Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 8(7): 1280-1292, 2023 07.
Article in English | MEDLINE | ID: mdl-37277533

ABSTRACT

For Plasmodium falciparum, the most widespread and virulent malaria parasite that infects humans, persistence depends on continuous asexual replication in red blood cells, while transmission to their mosquito vector requires asexual blood-stage parasites to differentiate into non-replicating gametocytes. This decision is controlled by stochastic derepression of a heterochromatin-silenced locus encoding AP2-G, the master transcription factor of sexual differentiation. The frequency of ap2-g derepression was shown to be responsive to extracellular phospholipid precursors but the mechanism linking these metabolites to epigenetic regulation of ap2-g was unknown. Through a combination of molecular genetics, metabolomics and chromatin profiling, we show that this response is mediated by metabolic competition for the methyl donor S-adenosylmethionine between histone methyltransferases and phosphoethanolamine methyltransferase, a critical enzyme in the parasite's pathway for de novo phosphatidylcholine synthesis. When phosphatidylcholine precursors are scarce, increased consumption of SAM for de novo phosphatidylcholine synthesis impairs maintenance of the histone methylation responsible for silencing ap2-g, increasing the frequency of derepression and sexual differentiation. This provides a key mechanistic link that explains how LysoPC and choline availability can alter the chromatin status of the ap2-g locus controlling sexual differentiation.


Subject(s)
Malaria , Parasites , Animals , Humans , Parasites/genetics , Parasites/metabolism , Histones/metabolism , Sex Differentiation , Methylation , Epigenesis, Genetic , Malaria/parasitology , Chromatin , Phosphatidylcholines , Phospholipids
2.
Commun Biol ; 6(1): 205, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36810637

ABSTRACT

Eukaryotes have canonical pathways for responding to amino acid (AA) availability. Under AA-limiting conditions, the TOR complex is repressed, whereas the sensor kinase GCN2 is activated. While these pathways have been highly conserved throughout evolution, malaria parasites are a rare exception. Despite auxotrophic for most AA, Plasmodium does not have either a TOR complex nor the GCN2-downstream transcription factors. While Ile starvation has been shown to trigger eIF2α phosphorylation and a hibernation-like response, the overall mechanisms mediating detection and response to AA fluctuation in the absence of such pathways has remained elusive. Here we show that Plasmodium parasites rely on an efficient sensing pathway to respond to AA fluctuations. A phenotypic screen of kinase knockout mutant parasites identified nek4, eIK1 and eIK2-the last two clustering with the eukaryotic eIF2α kinases-as critical for Plasmodium to sense and respond to distinct AA-limiting conditions. Such AA-sensing pathway is temporally regulated at distinct life cycle stages, allowing parasites to actively fine-tune replication and development in response to AA availability. Collectively, our data disclose a set of heterogeneous responses to AA depletion in malaria parasites, mediated by a complex mechanism that is critical for modulating parasite growth and survival.


Subject(s)
Amino Acids , Plasmodium , Amino Acids/deficiency , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Phosphorylation , Phosphotransferases/metabolism , Plasmodium/enzymology , Plasmodium/genetics
3.
Nature ; 611(7936): 563-569, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36352220

ABSTRACT

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Subject(s)
Gene Expression Regulation , Hepatocytes , Liver , Malaria , Parasites , Plasmodium berghei , Single-Cell Analysis , Animals , Hepatocytes/cytology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Liver/anatomy & histology , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Parasites/genetics , Parasites/immunology , Parasites/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Single Molecule Imaging , Sequence Analysis, RNA , Iron/metabolism , Fatty Acids/metabolism , Transcription, Genetic , Genes, Protozoan/genetics , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology
4.
Cell Rep ; 39(9): 110886, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35649358

ABSTRACT

Intracellular pathogens manipulate host cells to survive and thrive. Cellular sensing and signaling pathways are among the key host machineries deregulated to favor infection. In this study, we show that liver-stage Plasmodium parasites compete with the host to sequester a host endosomal-adaptor protein (APPL1) known to regulate signaling in response to endocytosis. The enrichment of APPL1 at the parasitophorous vacuole membrane (PVM) involves an atypical Plasmodium Rab5 isoform (Rab5b). Depletion of host APPL1 alters neither the infection nor parasite development; however, upon overexpression of a GTPase-deficient host Rab5 mutant (hRab5_Q79L), the parasites are smaller and their PVM is stripped of APPL1. Infection with the GTPase-deficient Plasmodium berghei Rab5b mutant (PbRab5b_Q91L) in this case rescues the PVM APPL1 signal and parasite size. In summary, we observe a robust correlation between the level of APPL1 retention at the PVM and parasite size during exoerythrocytic development.


