Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
Sci Rep ; 7(1): 2191, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28526861

ABSTRACT

The hepatic stage of the malaria parasite Plasmodium is accompanied by an autophagy-mediated host response directly targeting the parasitophorous vacuolar membrane (PVM) harbouring the parasite. Removal of the PVM-associated autophagic proteins such as ubiquitin, p62, and LC3 correlates with parasite survival. Yet, it is unclear how Plasmodium avoids the deleterious effects of selective autophagy. Here we show that parasites trap host autophagic factors in the tubovesicular network (TVN), an expansion of the PVM into the host cytoplasm. In proliferating parasites, PVM-associated LC3 becomes immediately redirected into the TVN, where it accumulates distally from the parasite's replicative centre. Finally, the host factors are shed as vesicles into the host cytoplasm. This strategy may enable the parasite to balance the benefits of the enhanced host catabolic activity with the risk of being eliminated by the cell's cytosolic immune defence.


Subject(s)
Autophagy , Host-Parasite Interactions , Malaria/metabolism , Malaria/parasitology , Plasmodium berghei/physiology , Vacuoles/metabolism , Animals , Cell Line , Cytoplasm/metabolism , Genes, Reporter , Humans , Liver/metabolism , Liver/parasitology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Biological , Protein Transport , Time-Lapse Imaging
4.
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
5.
Autophagy ; 9(4): 568-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388496

ABSTRACT

Analyzing molecular determinants of Plasmodium parasite cell death is a promising approach for exploring new avenues in the fight against malaria. Three major forms of cell death (apoptosis, necrosis and autophagic cell death) have been described in multicellular organisms but which cell death processes exist in protozoa is still a matter of debate. Here we suggest that all three types of cell death occur in Plasmodium liver-stage parasites. Whereas typical molecular markers for apoptosis and necrosis have not been found in the genome of Plasmodium parasites, we identified genes coding for putative autophagy-marker proteins and thus concentrated on autophagic cell death. We characterized the Plasmodium berghei homolog of the prominent autophagy marker protein Atg8/LC3 and found that it localized to the apicoplast. A relocalization of PbAtg8 to autophagosome-like vesicles or vacuoles that appear in dying parasites was not, however, observed. This strongly suggests that the function of this protein in liver-stage parasites is restricted to apicoplast biology.


Subject(s)
Autophagy , Life Cycle Stages , Liver/parasitology , Parasites/cytology , Parasites/growth & development , Plasmodium berghei/cytology , Plasmodium berghei/growth & development , Amino Acid Sequence , Animals , Conserved Sequence , Databases, Protein , Evolution, Molecular , Gene Knockout Techniques , Genetic Complementation Test , Green Fluorescent Proteins/metabolism , Hep G2 Cells , Humans , Lipid Metabolism , Mice , Molecular Sequence Data , Parasites/ultrastructure , Phagosomes/metabolism , Phagosomes/ultrastructure , Plasmodium berghei/ultrastructure , Protein Transport , Protozoan Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Schizonts/cytology , Schizonts/metabolism , Schizonts/ultrastructure , Sequence Homology, Amino Acid , Vacuoles/metabolism
6.
Antimicrob Agents Chemother ; 54(3): 1334-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028821

ABSTRACT

The stage-specific antimalarial activities of a panel of antiretroviral protease inhibitors (PIs), including two nonpeptidic PIs (tipranavir and darunavir), were tested in vitro against Plasmodium falciparum. While darunavir demonstrated limited antimalarial activity (effective concentration [EC(50)], >50 microM), tipranavir was active at clinically relevant concentrations (EC(50), 12 to 21 microM). Saquinavir, lopinavir, and tipranavir preferentially inhibited the growth of mature asexual-stage parasites (24 h postinvasion). While all of the PIs tested inhibited gametocytogenesis, tipranavir was the only one to exhibit gametocytocidal activity.


Subject(s)
Antimalarials/pharmacology , HIV Protease Inhibitors/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Animals , Darunavir , Erythrocytes/parasitology , Humans , Life Cycle Stages , Parasitic Sensitivity Tests , Pyridines/pharmacology , Pyrones/pharmacology , Sulfonamides/pharmacology
7.
J Virol ; 83(22): 11528-39, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726522

ABSTRACT

Nef, an important pathogenicity factor of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV), elevates virus replication in vivo. Among other activities, Nef affects T-cell receptor (TCR) signaling via several mechanisms. For HIV-1 Nef these include alteration of the organization and function of the immunological synapse (IS) such as relocalization of the Lck kinase, as well as early inhibition of TCR/CD3 complex (TCR-CD3)-mediated actin rearrangements and tyrosine phosphorylation. Although most SIV and HIV-2 Nef alleles (group 2) potently downregulate cell surface TCR-CD3, this activity was lost in the viral lineage that gave rise to HIV-1 and its SIV counterparts (group 1). To address the contribution of TCR-CD3 downregulation to Nef effects on TCR signal initiation, we compared the activities of 18 group 1 and group 2 Nef proteins, as well as SIV Nef mutants with defects in TCR-CD3 downmodulation. We found that alteration of Lck's subcellular localization is largely conserved and occurs independently of actin remodeling inhibition or TCR-CD3 downregulation. Surprisingly, Nef proteins of both groups also strongly reduced TCR-induced actin remodeling and tyrosine phosphorylation on TCR-stimulatory surfaces and TCR-CD3 downmodulation competence by group 2 Nef proteins only slightly elevated these effects. Furthermore, Nef proteins from HIV-1 and SIV reduced conjugation between infected primary human T lymphocytes and Raji B cells and potently prevented F-actin polarization at the IS independently of their ability to downmodulate TCR-CD3. These results establish alterations of early TCR signaling events at the IS, including F-actin remodeling and relocalization of Lck, as evolutionary conserved activities of highly divergent lentiviral Nef proteins.


Subject(s)
Actins/physiology , HIV-1/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Receptors, Antigen, T-Cell/physiology , nef Gene Products, Human Immunodeficiency Virus/physiology , Conserved Sequence , Down-Regulation , Humans , Jurkat Cells , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Viral Regulatory and Accessory Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...