Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 153(5): 1120-33, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23683579

ABSTRACT

Cell-cell communication is an important mechanism for information exchange promoting cell survival for the control of features such as population density and differentiation. We determined that Plasmodium falciparum-infected red blood cells directly communicate between parasites within a population using exosome-like vesicles that are capable of delivering genes. Importantly, communication via exosome-like vesicles promotes differentiation to sexual forms at a rate that suggests that signaling is involved. Furthermore, we have identified a P. falciparum protein, PfPTP2, that plays a key role in efficient communication. This study reveals a previously unidentified pathway of P. falciparum biology critical for survival in the host and transmission to mosquitoes. This identifies a pathway for the development of agents to block parasite transmission from the human host to the mosquito.


Subject(s)
Cell Communication , Erythrocytes/pathology , Erythrocytes/parasitology , Malaria, Falciparum/pathology , Malaria, Falciparum/parasitology , Plasmodium falciparum/physiology , Actins/antagonists & inhibitors , Animals , Culicidae/parasitology , Drug Resistance , Exosomes/parasitology , Humans , Microtubules/drug effects , Plasmids/genetics , Plasmodium falciparum/growth & development , Signal Transduction , Trophozoites/physiology
2.
PLoS Biol ; 21(4): e3002066, 2023 04.
Article in English | MEDLINE | ID: mdl-37053271

ABSTRACT

With emerging resistance to frontline treatments, it is vital that new antimalarial drugs are identified to target Plasmodium falciparum. We have recently described a compound, MMV020291, as a specific inhibitor of red blood cell (RBC) invasion, and have generated analogues with improved potency. Here, we generated resistance to MMV020291 and performed whole genome sequencing of 3 MMV020291-resistant populations. This revealed 3 nonsynonymous single nucleotide polymorphisms in 2 genes; 2 in profilin (N154Y, K124N) and a third one in actin-1 (M356L). Using CRISPR-Cas9, we engineered these mutations into wild-type parasites, which rendered them resistant to MMV020291. We demonstrate that MMV020291 reduces actin polymerisation that is required by the merozoite stage parasites to invade RBCs. Additionally, the series inhibits the actin-1-dependent process of apicoplast segregation, leading to a delayed death phenotype. In vitro cosedimentation experiments using recombinant P. falciparum proteins indicate that potent MMV020291 analogues disrupt the formation of filamentous actin in the presence of profilin. Altogether, this study identifies the first compound series interfering with the actin-1/profilin interaction in P. falciparum and paves the way for future antimalarial development against the highly dynamic process of actin polymerisation.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Plasmodium falciparum/metabolism , Actins/genetics , Actins/metabolism , Profilins/genetics , Profilins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Malaria, Falciparum/genetics , Erythrocytes/parasitology , Antimalarials/pharmacology
3.
Mol Microbiol ; 121(3): 529-542, 2024 03.
Article in English | MEDLINE | ID: mdl-38131156

ABSTRACT

An essential process in transmission of the malaria parasite to the Anopheles vector is the conversion of mature gametocytes into gametes within the mosquito gut, where they egress from the red blood cell (RBC). During egress, male gametocytes undergo exflagellation, leading to the formation of eight haploid motile microgametes, while female gametes retain their spherical shape. Gametocyte egress depends on sequential disruption of the parasitophorous vacuole membrane and the host cell membrane. In other life cycle stages of the malaria parasite, phospholipases have been implicated in membrane disruption processes during egress, however their importance for gametocyte egress is relatively unknown. Here, we performed comprehensive functional analyses of six putative phospholipases for their role during development and egress of Plasmodium falciparum gametocytes. We localize two of them, the prodrug activation and resistance esterase (PF3D7_0709700) and the lysophospholipase 1 (PF3D7_1476700), to the parasite plasma membrane. Subsequently, we show that disruption of most of the studied phospholipase genes does neither affect gametocyte development nor egress. The exception is the putative patatin-like phospholipase 3 (PF3D7_0924000), whose gene deletion leads to a delay in male gametocyte exflagellation, indicating an important, albeit not essential, role of this enzyme in male gametogenesis.