Subject(s)
Parasites , Plasmodium berghei , Animals , Endocytosis , GTP Phosphohydrolases/metabolism , Liver/metabolism
6.
Sci Rep ; 10(1): 21026, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273556

ABSTRACT

Iron homeostasis is an essential biological process that ensures the tissue distribution of iron for various cellular processes. As the major producer of hepcidin, the liver is central to the regulation of iron metabolism. The liver is also home to many immune cells, which upon activation may greatly impact iron metabolism. Here, we focus on the role of invariant natural killer T (iNKT) cells, a subset of T lymphocytes that, in mice, is most abundant in the liver. Activation of iNKT cells with the prototypical glycosphingolipid antigen, α-galactosylceramide, resulted in immune cell proliferation and biphasic changes in iron metabolism. This involved an early phase characterized by hypoferremia, hepcidin induction and ferroportin suppression, and a second phase associated with strong suppression of hepcidin despite elevated levels of circulating and tissue iron. We further show that these changes in iron metabolism are fully dependent on iNKT cell activation. Finally, we demonstrate that the biphasic regulation of hepcidin is independent of NK and Kupffer cells, and is initially driven by the STAT3 inflammatory pathway, whereas the second phase is regulated by repression of the BMP/SMAD signaling pathway. These findings indicate that iNKT activation and the resulting cell proliferation influence iron homeostasis.


Subject(s)
Homeostasis , Iron/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Proliferation , Galactosylceramides/immunology , Hepcidins/genetics , Hepcidins/metabolism , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred C57BL
7.
Nat Commun ; 11(1): 5654, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159090

ABSTRACT

Plasmodium parasites possess a translocon that exports parasite proteins into the infected erythrocyte. Although the translocon components are also expressed during the mosquito and liver stage of infection, their function remains unexplored. Here, using a combination of genetic and chemical assays, we show that the translocon component Exported Protein 2 (EXP2) is critical for invasion of hepatocytes. EXP2 is a pore-forming protein that is secreted from the sporozoite upon contact with the host cell milieu. EXP2-deficient sporozoites are impaired in invasion, which can be rescued by the exogenous administration of recombinant EXP2 and alpha-hemolysin (an S. aureus pore-forming protein), as well as by acid sphingomyelinase. The latter, together with the negative impact of chemical and genetic inhibition of acid sphingomyelinase on invasion, reveals that EXP2 pore-forming activity induces hepatocyte membrane repair, which plays a key role in parasite invasion. Overall, our findings establish a novel and critical function for EXP2 that leads to an active participation of the host cell in Plasmodium sporozoite invasion, challenging the current view of the establishment of liver stage infection.


Subject(s)
Hepatocytes/parasitology , Liver/parasitology , Malaria/parasitology , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Animals , Humans , Liver/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Protein Transport , Protozoan Proteins/genetics , Sporozoites/genetics , Sporozoites/metabolism
8.
Sci Rep ; 9(1): 7575, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31110285

ABSTRACT

Malaria causes hepatic inflammation and damage, which contribute to disease severity. The pro-inflammatory cytokine interleukin (IL)-1α is released by non-hematopoietic or hematopoietic cells during liver injury. This study established the role of IL-1α in the liver pathology caused by blood-stage P. chabaudi malaria. During acute infection, hepatic inflammation and necrosis were accompanied by NLRP3 inflammasome-independent IL-1α production. Systemically, IL-1α deficiency attenuated weight loss and hypothermia but had minor effects on parasitemia control. In the liver, the absence of IL-1α reduced the number of TUNEL+ cells and necrotic lesions. This finding was associated with a lower inflammatory response, including TNF-α production. The main source of IL-1α in the liver of infected mice was inflammatory cells, particularly neutrophils. The implication of IL-1α in liver inflammation and necrosis caused by P. chabaudi infection, as well as in weight loss and hypothermia, opens up new perspectives for improving malaria outcomes by inhibiting IL-1 signaling.


