Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 396
1.
Malar J ; 23(1): 151, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755636

BACKGROUND: Sporozoite invasion of hepatocytes is an essential step in the Plasmodium life-cycle and has similarities, at the cellular level, to merozoite invasion of erythrocytes. In the case of the Plasmodium blood-stage, efforts to identify host-pathogen protein-protein interactions have yielded important insights including vaccine candidates. In the case of sporozoite-hepatocyte invasion, the host-pathogen protein-protein interactions involved are poorly understood. METHODS: To gain a better understanding of the protein-protein interaction between the sporozoite ligands and host receptors, a systematic screen was performed. The previous Plasmodium falciparum and human surface protein ectodomain libraries were substantially extended, resulting in the creation of new libraries comprising 88 P. falciparum sporozoite protein coding sequences and 182 sequences encoding human hepatocyte surface proteins. Having expressed recombinant proteins from these sequences, a plate-based assay was used, capable of detecting low affinity interactions between recombinant proteins, modified for enhanced throughput, to screen the proteins for interactions. The novel interactions identified in the screen were characterized biochemically, and their essential role in parasite invasion was further elucidated using antibodies and genetically manipulated Plasmodium parasites. RESULTS: A total of 7540 sporozoite-hepatocyte protein pairs were tested under conditions capable of detecting interactions of at least 1.2 µM KD. An interaction between the human fibroblast growth factor receptor 4 (FGFR4) and the P. falciparum protein Pf34 is identified and reported here, characterizing its affinity and demonstrating the blockade of the interaction by reagents, including a monoclonal antibody. Furthermore, further interactions between Pf34 and a second P. falciparum rhoptry neck protein, PfRON6, and between human low-density lipoprotein receptor (LDLR) and the P. falciparum protein PIESP15 are identified. Conditional genetic deletion confirmed the essentiality of PfRON6 in the blood-stage, consistent with the important role of this protein in parasite lifecycle. Pf34 was refractory to attempted genetic modification. Antibodies to Pf34 abrogated the interaction and had a modest effect upon sporozoite invasion into primary human hepatocytes. CONCLUSION: Pf34 and PfRON6 may be members of a functionally important invasion complex which could be a target for future interventions. The modified interaction screening assay, protein expression libraries and P. falciparum mutant parasites reported here may be a useful tool for protein interaction discovery and antigen candidate screening which could be of wider value to the scientific community.


Hepatocytes , Plasmodium falciparum , Protozoan Proteins , Sporozoites , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Hepatocytes/parasitology , Humans , Sporozoites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Host-Pathogen Interactions , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Host-Parasite Interactions , Protein Binding
2.
Trends Parasitol ; 40(6): 466-476, 2024 Jun.
Article En | MEDLINE | ID: mdl-38714463

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.


Hepatocytes , Malaria , Plasmodium , Plasmodium/immunology , Plasmodium/physiology , Humans , Malaria/immunology , Malaria/parasitology , Hepatocytes/parasitology , Hepatocytes/immunology , Animals
3.
Infect Immun ; 92(6): e0002624, 2024 Jun 11.
Article En | MEDLINE | ID: mdl-38767360

Schistosomiasis is a serious public health problem, and previous studies found that liver function and hepatic cells are damaged. To evaluate the serum parameters of liver function and fibrosis in schistosomiasis patients infected with Schistosoma japonicum (Schistosoma J.) and analyze the correlations between liver function and serum fibrosis markers in patients infected with Schistosoma J., this retrospective study enrolled 133 patients. The study population was divided into four groups: healthy people control group (n = 20), chronic schistosomiasis without liver cirrhosis (CS) group (n = 21), schistosomiasis cirrhosis without hypoalbuminemia (SC-HA) group (n = 68), and schistosomiasis cirrhosis with hypoalbuminemia (SC +HA) group (n = 24). Clinical and laboratory data were collected for analysis. In the multiple comparison of abnormal rates of aspartate aminotransferase (AST) and total bilirubin (TBIL), the abnormal rate of the SC +HA group was significantly higher than that of the other three groups (P < 0.05), and the abnormal rate of γ-GT in the SC +HA group was significantly higher than that in the control group (P < 0.05). Multiple comparison results of serum levels of fibrosis markers showed that the SC group had a significantly higher level of indexes than other groups (P < 0.05). The levels of TGF-ß1 in the CS group, SC-HA group and SC +HA group were significantly higher than those in the control group (P < 0.001). Our study demonstrated that the liver function and hepatic cells were damaged with the progression of liver disease in patients infected with Schistosoma J., and they played an important role in the occurrence and development of liver fibrosis.


