Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Trends Parasitol ; 40(6): 466-476, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714463

ABSTRACT

The Plasmodium parasites that cause malaria undergo asymptomatic development in the parenchymal cells of the liver, the hepatocytes, prior to infecting erythrocytes and causing clinical disease. Traditionally, hepatocytes have been perceived as passive bystanders that allow hepatotropic pathogens such as Plasmodium to develop relatively unchallenged. However, now there is emerging evidence suggesting that hepatocytes can mount robust cell-autonomous immune responses that target Plasmodium, limiting its progression to the blood and reducing the incidence and severity of clinical malaria. Here we discuss our current understanding of hepatocyte cell-intrinsic immune responses that target Plasmodium and how these pathways impact malaria.


Subject(s)
Hepatocytes , Malaria , Plasmodium , Plasmodium/immunology , Plasmodium/physiology , Humans , Malaria/immunology , Malaria/parasitology , Hepatocytes/parasitology , Hepatocytes/immunology , Animals
2.
J Immunol ; 212(4): 596-606, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38149914

ABSTRACT

Inflammasome-mediated caspase-1 activation facilitates innate immune control of Plasmodium in the liver, thereby limiting the incidence and severity of clinical malaria. However, caspase-1 processing occurs incompletely in both mouse and human hepatocytes and precludes the generation of mature IL-1ß or IL-18, unlike in other cells. Why this is so or how it impacts Plasmodium control in the liver has remained unknown. We show that an inherently reduced expression of the inflammasome adaptor molecule apoptosis-associated specklike protein containing CARD (ASC) is responsible for the incomplete proteolytic processing of caspase-1 in murine hepatocytes. Transgenically enhancing ASC expression in hepatocytes enabled complete caspase-1 processing, enhanced pyroptotic cell death, maturation of the proinflammatory cytokines IL-1ß and IL-18 that was otherwise absent, and better overall control of Plasmodium infection in the liver of mice. This, however, impeded the protection offered by live attenuated antimalarial vaccination. Tempering ASC expression in mouse macrophages, on the other hand, resulted in incomplete processing of caspase-1. Our work shows how caspase-1 activation and function in host cells are fundamentally defined by ASC expression and offers a potential new pathway to create better disease and vaccination outcomes by modifying the latter.


Subject(s)
Inflammasomes , Malaria , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Hepatocytes/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...