Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 14(659): eabn9709, 2022 08 24.
Article in English | MEDLINE | ID: mdl-36001680

ABSTRACT

Genetically engineered live Plasmodium falciparum sporozoites constitute a potential platform for creating consistently attenuated, genetically defined, whole-parasite vaccines against malaria through targeted gene deletions. Such genetically attenuated parasites (GAPs) do not require attenuation by irradiation or concomitant drug treatment. We previously developed a P. falciparum (Pf) GAP with deletions in P52, P36, and SAP1 genes (PfGAP3KO) and demonstrated its safety and immunogenicity in humans. Here, we further assessed safety, tolerability, and immunogenicity of the PfGAP3KO vaccine and tested its efficacy against controlled human malaria infection (CHMI) in malaria-naïve subjects. The vaccine was delivered by three (n = 6) or five (n = 8) immunizations with ~200 PfGAP3KO-infected mosquito bites per immunization. PfGAP3KO was safe and well tolerated with no breakthrough P. falciparum blood stage infections. Vaccine-related adverse events were predominately localized urticaria related to the numerous mosquito bites administered per vaccination. CHMI via bites with mosquitoes carrying fully infectious Pf NF54 parasites was carried out 1 month after the last immunization. Half of the study participants who received either three or five PfGAP3KO immunizations remained P. falciparum blood stage negative, as shown by a lack of detection of Plasmodium 18S rRNA in the blood for 28 days after CHMI. Six protected study participants received a second CHMI 6 months later, and one remained completely protected. Thus, the PfGAP3KO vaccine was safe and immunogenic and was capable of inducing protection against sporozoite infection. These results warrant further evaluation of PfGAP3KO vaccine efficacy in dose-range finding trials with an injectable formulation.


Subject(s)
Insect Bites and Stings , Malaria Vaccines , Malaria, Falciparum , Malaria , Parasites , Animals , Humans , Insect Bites and Stings/chemically induced , Malaria/prevention & control , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/genetics , Sporozoites/genetics , Vaccines, Attenuated
2.
Cell Chem Biol ; 29(9): 1419-1433.e5, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35738280

ABSTRACT

Prior to initiating symptomatic malaria, a single Plasmodium sporozoite infects a hepatocyte and develops into thousands of merozoites, in part by scavenging host resources, likely delivered by vesicles. Here, we demonstrate that host microtubules (MTs) dynamically reorganize around the developing liver stage (LS) parasite to facilitate vesicular transport to the parasite. Using a genome-wide CRISPR-Cas9 screen, we identified host regulators of cytoskeleton organization, vesicle trafficking, and ER/Golgi stress that regulate LS development. Foci of γ-tubulin localized to the parasite periphery; depletion of centromere protein J (CENPJ), a novel regulator identified in the screen, exacerbated this re-localization and increased infection. We demonstrate that the Golgi acts as a non-centrosomal MT organizing center (ncMTOC) by positioning γ-tubulin and stimulating MT nucleation at parasite periphery. Together, these data support a model where the Plasmodium LS recruits host Golgi to form MT-mediated conduits along which host organelles are recruited to PVM and support parasite development.


Subject(s)
Malaria , Microtubule-Associated Proteins , Microtubules , CRISPR-Cas Systems , Humans , Liver/metabolism , Liver/parasitology , Malaria/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Plasmodium/metabolism , Tubulin/metabolism
3.
Cell Chem Biol ; 28(12): 1679-1692.e4, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34216546

ABSTRACT

Kinase inhibitors are promising drugs to stabilize the endothelial barrier following inflammatory damage. However, our limited knowledge of how kinase signaling activates barrier-restorative pathways and the complexity of multi-target drugs have hindered drug discovery and repurposing efforts. Here, we apply a kinase regression approach that exploits drug polypharmacology to investigate endothelial barrier regulation. A screen of 28 kinase inhibitors identified multiple inhibitors that promote endothelial barrier integrity and revealed divergent barrier phenotypes for BCR-ABL drugs. Target deconvolution predicted 50 barrier-regulating kinases from diverse kinase families. Using gene knockdowns, we identified kinases with a role in endothelial barrier regulation and dissected different mechanisms of action of barrier-protective kinase inhibitors. These results demonstrate the importance of polypharmacology in the endothelial barrier phenotype of kinase inhibitors and provide promising new leads for barrier-strengthening therapies.