Hepatocytes , Liver Cirrhosis , Schistosoma japonicum , Schistosomiasis japonica , Humans , Liver Cirrhosis/pathology , Liver Cirrhosis/parasitology , Schistosomiasis japonica/complications , Schistosomiasis japonica/pathology , Male , Female , Middle Aged , Animals , Adult , Retrospective Studies , Hepatocytes/pathology , Hepatocytes/parasitology , Biomarkers/blood , Aged , Liver/pathology , Liver/parasitology , Liver Function Tests
4.
ACS Infect Dis ; 10(6): 1904-1913, 2024 Jun 14.
Article En | MEDLINE | ID: mdl-38752809

Malaria parasites have a complex life cycle and undergo replication and population expansion within vertebrate hosts and mosquito vectors. These developmental transitions rely on changes in gene expression and chromatin reorganization that result in the activation and silencing of stage-specific genes. The ApiAp2 family of DNA-binding proteins plays an important role in regulating gene expression in malaria parasites. Here, we characterized the ApiAp2 protein in Plasmodium berghei, which we termed Ap2-D. In silico analysis revealed that Ap2-D has three beta-sheets followed by a helix at the C-terminus for DNA binding. Using gene tagging with 3XHA-mCherry, we found that Ap2-D is expressed in Plasmodium blood stages and is present in the parasite cytoplasm and nucleus. Surprisingly, our gene deletion study revealed a completely dispensable role for Ap2-D in the entirety of the P. berghei life cycle. Ap2-D KO parasites were found to grow in the blood successfully and progress through the mosquito midgut and salivary glands. Sporozoites isolated from mosquito salivary glands were infective for hepatocytes and achieved similar patency as WT in mice. We emphasize the importance of genetic validation of antimalarial drug targets before progressing them to drug discovery.


Life Cycle Stages , Plasmodium berghei , Protozoan Proteins , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Animals , Mice , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Malaria/parasitology , Transcription Factors/genetics , Transcription Factors/metabolism , Sporozoites/growth & development , Sporozoites/metabolism , Sporozoites/physiology , Salivary Glands/parasitology , Mosquito Vectors/parasitology , Female , Anopheles/parasitology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hepatocytes/parasitology
5.
Mol Microbiol ; 121(6): 1095-1111, 2024 Jun.
Article En | MEDLINE | ID: mdl-38574236

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.


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
6.
ACS Infect Dis ; 10(4): 1116-1125, 2024 04 12.
Article En | MEDLINE | ID: mdl-38421807

The O-fucosylation of the thrombospondin type I repeat (TSR) domain is important for TSR-containing proteins' optimal folding and stability. However, the importance of Plasmodium O-fucosyltransferase 2 (POFut2) remains unclear due to two different reports. Here, we disrupted the POFut2 gene in Plasmodium berghei and demonstrated that POFut2 KO parasites develop normally in blood and mosquito stages but show reduced infectivity in mice. We found that the reduced infectivity of POFut2 KO sporozoites was due to a diminished level of TRAP that affected the parasite gliding motility and hepatocyte infectivity. Using all-atom MD simulation, we also hypothesize that O-fucosylation impacts the TSR domain's stability more than its heparin binding capacity.


