Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Mol Microbiol ; 121(6): 1095-1111, 2024 06.
Article in English | MEDLINE | ID: mdl-38574236

ABSTRACT

The protozoan parasite Plasmodium, the causative agent of malaria, undergoes an obligatory stage of intra-hepatic development before initiating a blood-stage infection. Productive invasion of hepatocytes involves the formation of a parasitophorous vacuole (PV) generated by the invagination of the host cell plasma membrane. Surrounded by the PV membrane (PVM), the parasite undergoes extensive replication. During intracellular development in the hepatocyte, the parasites provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterized by a long-lasting association of the autophagy marker protein, and ATG8 family member, LC3B with the PVM. LC3B localization at the PVM does not follow the canonical autophagy pathway since upstream events specific to canonical autophagy are dispensable. Here, we describe that LC3B localization at the PVM of Plasmodium parasites requires the V-ATPase and its interaction with ATG16L1. The WD40 domain of ATG16L1 is crucial for its recruitment to the PVM. Thus, we provide new mechanistic insight into the previously described PAAR response targeting Plasmodium liver stage parasites.


Subject(s)
Autophagy-Related Proteins , Autophagy , Hepatocytes , Liver , Microtubule-Associated Proteins , Plasmodium berghei , Vacuolar Proton-Translocating ATPases , Vacuoles , Vacuoles/metabolism , Vacuoles/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium berghei/enzymology , Animals , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Liver/parasitology , Mice , Hepatocytes/parasitology , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Malaria/parasitology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Humans
2.
Cell Microbiol ; 23(1): e13271, 2021 01.
Article in English | MEDLINE | ID: mdl-32979009

ABSTRACT

The protozoan parasite Plasmodium, causative agent of malaria, invades hepatocytes by invaginating the host cell plasma membrane and forming a parasitophorous vacuole membrane (PVM). Surrounded by this PVM, the parasite undergoes extensive replication. Parasites inside a PVM provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterised by a long-lasting association of the autophagy marker protein LC3 with the PVM, which is not preceded by phosphatidylinositol 3-phosphate (PI3P)-labelling. Prior to productive invasion, sporozoites transmigrate several cells and here we describe that a proportion of traversing sporozoites become trapped in a transient traversal vacuole, provoking a host cell response that clearly differs from the PAAR response. These trapped sporozoites provoke PI3P-labelling of the surrounding vacuolar membrane immediately after cell entry, followed by transient LC3-labelling and elimination of the parasite by lysosomal acidification. Our data suggest that this PI3P response is not only restricted to sporozoites trapped during transmigration but also affects invaded parasites residing in a compromised vacuole. Thus, host cells can employ a pathway distinct from the previously described PAAR response to efficiently recognise and eliminate Plasmodium parasites.


Subject(s)
Autophagy , Hepatocytes/parasitology , Phosphatidylinositol Phosphates/metabolism , Plasmodium berghei/metabolism , Plasmodium berghei/parasitology , Sporozoites/metabolism , Vacuoles/parasitology , Animals , Cell Line , Female , HeLa Cells , Host-Parasite Interactions , Humans , Malaria/parasitology , Mice , Microtubule-Associated Proteins/metabolism , Organisms, Genetically Modified
3.
Cell Microbiol ; 21(9): e13046, 2019 09.
Article in English | MEDLINE | ID: mdl-31099152

ABSTRACT

The virulence strategy of pathogenic Yersinia spp. involves cell-invasive as well as phagocytosis-preventing tactics to enable efficient colonisation of the host organism. Enteropathogenic yersiniae display an invasive phenotype in early infection stages, which facilitates penetration of the intestinal mucosa. Here we show that invasion of epithelial cells by Yersinia enterocolitica is followed by intracellular survival and multiplication of a subset of ingested bacteria. The replicating bacteria were enclosed in vacuoles with autophagy-related characteristics, showing phagophore formation, xenophagy, and recruitment of cytoplasmic autophagosomes to the bacteria-containing compartments. The subsequent fusion of these vacuoles with lysosomes and concomitant vesicle acidification were actively blocked by Yersinia. This resulted in increased intracellular proliferation and detectable egress of yersiniae from infected cells. Notably, deficiency of the core autophagy machinery component FIP200 impaired the development of autophagic features at Yersinia-containing vacuoles as well as intracellular replication and release of bacteria to the extracellular environment. These results suggest that Y. enterocolitica may take advantage of the macroautophagy pathway in epithelial cells to create an autophagosomal niche that supports intracellular bacterial survival, replication, and, eventually, spread of the bacteria from infected cells.