Subject(s)
Inflammation/immunology , Interleukin-1alpha/immunology , Liver/pathology , Malaria/immunology , Plasmodium chabaudi/immunology , Animals , Inflammation/parasitology , Inflammation/pathology , Liver/immunology , Liver/parasitology , Malaria/parasitology , Malaria/pathology , Male , Mice, Inbred C57BL , Necrosis , Tumor Necrosis Factor-alpha/immunology
9.
Proc Natl Acad Sci U S A ; 116(20): 9979-9988, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028144

ABSTRACT

Cerebral malaria (CM) is a major cause of death due to Plasmodium infection. Both parasite and host factors contribute to the onset of CM, but the precise cellular and molecular mechanisms that contribute to its pathogenesis remain poorly characterized. Unlike conventional αß-T cells, previous studies on murine γδ-T cells failed to identify a nonredundant role for this T cell subset in experimental cerebral malaria (ECM). Here we show that mice lacking γδ-T cells are resistant to ECM when infected with Plasmodium berghei ANKA sporozoites, the liver-infective form of the parasite and the natural route of infection, in contrast with their susceptible phenotype if challenged with P. berghei ANKA-infected red blood cells that bypass the liver stage of infection. Strikingly, the presence of γδ-T cells enhanced the expression of Plasmodium immunogenic factors and exacerbated subsequent systemic and brain-infiltrating inflammatory αß-T cell responses. These phenomena were dependent on the proinflammatory cytokine IFN-γ, which was required during liver stage for modulation of the parasite transcriptome, as well as for downstream immune-mediated pathology. Our work reveals an unanticipated critical role of γδ-T cells in the development of ECM upon Plasmodium liver-stage infection.


Subject(s)
Intraepithelial Lymphocytes/physiology , Liver/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/pathogenicity , Sporozoites/pathogenicity , Animals , Liver/parasitology , Male , Mice , Mice, Inbred C57BL , Sporozoites/growth & development
10.
Article in English | MEDLINE | ID: mdl-32010639

ABSTRACT

Plasmodium parasites undergo a dramatic transformation during the liver stage of their life cycle, amplifying over 10,000-fold inside infected hepatocytes within a few days. Such a rapid growth requires large-scale interactions with, and manipulations of, host cell functions. Whereas hepatocyte polarity is well-known to be critical for liver function, little is presently known about its involvement during the liver stage of Plasmodium development. Apical domains of hepatocytes are critical components of their polarity machinery and constitute the bile canalicular network, which is central to liver function. Here, we employed high resolution 3-D imaging and advanced image analysis of Plasmodium-infected liver tissues to show that the parasite associates preferentially with the apical domain of hepatocytes and induces alterations in the organization of these regions, resulting in localized changes in the bile canalicular architecture in the liver tissue. Pharmacological perturbation of the bile canalicular network by modulation of AMPK activity reduces the parasite's association with bile canaliculi and arrests the parasite development. Our findings using Plasmodium-infected liver tissues reveal a host-Plasmodium interaction at the level of liver tissue organization. We demonstrate for the first time a role for bile canaliculi, a central component of the hepatocyte polarity machinery, during the liver stage of Plasmodium development.


Subject(s)
Hepatocytes/parasitology , Host-Pathogen Interactions/physiology , Liver/parasitology , Malaria/parasitology , Plasmodium berghei/physiology , Animals , Bile Acids and Salts/analysis , Bile Canaliculi/diagnostic imaging , Bile Canaliculi/parasitology , Bile Canaliculi/pathology , Disease Models, Animal , Imaging, Three-Dimensional , Life Cycle Stages , Liver/diagnostic imaging , Liver/pathology , Malaria/diagnostic imaging , Malaria/pathology , Mice , Mice, Inbred C57BL
11.
Cell Host Microbe ; 23(6): 749-758, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29902440

ABSTRACT

Parasites undergo complex life cycles that comprise a wide variety of cellular differentiation events in different host compartments and transmission across multiple hosts. As parasites depend on host resources, it is not surprising they have developed efficient mechanisms to sense alterations and adapt to the available resources in a wide range of environments. Here we provide an overview of the nutritional needs of different parasites throughout their diverse life stages and highlight recent insights into strategies that both hosts and parasites have developed to meet these nutritional requirements needed for defense, survival, and replication. These studies will provide the foundation for a systems-level understanding of host-parasite interactions, which will require the integration of molecular, epidemiologic, and mechanistic data and the application of interdisciplinary approaches to model parasite regulatory networks that are triggered by alterations in host resources.