Fucosyltransferases , Plasmodium berghei , Animals , Mice , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Plasmodium berghei/genetics , Sporozoites , Protozoan Proteins/metabolism , Hepatocytes/parasitology
7.
Int J Antimicrob Agents ; 63(5): 107112, 2024 May.
Article En | MEDLINE | ID: mdl-38367843

The control and elimination of malaria caused by Plasmodium vivax is hampered by the threat of relapsed infection resulting from the activation of dormant hepatic hypnozoites. Currently, only the 8-aminoquinolines, primaquine and tafenoquine, have been approved for the elimination of hypnozoites, although their use is hampered by potential toxicity. Therefore, an alternative radical curative drug that safely eliminates hypnozoites is a pressing need. This study assessed the potential hypnozoiticidal activity of the antibiotic azithromycin, which is thought to exert antimalarial activity by inhibiting prokaryote-like ribosomal translation within the apicoplast, an indispensable organelle. The results show that azithromycin inhibited apicoplast development during liver-stage schizogony in P. vivax and Plasmodium cynomolgi, leading to impaired parasite maturation. More importantly, this study found that azithromycin is likely to impair the hypnozoite's apicoplast, resulting in the loss of this organelle. Subsequently, using a recently developed long-term hepatocyte culture system, this study found that this loss likely induces a delay in the hypnozoite activation rate, and that those parasites that do proceed to schizogony display liver-stage arrest prior to differentiating into hepatic merozoites, thus potentially preventing relapse. Overall, this work provides evidence for the potential use of azithromycin for the radical cure of relapsing malaria, and identifies apicoplast functions as potential drug targets in quiescent hypnozoites.


Antimalarials , Apicoplasts , Azithromycin , Liver , Plasmodium cynomolgi , Plasmodium vivax , Azithromycin/pharmacology , Plasmodium vivax/drug effects , Plasmodium cynomolgi/drug effects , Antimalarials/pharmacology , Liver/parasitology , Liver/drug effects , Apicoplasts/drug effects , Animals , Hepatocytes/parasitology , Hepatocytes/drug effects , Humans , Organelle Biogenesis , Malaria, Vivax/parasitology , Malaria, Vivax/drug therapy , Mice , Malaria/parasitology , Malaria/drug therapy
8.
Acta Parasitol ; 69(1): 700-709, 2024 Mar.
Article En | MEDLINE | ID: mdl-38372909

Cystic Echinococcosis (CE) is a zoonotic infection caused by the larval form of Echinococcus granulosus in humans. Emerging evidence suggests an intriguing inverse association between E. granulosus infection and the occurrence of cancer. This study aimed to investigate the influence of diverse host-derived hydatid cyst fluids (HCF) with distinct genotypes on human liver hepatocytes (HC) and hepatocellular carcinoma cells (HepG2). Specifically, we examined their effects on cell proliferation, apoptosis sensitivity (BAX/BCL-2), apoptosis-related p53 expression, and the expression of cancer-related microRNA (hsa-miR-181b-3p). Cell proliferation assays, real-time PCR, and ELISA studies were conducted to evaluate potential anti-cancer properties. The findings revealed that animal-origin HCF (G1(A)) induced direct cell death by augmenting the susceptibility of HepG2 cells to apoptosis. Treatment with both G1(A) and G1(H) HCF sensitized HepG2 and HC cell lines to apoptosis by modulating the BAX/BCL-2 ratio, accompanied by upregulation of the p53 gene. Additionally, G1(A) HCF and human-derived HCFs (G1(H), G7(H)) reduced the expression of miR-181b-3p in HepG2 cells. Consequently, this study demonstrates the potential anti-cancer effect of HCF in HepG2 cells and provides the first comparative assessment of HCFs from human and animal sources with diverse genotypes, offering novel insights into this field.