Subject(s)
Autophagosomes/microbiology , Epithelial Cells/microbiology , Yersinia enterocolitica/pathogenicity , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Cell Death , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , HeLa Cells , Host Microbial Interactions , Humans , Lysosomes/metabolism , Lysosomes/microbiology , Lysosomes/ultrastructure , Mice , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Vacuoles/metabolism , Vacuoles/microbiology , Vacuoles/ultrastructure , Yersinia enterocolitica/growth & development , Yersinia enterocolitica/metabolism
4.
FEMS Microbiol Rev ; 42(3): 324-334, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29529207

ABSTRACT

Recent years have witnessed a great gain in knowledge regarding parasite-host cell interactions during Plasmodium liver stage development. It is now an accepted fact that a large percentage of sporozoites invading hepatocytes fail to form infectious merozoites. There appears to be a delicate balance between parasite survival and elimination and we now start to understand why this is so. Plasmodium liver stage parasites replicate within the parasitophorous vacuole (PV), formed during invasion by invagination of the host cell plasma membrane. The main interface between the parasite and hepatocyte is the parasitophorous vacuole membrane (PVM) that surrounds the PV. Recently, it was shown that autophagy marker proteins decorate the PVM of Plasmodium liver stage parasites and eliminate a proportion of them by an autophagy-like mechanism. Successfully developing Plasmodium berghei parasites are initially also labeled but in the course of development, they are able to control this host defense mechanism by shedding PVM material into the tubovesicular network (TVN), an extension of the PVM that releases vesicles into the host cell cytoplasm. Better understanding of the molecular events at the PVM/TVN during parasite elimination could be the basis of new antimalarial measures.


Subject(s)
Cytosol/immunology , Cytosol/parasitology , Host-Parasite Interactions/immunology , Liver/immunology , Liver/parasitology , Plasmodium/growth & development , Plasmodium/immunology , Animals , Hepatocytes/immunology , Hepatocytes/parasitology , Humans , Life Cycle Stages
5.
PLoS One ; 12(8): e0183797, 2017.
Article in English | MEDLINE | ID: mdl-28841718

ABSTRACT

Selective autophagy and related mechanisms can act as variable defense mechanisms against pathogens and can therefore be considered as intracellular immune responses. When in hepatocytes, Plasmodium parasites reside in a parasitophorous vacuole (PV) and the PV membrane (PVM) is the main contact site between host cell and parasite. Early in infection, the PVM is directly labeled with host cell autophagy proteins LC3B and p62 (nucleoporin 62). We investigated the recruitment of different selective autophagy receptors and could show that mainly p62 and NBR1 (neighbour of BRCA1 gene 1) and to a lesser extent NDP52 (nuclear dot protein 52) associate with the PVM. To investigate the recruitment of these receptors to the PVM in Plasmodium-infected cells, we generated LC3B knock out HeLa cells. In these cell lines, autophagosome formation and autophagic flux are not different to those in WT cells. Unexpectedly, p62 and NBR1 recruitment to the PVM was strongly impaired in LC3B-negative host cells, suggesting that LC3B recruits both receptors to the PVM of Plasmodium parasites. We also noticed that LC3B recruited ubiquitin to the PVM. This indicates that, in comparison to classical selective autophagy, in P. berghei-infected cells the order of membrane labeling with autophagy proteins appears to be inverted from canonical ubiquitin-receptor-LC3B recruitment to LC3B-receptor and possibly ubiquitin.


Subject(s)
Autophagy , Plasmodium berghei/physiology , Animals , HeLa Cells , Humans , Vacuoles/metabolism
6.
Cell Microbiol ; 19(10)2017 10.
Article in English | MEDLINE | ID: mdl-28573684

ABSTRACT

Eukaryotic cells can employ autophagy to defend themselves against invading pathogens. Upon infection by Plasmodium berghei sporozoites, the host hepatocyte targets the invader by labelling the parasitophorous vacuole membrane (PVM) with the autophagy marker protein LC3. Until now, it has not been clear whether LC3 recruitment to the PVM is mediated by fusion of autophagosomes or by direct incorporation. To distinguish between these possibilities, we knocked out genes that are essential for autophagosome formation and for direct LC3 incorporation into membranes. The CRISPR/Cas9 system was employed to generate host cell lines deficient for either FIP200, a member of the initiation complex for autophagosome formation, or ATG5, responsible for LC3 lipidation and incorporation of LC3 into membranes. Infection of these knockout cell lines with P. berghei sporozoites revealed that LC3 recruitment to the PVM indeed depends on functional ATG5 and the elongation machinery, but not on FIP200 and the initiation complex, suggesting a direct incorporation of LC3 into the PVM. Importantly, in P. berghei-infected ATG5-/- host cells, lysosomes still accumulated at the PVM, indicating that the recruitment of lysosomes follows an LC3-independent pathway.