Subject(s)
Host-Parasite Interactions/physiology , Nutrients/metabolism , Parasites/metabolism , Adaptation, Biological , Animals , Life Cycle Stages , Nutritional Status/physiology , Parasites/pathogenicity , Plasmodium/metabolism , Toxoplasma/metabolism , Trypanosoma/metabolism
12.
Cell Metab ; 27(4): 708-709, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617637

ABSTRACT

Several vacuolar bacteria and parasites, such as Legionella, Chlamydia, and Toxoplasma, have been reported to grow associated with host mitochondria. The reason behind this phenomenon remains elusive. In this issue of Cell Metabolism, Pernas et al. (2018) propose that fusion of host mitochondria limits the availability of fatty acids needed for Toxoplasma gondii replication.


Subject(s)
Parasites , Toxoplasma , Animals , Fatty Acids , Host-Parasite Interactions , Mitochondria
13.
Nat Microbiol ; 3(1): 17-25, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29109477

ABSTRACT

The causative agent of malaria, Plasmodium, replicates inside a membrane-bound parasitophorous vacuole (PV), which shields this intracellular parasite from the cytosol of the host cell 1 . One common threat for intracellular pathogens is the homeostatic process of autophagy, through which cells capture unwanted intracellular material for lysosomal degradation 2 . During the liver stage of a malaria infection, Plasmodium parasites are targeted by the autophagy machinery of the host cell, and the PV membrane (PVM) becomes decorated with several autophagy markers, including LC3 (microtubule-associated protein 1 light chain 3) 3,4 . Here we show that Plasmodium berghei parasites infecting hepatic cells rely on the PVM transmembrane protein UIS3 to avoid elimination by host-cell-mediated autophagy. We found that UIS3 binds host LC3 through a non-canonical interaction with a specialized surface on LC3 where host proteins with essential functions during autophagy also bind. UIS3 acts as a bona fide autophagy inhibitor by competing with host LC3-interacting proteins for LC3 binding. Our work identifies UIS3, one of the most promising candidates for a genetically attenuated vaccine against malaria 5 , as a unique and potent mediator of autophagy evasion in Plasmodium. We propose that the protein-protein interaction between UIS3 and host LC3 represents a target for antimalarial drug development.


Subject(s)
Autophagy/physiology , Hepatocytes/pathology , Malaria/pathology , Malaria/parasitology , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Plasmodium berghei/genetics , Animals , Autophagosomes/metabolism , Cell Line , HEK293 Cells , Hep G2 Cells , Hepatocytes/parasitology , Hepatocytes/ultrastructure , Host-Pathogen Interactions , Humans , Malaria/physiopathology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Models, Molecular , Plasmodium berghei/metabolism , Plasmodium berghei/pathogenicity , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Vacuoles/metabolism
14.
Nat Microbiol ; 2(12): 1600-1607, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28947801

ABSTRACT

The relevance of genetic factors in conferring protection to severe malaria has been demonstrated, as in the case of sickle cell trait and G6PD deficiency 1 . However, it remains unknown whether environmental components, such as dietary or metabolic variations, can contribute to the outcome of infection 2 . Here, we show that administration of a high-fat diet to mice for a period as short as 4 days impairs Plasmodium liver infection by over 90%. Plasmodium sporozoites can successfully invade and initiate replication but die inside hepatocytes, thereby are unable to cause severe disease. Transcriptional analyses combined with genetic and chemical approaches reveal that this impairment of infection is mediated by oxidative stress. We show that reactive oxygen species, probably spawned from fatty acid ß-oxidation, directly impact Plasmodium survival inside hepatocytes, and parasite load can be rescued by exogenous administration of the antioxidant N-acetylcysteine or the ß-oxidation inhibitor etomoxir. Together, these data reveal that acute and transient dietary alterations markedly impact the establishment of a Plasmodium infection and disease outcome.