Apoptosis , Carcinoma, Hepatocellular , Hepatocytes , Humans , Apoptosis/drug effects , Hepatocytes/parasitology , Hep G2 Cells , Carcinoma, Hepatocellular/parasitology , Liver Neoplasms/parasitology , Cyst Fluid/chemistry , Animals , Echinococcosis/parasitology , Cell Proliferation/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Echinococcus granulosus/genetics , Echinococcus granulosus/drug effects
9.
Mol Microbiol ; 121(3): 328-340, 2024 03.
Article En | MEDLINE | ID: mdl-37602900

An essential step in the life cycle of malaria parasites is their egress from hepatocytes, which enables the transition from the asymptomatic liver stage to the pathogenic blood stage of infection. To exit the liver, Plasmodium parasites first disrupt the parasitophorous vacuole membrane that surrounds them during their intracellular replication. Subsequently, parasite-filled structures called merosomes emerge from the infected cell. Shrouded by host plasma membrane, like in a Trojan horse, parasites enter the vasculature undetected by the host immune system and travel to the lung where merosomes rupture, parasites are released, and the blood infection stage begins. This complex, multi-step process must be carefully orchestrated by the parasite and requires extensive manipulation of the infected host cell. This review aims to outline the known signaling pathways that trigger exit, highlight Plasmodium proteins that contribute to the release of liver-stage merozoites, and summarize the accompanying changes to the hepatic host cell.


Malaria , Parasites , Plasmodium , Animals , Humans , Parasites/metabolism , Liver/parasitology , Hepatocytes/parasitology , Plasmodium/metabolism , Malaria/parasitology , Erythrocytes/parasitology , Protozoan Proteins/metabolism
10.
mSphere ; 8(6): e0054423, 2023 Dec 20.
Article En | MEDLINE | ID: mdl-37909773

IMPORTANCE: Plasmodium parasites cause malaria in humans. New multistage active antimalarial drugs are needed, and a promising class of drugs targets the core cellular process of translation, which has many potential molecular targets. During the obligate liver stage, Plasmodium parasites grow in metabolically active hepatocytes, making it challenging to study core cellular processes common to both host cells and parasites, as the signal from the host typically overwhelms that of the parasite. Here, we present and validate a flexible assay to quantify Plasmodium liver stage translation using a technique to fluorescently label the newly synthesized proteins of both host and parasite followed by computational separation of their respective nascent proteomes in confocal image sets. We use the assay to determine whether a test set of known compounds are direct or indirect liver stage translation inhibitors and show that the assay can also predict the mode of action for novel antimalarial compounds.


Antimalarials , Malaria , Parasites , Animals , Humans , Plasmodium berghei , Liver/parasitology , Hepatocytes/parasitology , Malaria/parasitology , Antimalarials/pharmacology , Antimalarials/metabolism
11.
Proc Biol Sci ; 290(2011): 20232280, 2023 Nov 29.
Article En | MEDLINE | ID: mdl-38018100

Vaccination strategies in mice inducing high numbers of memory CD8+ T cells specific to a single epitope are able to provide sterilizing protection against infection with Plasmodium sporozoites. We have recently found that Plasmodium-specific CD8+ T cells cluster around sporozoite-infected hepatocytes but whether such clusters are important in elimination of the parasite remains incompletely understood. Here, we used our previously generated data in which we employed intravital microscopy to longitudinally image 32 green fluorescent protein (GFP)-expressing Plasmodium yoelii parasites in livers of mice that had received activated Plasmodium-specific CD8+ T cells after sporozoite infection. We found significant heterogeneity in the dynamics of the normalized GFP signal from the parasites (termed 'vitality index' or VI) that was weakly correlated with the number of T cells near the parasite. We also found that a simple model assuming mass-action, additive killing by T cells well describes the VI dynamics for most parasites and predicts a highly variable killing efficacy by individual T cells. Given our estimated median per capita kill rate of k = 0.031/h we predict that a single T cell is typically incapable of killing a parasite within the 48 h lifespan of the liver stage in mice. Stochastic simulations of T cell clustering and killing of the liver stage also suggested that: (i) three or more T cells per infected hepatocyte are required to ensure sterilizing protection; (ii) both variability in killing efficacy of individual T cells and resistance to killing by individual parasites may contribute to the observed variability in VI decline, and (iii) the stable VI of some clustered parasites cannot be explained by measurement noise. Taken together, our analysis for the first time provides estimates of efficiency at which individual CD8+ T cells eliminate intracellular parasitic infection in vivo.