Subject(s)
Liver/physiopathology , Plasmodium berghei/metabolism , Plasmodium berghei/pathogenicity , Vacuoles/metabolism , Autophagosomes/metabolism , Autophagy/physiology , CRISPR-Cas Systems/physiology , Lysosomes/metabolism , Signal Transduction/physiology , Sporozoites/metabolism
7.
Malar J ; 15: 232, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27102897

ABSTRACT

BACKGROUND: Bioluminescence imaging is widely used for cell-based assays and animal imaging studies, both in biomedical research and drug development. Its main advantages include its high-throughput applicability, affordability, high sensitivity, operational simplicity, and quantitative outputs. In malaria research, bioluminescence has been used for drug discovery in vivo and in vitro, exploring host-pathogen interactions, and studying multiple aspects of Plasmodium biology. While the number of fluorescent proteins available for imaging has undergone a great expansion over the last two decades, enabling simultaneous visualization of multiple molecular and cellular events, expansion of available luciferases has lagged behind. The most widely used bioluminescent probe in malaria research is the Photinus pyralis firefly luciferase, followed by the more recently introduced Click-beetle and Renilla luciferases. Ultra-sensitive imaging of Plasmodium at low parasite densities has not been previously achieved. With the purpose of overcoming these challenges, a Plasmodium berghei line expressing the novel ultra-bright luciferase enzyme NanoLuc, called PbNLuc has been generated, and is presented in this work. RESULTS: NanoLuc shows at least 150 times brighter signal than firefly luciferase in vitro, allowing single parasite detection in mosquito, liver, and sexual and asexual blood stages. As a proof-of-concept, the PbNLuc parasites were used to image parasite development in the mosquito, liver and blood stages of infection, and to specifically explore parasite liver stage egress, and pre-patency period in vivo. CONCLUSIONS: PbNLuc is a suitable parasite line for sensitive imaging of the entire Plasmodium life cycle. Its sensitivity makes it a promising line to be used as a reference for drug candidate testing, as well as the characterization of mutant parasites to explore the function of parasite proteins, host-parasite interactions, and the better understanding of Plasmodium biology. Since the substrate requirements of NanoLuc are different from those of firefly luciferase, dual bioluminescence imaging for the simultaneous characterization of two lines, or two separate biological processes, is possible, as demonstrated in this work.


Subject(s)
Luminescent Measurements/methods , Malaria/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Animals , Culicidae/parasitology , Host-Parasite Interactions , Humans , Liver/parasitology , Luciferases/genetics , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/metabolism , Plasmodium berghei/isolation & purification
8.
Autophagy ; 11(9): 1561-79, 2015.
Article in English | MEDLINE | ID: mdl-26208778

ABSTRACT

Plasmodium parasites are transmitted by Anopheles mosquitoes to the mammalian host and actively infect hepatocytes after passive transport in the bloodstream to the liver. In their target host hepatocyte, parasites reside within a parasitophorous vacuole (PV). In the present study it was shown that the parasitophorous vacuole membrane (PVM) can be targeted by autophagy marker proteins LC3, ubiquitin, and SQSTM1/p62 as well as by lysosomes in a process resembling selective autophagy. The dynamics of autophagy marker proteins in individual Plasmodium berghei-infected hepatocytes were followed by live imaging throughout the entire development of the parasite in the liver. Although the host cell very efficiently recognized the invading parasite in its vacuole, the majority of parasites survived this initial attack. Successful parasite development correlated with the gradual loss of all analyzed autophagy marker proteins and associated lysosomes from the PVM. However, other autophagic events like nonselective canonical autophagy in the host cell continued. This was indicated as LC3, although not labeling the PVM anymore, still localized to autophagosomes in the infected host cell. It appears that growing parasites even benefit from this form of nonselective host cell autophagy as an additional source of nutrients, as in host cells deficient for autophagy, parasite growth was retarded and could partly be rescued by the supply of additional amino acid in the medium. Importantly, mouse infections with P. berghei sporozoites confirmed LC3 dynamics, the positive effect of autophagy activation on parasite growth, and negative effects upon autophagy inhibition.


Subject(s)
Cytosol/immunology , Hepatocytes/immunology , Imaging, Three-Dimensional , Immune Evasion , Immunity , Malaria/immunology , Parasites/immunology , Plasmodium berghei/pathogenicity , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy , Biomarkers/metabolism , Galectins/metabolism , Heat-Shock Proteins/metabolism , Hep G2 Cells , Hepatocytes/parasitology , Hepatocytes/ultrastructure , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Life Cycle Stages , Liver/parasitology , Lysosomes/metabolism , Lysosomes/ultrastructure , Malaria/parasitology , Mice , Microtubule-Associated Proteins/metabolism , Parasites/growth & development , Parasites/pathogenicity , Parasites/ultrastructure , Plasmodium berghei/growth & development , Plasmodium berghei/ultrastructure , Sequestosome-1 Protein , Sporozoites/physiology , Sporozoites/ultrastructure , Survival Analysis , Time Factors , Ubiquitin/metabolism , Ubiquitination , Vacuoles/metabolism , Vacuoles/ultrastructure , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...