Subject(s)
Diet, High-Fat/methods , Host-Parasite Interactions/genetics , Malaria/diet therapy , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Glucose Tolerance Test , Glucosephosphate Dehydrogenase Deficiency/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/parasitology , Humans , Liver/metabolism , Liver/parasitology , Liver Diseases/metabolism , Liver Diseases/parasitology , Macrophages/parasitology , Macrophages/pathology , Malaria/blood , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Parasite Load , Plasmodium berghei , Reactive Oxygen Species , Sickle Cell Trait/metabolism , Sporozoites/metabolism
15.
Elife ; 62017 05 02.
Article in English | MEDLINE | ID: mdl-28525314

ABSTRACT

Gliding motility allows malaria parasites to migrate and invade tissues and cells in different hosts. It requires parasite surface proteins to provide attachment to host cells and extracellular matrices. Here, we identify the Plasmodium protein LIMP (the name refers to a gliding phenotype in the sporozoite arising from epitope tagging of the endogenous protein) as a key regulator for adhesion during gliding motility in the rodent malaria model P. berghei. Transcribed in gametocytes, LIMP is translated in the ookinete from maternal mRNA, and later in the sporozoite. The absence of LIMP reduces initial mosquito infection by 50%, impedes salivary gland invasion 10-fold, and causes a complete absence of liver invasion as mutants fail to attach to host cells. GFP tagging of LIMP caused a limping defect during movement with reduced speed and transient curvature changes of the parasite. LIMP is an essential motility and invasion factor necessary for malaria transmission.


Subject(s)
Culicidae/parasitology , Locomotion , Lysosomal Membrane Proteins/metabolism , Plasmodium berghei/physiology , Protozoan Proteins/metabolism , Sporozoites/physiology , Virulence Factors/metabolism , Animals , Disease Models, Animal , Liver/parasitology , Malaria/parasitology , Membrane Proteins/metabolism , Mice
16.
Cell Microbiol ; 18(3): 437-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26399761

ABSTRACT

Autophagy plays an important role in the defence against intracellular pathogens. However, some microorganisms can manipulate this host cell pathway to their advantage. In this study, we addressed the role of host cell autophagy during Plasmodium berghei liver infection. We show that vesicles containing the autophagic marker LC3 surround parasites from early time-points after invasion and throughout infection and colocalize with the parasitophorous vacuole membrane. Moreover, we show that the LC3-positive vesicles that surround Plasmodium parasites are amphisomes that converge from the endocytic and autophagic pathways, because they contain markers of both pathways. When the host autophagic pathway was inhibited by silencing several of its key regulators such as LC3, Beclin1, Vps34 or Atg5, we observed a reduction in parasite size. We also found that LC3 surrounds parasites in vivo and that parasite load is diminished in a mouse model deficient for autophagy. Together, these results show the importance of the host autophagic pathway for parasite development during the liver stage of Plasmodium infection.


Subject(s)
Autophagy/physiology , Host-Parasite Interactions/physiology , Liver/parasitology , Malaria/pathology , Plasmodium berghei/pathogenicity , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Liver/pathology , Malaria/parasitology , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism
17.
Nat Rev Microbiol ; 13(9): 589-98, 2015 09.
Article in English | MEDLINE | ID: mdl-26189591

ABSTRACT

Many clinically relevant pathogens, including certain bacteria and protozoan parasites, have developed an intracellular lifestyle that enables them to nestle in customized vacuoles. Although these pathogens are protected from extracellular defences, recent findings indicate that host cells have evolved multiple strategies to unmask the pathogen disguised by the vacuole and thereby initiate innate immune responses. In this Opinion article, we propose and discuss models by which hosts can sense 'professional' vacuolar pathogens, and we highlight the ability of the host to target these stealthy bacteria and parasites.


Subject(s)
Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Host-Pathogen Interactions , Parasitic Diseases/parasitology , Vacuoles , Animals , Bacterial Infections/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Parasitic Diseases/immunology
18.
EMBO Rep ; 16(8): 955-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26113366

ABSTRACT

Upon infection of a mammalian host, Plasmodium parasites first replicate inside hepatocytes, generating thousands of new parasites. Although Plasmodium intra-hepatic development represents a substantial metabolic challenge to the host hepatocyte, how infected cells respond to and integrate this stress remains poorly understood. Here, we present proteomic and transcriptomic analyses, revealing that the endoplasmic reticulum (ER)-resident unfolded protein response (UPR) is activated in host hepatocytes upon Plasmodium berghei infection. The expression of XBP1s--the active form of the UPR mediator XBP1--and the liver-specific UPR mediator CREBH is induced by P. berghei infection in vivo. Furthermore, this UPR induction increases parasite liver burden. Altogether, our data suggest that ER stress is a central feature of P. berghei intra-hepatic development, contributing to the success of infection.


