and comparable gender distribution, groups of mice undergoing higher number of infections treatment cycles over a longer period, remained susceptible to reinfection by the parasite, as judged by the presence of eggs and the associated increasing pathology in the liver tissues. However, notably, there was a clear and significantly higher propensity to lower egg burden upon reinfection when compared to counterparts undergoing a lower number of infection-treatment cycles. This relative reduction of susceptibility to infection was paralleled by a more robust humoral response against parasite antigens, elevated serum IL-4 and liver cytokines. Of note, praziquantel treatment of infected mice left them at a higher baseline of serum IL-4, IgE and liver cytokines but lower CD4+ T cell -derived cytokines when compared to infected non-treated mice supporting an immunological treatment-induced advantage of previously infected mice over naïve mice and infected/not treated mice. Notably, repeated infection-treatment cycles did not preclude the infection-driven aggravation of collagen deposition in the livers over time and was corroborated by a more robust local production of inflammatory cytokines in the most exposed livers. Taken together, our data reveal that treatment of S. mansoni-infected hosts with praziquantel rewires the immune system to a conformation less permissive to subsequent reinfection in mice. Provided the data are translatable from mouse to human, our findings may provide mechanistic support to the potential benefits of more frequent MDAs in high transmission areas to allow rapid acquisition of protective immunity against reinfection.

Highlights

  • Schistosomiasis heavily cripples and kills many, predominantly in Africa [1,2,3]

  • From the early endpoint of week 15 (P1), a clear and robust trend of hepato and splenomegaly was apparent in infected mice when compared to naïve mice, though no difference in spleen or liver weights was observed between animals from the groups of different numbers of infection-treatment cycles

  • Reports over the last three decades have constantly emerged on the non-conventional additional benefits of repeated PZQ treatment in endemic areas [22,23,24,25,26,27] and in experimental models [19]

Read more

Summary

Introduction

Schistosomiasis heavily cripples and kills many, predominantly in Africa [1,2,3]. As no vaccine or drug is available to prevent infection, the mainstay of morbidity control in endemic communities is a yearly treatment with praziquantel (PZQ) e.g. mass drug administration programs (MDA) to school-aged children [4]. To support potential upcoming clinical studies and strategies on the field, the S. mansoni infected mouse model offers a unique possibility to assess in an accelerated view the development of schistosomiasis infection that could take decades to unfold in clinical settings [8,9,10]. Inherently resistant to the course of repeated cycles of reinfection as a result of the portal shunting of eggs [11], the mouse model still closely recapitulates some features of human schistosomiasis infections such as the anatomo-pathologic and pathophysiologic features of the infection including the immunological specifics and when used with a single inbred strain, all prone to portal shunting, differences in resistance to infection could be reliably assigned to additional immunological changes [12]. The key areas of focus in this study were the alterations in egg burden (as a proxy for susceptibility to infection) and specific immune responses prompted by PZQ treatment of infected mice

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call