Malaria , Plasmodium yoelii , Mice , Animals , CD8-Positive T-Lymphocytes , Liver/parasitology , Hepatocytes/parasitology , Sporozoites , Plasmodium berghei/metabolism
12.
Mol Biochem Parasitol ; 256: 111589, 2023 12.
Article En | MEDLINE | ID: mdl-37604406

Plasmodium sporozoites can block apoptotic pathways within host hepatocytes, ensuring the survival of the parasite. However, attenuated plasmodial sporozoites are unable to prevent apoptosis, which provides many parasite antigens to immune cells. This exposure leads to protection against Malaria in both human and animal models. If these hosts are later inoculated with infectious sporozoites, apoptosis of infected hepatocytes will occur, preventing parasite development. Considering that hydrogen peroxide can induce apoptosis, it is plausible that it plays a role in the mechanisms associated with the protection mediated by attenuated plasmodial sporozoites. Based on published results that describe the relationship between Plasmodium, hydrogen peroxide, and apoptosis, a rational explanation can be provided for this hypothesis.


Malaria Vaccines , Malaria , Plasmodium , Animals , Humans , Sporozoites , Hydrogen Peroxide/pharmacology , Malaria/prevention & control , Malaria/parasitology , Hepatocytes/parasitology
13.
Trends Parasitol ; 39(10): 808-811, 2023 10.
Article En | MEDLINE | ID: mdl-37574429

Attenuated plasmodial sporozoite-induced immune response includes intrahepatic nitric oxide (NO) production, which promotes apoptosis of infected hepatocytes and consequent parasite clearance. NO in excess reacts with superoxide, forming peroxynitrite, a powerful cytotoxic agent. Here, I suggest that peroxynitrite proapoptotic action may contribute to the attenuated malarial sporozoite-mediated protection.


Malaria Vaccines , Malaria , Plasmodium , Animals , Sporozoites , Peroxynitrous Acid , Malaria/prevention & control , Hepatocytes/parasitology , Nitric Oxide
14.
Cell Rep ; 42(7): 112727, 2023 07 25.
Article En | MEDLINE | ID: mdl-37392389

Dormancy enables relapsing malaria parasites, such as Plasmodium vivax and cynomolgi, to survive unfavorable conditions. It is enabled by hypnozoites, parasites remaining quiescent inside hepatocytes before reactivating and establishing blood-stage infection. We integrate omics approaches to explore gene-regulatory mechanisms underlying hypnozoite dormancy. Genome-wide profiling of activating and repressing histone marks identifies a few genes that get silenced by heterochromatin during hepatic infection of relapsing parasites. By combining single-cell transcriptomics, chromatin accessibility profiling, and fluorescent in situ RNA hybridization, we show that these genes are expressed in hypnozoites and that their silencing precedes parasite development. Intriguingly, these hypnozoite-specific genes mainly encode proteins with RNA-binding domains. We hence hypothesize that these likely repressive RNA-binding proteins keep hypnozoites in a developmentally competent but dormant state and that heterochromatin-mediated silencing of the corresponding genes aids reactivation. Exploring the regulation and exact function of these proteins hence could provide clues for targeted reactivation and killing of these latent pathogens.


Malaria , Plasmodium cynomolgi , Humans , Heterochromatin , Plasmodium cynomolgi/genetics , Malaria/parasitology , Hepatocytes/parasitology , Gene Expression Profiling
15.
mSphere ; 8(4): e0058722, 2023 08 24.
Article En | MEDLINE | ID: mdl-37272704