Subject(s)
Endoplasmic Reticulum Stress , Hepatocytes/parasitology , Host-Parasite Interactions , Malaria/parasitology , Plasmodium berghei/growth & development , Unfolded Protein Response , Animals , DNA-Binding Proteins/genetics , Gene Expression Profiling , Hepatocytes/physiology , Hepatocytes/ultrastructure , Life Cycle Stages , Malaria/physiopathology , Male , Mice, Inbred C57BL , Parasite Load , Plasmodium berghei/pathogenicity , Proteomics , Regulatory Factor X Transcription Factors , Signal Transduction/genetics , Transcription Factors/genetics , X-Box Binding Protein 1
19.
Sci Transl Med ; 7(288): 288ra77, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995223

ABSTRACT

The emergence of drug resistance is a major limitation of current antimalarials. The discovery of new druggable targets and pathways including those that are critical for multiple life cycle stages of the malaria parasite is a major goal for developing next-generation antimalarial drugs. Using an integrated chemogenomics approach that combined drug resistance selection, whole-genome sequencing, and an orthogonal yeast model, we demonstrate that the cytoplasmic prolyl-tRNA (transfer RNA) synthetase (PfcPRS) of the malaria parasite Plasmodium falciparum is a biochemical and functional target of febrifugine and its synthetic derivative halofuginone. Febrifugine is the active principle of a traditional Chinese herbal remedy for malaria. We show that treatment with febrifugine derivatives activated the amino acid starvation response in both P. falciparum and a transgenic yeast strain expressing PfcPRS. We further demonstrate in the Plasmodium berghei mouse model of malaria that halofuginol, a new halofuginone analog that we developed, is active against both liver and asexual blood stages of the malaria parasite. Halofuginol, unlike halofuginone and febrifugine, is well tolerated at efficacious doses and represents a promising lead for the development of dual-stage next-generation antimalarials.


Subject(s)
Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Malaria, Falciparum/drug therapy , Piperidines/pharmacology , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Quinazolinones/pharmacology , Amino Acyl-tRNA Synthetases/metabolism , Animals , Antimalarials/chemistry , Antimalarials/toxicity , Computer-Aided Design , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Drug Resistance , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Erythrocytes/parasitology , Liver/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Mice , Models, Molecular , Molecular Structure , Molecular Targeted Therapy , Piperidines/chemistry , Piperidines/toxicity , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Quinazolines/chemistry , Quinazolines/toxicity , Quinazolinones/chemistry , Quinazolinones/toxicity , Structure-Activity Relationship , Time Factors
20.
Cell Host Microbe ; 16(6): 778-86, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25498345

ABSTRACT

During invasion, Plasmodium, the causative agent of malaria, wraps itself in a parasitophorous vacuole membrane (PVM), which constitutes a critical interface between the parasite and its host cell. Within hepatocytes, each Plasmodium sporozoite generates thousands of new parasites, creating high demand for lipids to support this replication and enlarge the PVM. Here, a global analysis of the total lipid repertoire of Plasmodium-infected hepatocytes reveals an enrichment of neutral lipids and the major membrane phospholipid, phosphatidylcholine (PC). While infection is unaffected in mice deficient in key enzymes involved in neutral lipid synthesis and lipolysis, ablation of rate-limiting enzymes in hepatic PC biosynthetic pathways significantly decreases parasite numbers. Host PC is taken up by both P. berghei and P. falciparum and is necessary for correct localization of parasite proteins to the PVM, which is essential for parasite survival. Thus, Plasmodium relies on the abundance of these lipids within hepatocytes to support infection.


Subject(s)
Liver/parasitology , Malaria/metabolism , Phosphatidylcholines/biosynthesis , Plasmodium berghei/metabolism , Plasmodium falciparum/metabolism , Animals , Cell Line , Cell Survival , Female , Host-Parasite Interactions , Humans , Lipid Metabolism , Liver/metabolism , Malaria/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/growth & development , Plasmodium falciparum/growth & development , Sporozoites/growth & development , Sporozoites/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...