Subject(s)
Aniline Compounds/pharmacology , Carbazoles/pharmacology , Indole Alkaloids/pharmacology , Nitriles/pharmacology , Phosphotransferases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Aniline Compounds/chemistry , Carbazoles/chemistry , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Indole Alkaloids/chemistry , Nitriles/chemistry , Phosphotransferases/genetics , Phosphotransferases/metabolism , Polypharmacology , Protein Kinase Inhibitors/chemistry , Quinolines/chemistry , Signal Transduction/drug effects
4.
Cell Death Differ ; 27(1): 44-54, 2020 01.
Article in English | MEDLINE | ID: mdl-31065106

ABSTRACT

The facets of host control during Plasmodium liver infection remain largely unknown. We find that the SLC7a11-GPX4 pathway, which has been associated with the production of reactive oxygen species, lipid peroxidation, and a form of cell death called ferroptosis, plays a critical role in control of Plasmodium liver stage infection. Specifically, blocking GPX4 or SLC7a11 dramatically reduces Plasmodium liver stage parasite infection. In contrast, blocking negative regulators of this pathway, NOX1 and TFR1, leads to an increase in liver stage infection. We have shown previously that increased levels of P53 reduces Plasmodium LS burden in an apoptosis-independent manner. Here, we demonstrate that increased P53 is unable to control parasite burden during NOX1 or TFR1 knockdown, or in the presence of ROS scavenging or when lipid peroxidation is blocked. Additionally, SLC7a11 inhibitors Erastin and Sorafenib reduce infection. Thus, blocking the host SLC7a11-GPX4 pathway serves to selectively elevate lipid peroxides in infected cells, which localize within the parasite and lead to the elimination of liver stage parasites.


Subject(s)
Amino Acid Transport System y+/metabolism , Lipid Peroxidation , Liver Diseases/metabolism , Liver Diseases/parasitology , Malaria/metabolism , Amino Acid Transport System y+/antagonists & inhibitors , Animals , Cell Line , Cells, Cultured , Ferroptosis , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 1/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Receptors, Transferrin/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism
5.
iScience ; 21: 603-611, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31731198

ABSTRACT

The invasion of a suitable host hepatocyte by Plasmodium sporozoites is an essential step in malaria infection. We demonstrate that in infected hepatocytes, lysosomes are redistributed away from the nucleus, and surface exposure of lysosome-associated membrane protein 1 (LAMP1) is increased. Lysosome exocytosis in infected cells occurs independently of sporozoite traversal. Instead, a sporozoite-secreted factor is sufficient for the process. Knockdown of SNARE proteins involved in lysosome-plasma membrane fusion reduces lysosome exocytosis and Plasmodium infection. In contrast, promoting fusion between the lysosome and plasma membrane dramatically increases infection. Our work demonstrates parallels between Plasmodium sporozoite entry of hepatocytes and infection by the excavate pathogen Trypanosoma cruzi and raises the question of whether convergent evolution has shaped host cell invasion by divergent pathogens.

6.
Cell Rep ; 26(12): 3391-3399.e4, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893610

ABSTRACT

Plasmodium parasites are highly selective when infecting hepatocytes and induce many changes within the host cell upon infection. While several host cell factors have been identified that are important for liver infection, our understanding of what facilitates the maintenance of infection remains incomplete. Here, we describe a role for phosphorylated ribosomal protein S6 (Ser235/236) (p-RPS6) in Plasmodium yoelii-infected hepatocytes. Blocking RPS6 phosphorylation prior to infection decreases the number of liver stage parasites within 24 h. Infected hepatocytes exhibit elevated levels of p-RPS6 while simultaneously abrogating the induction of phosphorylation of RPS6 in response to insulin stimulation. This is in contrast with the regulation of p-RPS6 by Toxoplasma gondii, which elevates levels of p-RPS6 after infection but does not alter the response to insulin. Our data support a model in which RPS6 phosphorylation is uncoupled from canonical regulators in Plasmodium-infected hepatocytes and is relied on by the parasite to maintain infection.