During invasion, Plasmodium parasites secrete proteins from rhoptry and microneme apical end organelles, which have crucial roles in attaching to and invading target cells. A sporozoite stage-specific gene silencing system revealed that rhoptry neck protein 2 (RON2), RON4, and RON5 are important for sporozoite invasion of mosquito salivary glands. Here, we further investigated the roles of RON4 during sporozoite infection of the liver in vivo. Following intravenous inoculation of RON4-knockdown sporozoites into mice, we demonstrated that sporozoite RON4 has multiple functions during sporozoite traversal of sinusoidal cells and infection of hepatocytes. In vitro infection experiments using a hepatoma cell line revealed that secreted RON4 is involved in sporozoite adhesion to hepatocytes and has an important role in the early steps of hepatocyte infection. In addition, in vitro motility assays indicated that RON4 is required for sporozoite attachment to the substrate and the onset of migration. These findings indicate that RON4 is crucial for sporozoite migration toward and invasion of hepatocytes via attachment ability and motility.IMPORTANCEMalarial parasite transmission to mammals is established when sporozoites are inoculated by mosquitoes and migrate through the bloodstream to infect hepatocytes. Many aspects of the molecular mechanisms underpinning migration and cellular invasion remain largely unelucidated. By applying a sporozoite stage-specific gene silencing system in the rodent malarial parasite, Plasmodium berghei, we demonstrated that rhoptry neck protein 4 (RON4) is crucial for sporozoite infection of the liver in vivo. Combined with in vitro investigations, it was revealed that RON4 functions during a crossing of the sinusoidal cell layer and invading hepatocytes, at an early stage of liver infection, by mediating the sporozoite capacity for adhesion and the onset of motility. Since RON4 is also expressed in Plasmodium merozoites and Toxoplasma tachyzoites, our findings contribute to understanding the conserved invasion mechanisms of Apicomplexa parasites.


Malaria , Plasmodium berghei , Sporozoites , Animals , Mice , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria/metabolism , Malaria/parasitology , Malaria/pathology , Sporozoites/physiology , Protozoan Proteins/metabolism , Hepatocytes/metabolism , Hepatocytes/parasitology , Hepatocytes/pathology
16.
PLoS One ; 18(3): e0279144, 2023.
Article En | MEDLINE | ID: mdl-36928885

Early Plasmodium falciparum and P. vivax infection requires parasite replication within host hepatocytes, referred to as liver stage (LS). However, limited understanding of infection dynamics in human LS exists due to species-specificity challenges. Reported here is a reproducible, easy-to-manipulate, and moderate-cost in vivo model to study human Plasmodium LS in mice; the ectopic huLiver model. Ectopic huLiver tumors were generated through subcutaneous injection of the HC-04 cell line and shown to be infectible by both freshly dissected sporozoites and through the bite of infected mosquitoes. Evidence for complete LS development was supported by the transition to blood-stage infection in mice engrafted with human erythrocytes. Additionally, this model was successfully evaluated for its utility in testing antimalarial therapeutics, as supported by primaquine acting as a causal prophylactic against P. falciparum. Presented here is a new platform for the study of human Plasmodium infection with the potential to aid in drug discovery.


Communicable Diseases , Liver Diseases , Malaria, Falciparum , Malaria, Vivax , Malaria , Plasmodium , Mice , Animals , Humans , Liver/parasitology , Malaria/drug therapy , Malaria, Falciparum/parasitology , Hepatocytes/parasitology , Plasmodium falciparum , Sporozoites
17.
Trends Parasitol ; 39(5): 319-320, 2023 05.
Article En | MEDLINE | ID: mdl-36931922

Productive invasion of hepatocytes by Plasmodium sporozoites is a key step of infection. The parasites traverse hepatocytes before targeting one of them to form a parasitophorous vacuole for parasite expansion. Schepis et al. show the induction of membrane ruffling via host Rho GTPases by Plasmodium sporozoites facilitating productive invasion.