Subject(s)
Hepatocytes/metabolism , Malaria/metabolism , Plasmodium yoelii/metabolism , Ribosomal Protein S6/metabolism , Animals , Cell Line , Hepatocytes/parasitology , Hepatocytes/pathology , Humans , Malaria/pathology , Mice , Mice, Inbred BALB C , Phosphorylation , Toxoplasma/metabolism , Toxoplasmosis/metabolism , Toxoplasmosis/pathology
7.
Nat Commun ; 8(1): 1232, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29089541

ABSTRACT

Plasmodium parasites have extensive needs from their host hepatocytes during the obligate liver stage of infection, yet there remains sparse knowledge of specific host regulators. Here we assess 34 host-targeted kinase inhibitors for their capacity to eliminate Plasmodium yoelii-infected hepatocytes. Using pre-existing activity profiles of each inhibitor, we generate a predictive computational model that identifies host kinases, which facilitate Plasmodium yoelii liver stage infection. We predict 47 kinases, including novel and previously described kinases that impact infection. The impact of a subset of kinases is experimentally validated, including Receptor Tyrosine Kinases, members of the MAP Kinase cascade, and WEE1. Our approach also predicts host-targeted kinase inhibitors of infection, including compounds already used in humans. Three of these compounds, VX-680, Roscovitine and Sunitinib, each eliminate >85% of infection. Our approach is well-suited to uncover key host determinants of infection in difficult model systems, including field-isolated parasites and/or emerging pathogens.


Subject(s)
Liver/drug effects , Malaria/prevention & control , Plasmodium yoelii/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/parasitology , Host-Parasite Interactions/drug effects , Humans , Indoles/pharmacology , Liver/enzymology , Liver/parasitology , Malaria/enzymology , Malaria/parasitology , Mice , Piperazines/pharmacology , Plasmodium yoelii/physiology , Protein Kinases/genetics , Protein Kinases/metabolism , Purines/pharmacology , Pyrroles/pharmacology , RNA Interference , Roscovitine , Sporozoites/drug effects , Sporozoites/physiology , Sunitinib
8.
Sci Transl Med ; 9(371)2017 01 04.
Article in English | MEDLINE | ID: mdl-28053159

ABSTRACT

Immunization of humans with whole sporozoites confers complete, sterilizing immunity against malaria infection. However, achieving consistent safety while maintaining immunogenicity of whole parasite vaccines remains a formidable challenge. We generated a genetically attenuated Plasmodium falciparum (Pf) malaria parasite by deleting three genes expressed in the pre-erythrocytic stage (Pf p52-/p36-/sap1-). We then tested the safety and immunogenicity of the genetically engineered (Pf GAP3KO) sporozoites in human volunteers. Pf GAP3KO sporozoites were delivered to 10 volunteers using infected mosquito bites with a single exposure consisting of 150 to 200 bites per subject. All subjects remained blood stage-negative and developed inhibitory antibodies to sporozoites. GAP3KO rodent malaria parasites engendered complete, protracted immunity against infectious sporozoite challenge in mice. The results warrant further clinical testing of Pf GAP3KO and its potential development into a vaccine strain.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/genetics , Sporozoites/genetics , Adult , Animals , Antibodies, Protozoan/blood , Female , Gene Deletion , Genetic Engineering , Humans , Immunoglobulin G/blood , Malaria Vaccines/genetics , Male , Mice , Mice, Inbred BALB C , Middle Aged , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Sporozoites/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Young Adult
9.
Science ; 350(6264): 1089-92, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26612952

ABSTRACT

The invasion of a suitable host hepatocyte by mosquito-transmitted Plasmodium sporozoites is an essential early step in successful malaria parasite infection. Yet precisely how sporozoites target their host cell and facilitate productive infection remains largely unknown. We found that the hepatocyte EphA2 receptor was critical for establishing a permissive intracellular replication compartment, the parasitophorous vacuole. Sporozoites productively infected hepatocytes with high EphA2 expression, and the deletion of EphA2 protected mice from liver infection. Lack of host EphA2 phenocopied the lack of the sporozoite proteins P52 and P36. Our data suggest that P36 engages EphA2, which is likely to be a key step in establishing the permissive replication compartment.