Malaria , Parasites , Plasmodium , Animals , Actins , Malaria/parasitology , Hepatocytes/parasitology , Sporozoites , Plasmodium berghei , Protozoan Proteins
18.
Parasitol Int ; 93: 102700, 2023 Apr.
Article En | MEDLINE | ID: mdl-36403748

Plasmodium sporozoites travel a long way from the site where they are released by a mosquito bite to the liver, where they infect hepatocytes and develop into erythrocyte-invasive forms. The success of this infection depends on the ability of the sporozoites to correctly recognize the hepatocyte as a target and change their behavior from migration to infection. However, how this change is accomplished remains incompletely understood. In this paper, we report that 6-cysteine protein family members expressed in sporozoites including B9 are responsible for this ability. Experiments on parasites using double knockouts of B9 and SPECT2, which is essential for sporozoite to migrate through the hepatocyte, showed that the parasites lacked the capacity to stop migration. This finding suggests that interactions between these parasite proteins and hepatocyte-specific cell surface ligands mediate correct recognition of hepatocytes by sporozoites, which is an essential step in malaria transmission to humans.


Liver Diseases , Plasmodium , Humans , Animals , Sporozoites , Cysteine , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Hepatocytes/parasitology
19.
Nature ; 612(7940): 534-539, 2022 12.
Article En | MEDLINE | ID: mdl-36477528

An effective vaccine is needed for the prevention and elimination of malaria. The only immunogens that have been shown to have a protective efficacy of more than 90% against human malaria are Plasmodium falciparum (Pf) sporozoites (PfSPZ) manufactured in mosquitoes (mPfSPZ)1-7. The ability to produce PfSPZ in vitro (iPfSPZ) without mosquitoes would substantially enhance the production of PfSPZ vaccines and mosquito-stage malaria research, but this ability is lacking. Here we report the production of hundreds of millions of iPfSPZ. iPfSPZ invaded human hepatocytes in culture and developed to mature liver-stage schizonts expressing P. falciparum merozoite surface protein 1 (PfMSP1) in numbers comparable to mPfSPZ. When injected into FRGhuHep mice containing humanized livers, iPfSPZ invaded the human hepatocytes and developed to PfMSP1-expressing late liver stage parasites at 45% the quantity of cryopreserved mPfSPZ. Human blood from FRGhuHep mice infected with iPfSPZ produced asexual and sexual erythrocytic-stage parasites in culture, and gametocytes developed to PfSPZ when fed to mosquitoes, completing the P. falciparum life cycle from infectious gametocyte to infectious gametocyte without mosquitoes or primates.


Plasmodium falciparum , Sporozoites , Animals , Humans , Mice , Culicidae/parasitology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/biosynthesis , Malaria Vaccines/chemistry , Malaria, Falciparum/parasitology , Plasmodium falciparum/growth & development , Sporozoites/growth & development , Sporozoites/pathogenicity , Hepatocytes/parasitology , Liver/parasitology , Merozoite Surface Protein 1 , Erythrocytes/parasitology , In Vitro Techniques
20.
Malar J ; 21(1): 393, 2022 Dec 23.
Article En | MEDLINE | ID: mdl-36564750

BACKGROUND: The zoonotic simian parasite Plasmodium cynomolgi develops into replicating schizonts and dormant hypnozoites during the infection of hepatocytes and is used as a model organism to study relapsing malaria. The transcriptional profiling of P. cynomolgi liver stages was previously reported and revealed many important biological features of the parasite but left out the host response to malaria infection. METHODS: Previously published RNA sequencing data were used to quantify the expression of host genes in rhesus macaque hepatocytes infected with P. cynomolgi in comparison to either cells from uninfected samples or uninfected bystander cells. RESULTS: Although the dataset could not be used to resolve the transcriptional profile of hypnozoite-infected hepatocytes, it provided a snapshot of the host response to liver stage schizonts at 9-10 day post-infection and identified specific host pathways that are modulated during the exo-erythrocytic stage of P. cynomolgi. CONCLUSIONS: This study constitutes a valuable resource characterizing the hepatocyte response to P. cynomolgi infection and provides a framework to build on future research that aims at understanding hepatocyte-parasite interactions during relapsing malaria infection.


Malaria , Parasites , Plasmodium cynomolgi , Animals , Plasmodium cynomolgi/genetics , Macaca mulatta/parasitology , Hepatocytes/parasitology , Malaria/parasitology , Liver/parasitology
...