Subject(s)
Hepatocytes/enzymology , Hepatocytes/parasitology , Malaria/enzymology , Malaria/parasitology , Plasmodium/physiology , Protozoan Proteins/metabolism , Receptor, EphA2/metabolism , Sporozoites/physiology , Animals , Anopheles/parasitology , Cell Line, Tumor , Humans , Malaria/genetics , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Plasmodium/genetics , Receptor, EphA2/genetics
10.
Cell Microbiol ; 17(12): 1848-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26118838

ABSTRACT

The bud emergence (BEM)46 proteins are evolutionarily conserved members of the α/ß-hydrolase superfamily, which includes enzymes with diverse functions and a wide range of substrates. Here, we identified a Plasmodium BEM46-like protein (PBLP) and characterized it throughout the life cycle of the rodent malaria parasite Plasmodium yoelii. The Plasmodium BEM46-like protein is shown to be closely associated with the parasite plasma membrane of asexual erythrocytic stage schizonts and exo-erythrocytic schizonts; however, PBLP localizes to unique intracellular structures in sporozoites. Generation and analysis of P. yoelii knockout (Δpblp) parasite lines showed that PBLP has an important role in erythrocytic stage merozoite development with Δpblp parasites forming fewer merozoites during schizogony, which results in decreased parasitemia when compared with wild-type (WT) parasites. Δpblp parasites showed no defects in gametogenesis or transmission to mosquitoes; however, because they formed fewer oocysts there was a reduction in the number of developed sporozoites in infected mosquitoes when compared with WT. Although Δpblp sporozoites showed no apparent defect in mosquito salivary gland infection, they showed decreased infectivity in hepatocytes in vitro. Similarly, mice infected with Δpblp sporozoites exhibited a delay in the onset of blood-stage patency, which is likely caused by reduced sporozoite infectivity and a discernible delay in exo-erythrocytic merozoite formation. These data are consistent with the model that PBLP has an important role in parasite invasive-stage morphogenesis throughout the parasite life cycle.


Subject(s)
Hydrolases/metabolism , Plasmodium yoelii/enzymology , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/enzymology , Culicidae , Gene Deletion , Hydrolases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Merozoites/enzymology , Merozoites/growth & development , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Plasmodium yoelii/genetics , Plasmodium yoelii/growth & development , Sporozoites/enzymology , Sporozoites/growth & development
11.
Mol Ther ; 23(5): 857-865, 2015 May.
Article in English | MEDLINE | ID: mdl-25648263

ABSTRACT

Eliminating malaria parasites during the asymptomatic but obligate liver stages (LSs) of infection would stop disease and subsequent transmission. Unfortunately, only a single licensed drug that targets all LSs, Primaquine, is available. Targeting host proteins might significantly expand the repertoire of prophylactic drugs against malaria. Here, we demonstrate that both Bcl-2 inhibitors and P53 agonists dramatically reduce LS burden in a mouse malaria model in vitro and in vivo by altering the activity of key hepatocyte factors on which the parasite relies. Bcl-2 inhibitors act primarily by inducing apoptosis in infected hepatocytes, whereas P53 agonists eliminate parasites in an apoptosis-independent fashion. In combination, Bcl-2 inhibitors and P53 agonists act synergistically to delay, and in some cases completely prevent, the onset of blood stage disease. Both families of drugs are highly effective at doses that do not cause substantial hepatocyte cell death in vitro or liver damage in vivo. P53 agonists and Bcl-2 inhibitors were also effective when administered to humanized mice infected with Plasmodium falciparum. Our data demonstrate that host-based prophylaxis could be developed into an effective intervention strategy that eliminates LS parasites before the onset of clinical disease and thus opens a new avenue to prevent malaria.


Subject(s)
Antimalarials/pharmacology , Life Cycle Stages/drug effects , Liver/parasitology , Malaria/parasitology , Plasmodium/drug effects , Plasmodium/physiology , Post-Exposure Prophylaxis , Animals , Antimalarials/administration & dosage , Cell Line , Disease Models, Animal , Female , Imidazoles/administration & dosage , Imidazoles/pharmacology , Indoles , Malaria/drug therapy , Malaria/metabolism , Malaria/prevention & control , Malaria, Falciparum/drug therapy , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Mice , Mice, Transgenic , Parasite Load , Piperazines/administration & dosage , Piperazines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/administration & dosage , Pyrroles/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...