Subject(s)
Malaria , Plasmodium falciparum , Animals , Male , Female , Phospholipases/genetics , Mosquito Vectors , Erythrocytes/parasitology
4.
Cell Mol Life Sci ; 80(3): 74, 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36847896

ABSTRACT

Plasmodium falciparum and P. vivax are the major causes of human malaria, and P. knowlesi is an important additional cause in SE Asia. Binding of apical membrane antigen 1 (AMA1) to rhoptry neck protein 2 (RON2) was thought to be essential for merozoite invasion of erythrocytes by Plasmodium spp. Our findings reveal that P. falciparum and P. vivax have diverged and show species-specific binding of AMA1 to RON2, determined by a ß-hairpin loop in RON2 and specific residues in AMA1 Loop1E. In contrast, cross-species binding of AMA1 to RON2 is retained between P. vivax and P. knowlesi. Mutation of specific amino acids in AMA1 Loop1E in P. falciparum or P. vivax ablated RON2 binding without impacting erythrocyte invasion. This indicates that the AMA1-RON2-loop interaction is not essential for invasion and additional AMA1 interactions are involved. Mutations in AMA1 that disrupt RON2 binding also enable escape of invasion inhibitory antibodies. Therefore, vaccines and therapeutics will need to be broader than targeting only the AMA1-RON2 interaction. Antibodies targeting AMA1 domain 3 had greater invasion-inhibitory activity when RON2-loop binding was ablated, suggesting this domain is a promising additional target for vaccine development. Targeting multiple AMA1 interactions involved in invasion may enable vaccines that generate more potent inhibitory antibodies and address the capacity for immune evasion. Findings on specific residues for invasion function and species divergence and conservation can inform novel vaccines and therapeutics against malaria caused by three species, including the potential for cross-species vaccines.


Subject(s)
Erythrocytes , Malaria , Membrane Proteins , Protozoan Proteins , Humans , Cell Membrane/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Malaria/genetics , Malaria/metabolism , Malaria/parasitology , Malaria/prevention & control , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
5.
J Bacteriol ; 205(12): e0032023, 2023 12 19.
Article in English | MEDLINE | ID: mdl-37991380

ABSTRACT

IMPORTANCE: Bacterial pathogens have vastly distinct sites that they inhabit during infection. This requires adaptation due to changes in nutrient availability and antimicrobial stress. The bacterial surface is a primary barrier, and here, we show that the bacterial pathogen Shigella flexneri increases its surface decorations when it transitions to an intracellular lifestyle. We also observed changes in bacterial and host cell fatty acid homeostasis. Specifically, intracellular S. flexneri increased the expression of their fatty acid degradation pathway, while the host cell lipid pool was significantly depleted. Importantly, bacterial proliferation could be inhibited by fatty acid supplementation of host cells, thereby providing novel insights into the possible link between human malnutrition and susceptibility to S. flexneri.


Subject(s)
Bacterial Proteins , Shigella flexneri , Humans , Bacterial Proteins/metabolism , Shigella flexneri/metabolism , Fatty Acids/metabolism , Lipids
6.
J Biol Chem ; 298(9): 102360, 2022 09.
Article in English | MEDLINE | ID: mdl-35961464

ABSTRACT

Malaria is responsible for hundreds of thousands of deaths every year. The lack of an effective vaccine and the global spread of multidrug resistant parasites hampers the fight against the disease and underlines the need for new antimalarial drugs. Central to the pathogenesis of malaria is the proliferation of Plasmodium parasites within human erythrocytes. Parasites invade erythrocytes via a coordinated sequence of receptor-ligand interactions between the parasite and the host cell. Posttranslational modifications such as protein phosphorylation are known to be key regulators in this process and are mediated by protein kinases. For several parasite kinases, including the Plasmodium falciparum glycogen synthase kinase 3 (PfGSK3), inhibitors have been shown to block erythrocyte invasion. Here, we provide an assessment of PfGSK3 function by reverse genetics. Using targeted gene disruption, we show the active gene copy, PfGSK3ß, is not essential for asexual blood stage proliferation, although it modulates efficient erythrocyte invasion. We found functional inactivation leads to a 69% decreased growth rate and confirmed this growth defect by rescue experiments with wildtype and catalytically inactive mutants. Functional knockout of PfGSK3ß does not lead to transcriptional upregulation of the second copy of PfGSK3. We further analyze expression, localization, and function of PfGSK3ß during gametocytogenesis using a parasite line allowing conditional induction of sexual commitment. We demonstrate PfGSK3ß-deficient gametocytes show a strikingly malformed morphology leading to the death of parasites in later stages of gametocyte development. Taken together, these findings are important for our understanding and the development of PfGSK3 as an antimalarial target.


Subject(s)
Antimalarials , Malaria, Falciparum , Antimalarials/pharmacology , Erythrocytes/metabolism , Glycogen Synthase Kinase 3/genetics , Humans , Ligands , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
7.
J Antimicrob Chemother ; 76(8): 1955-1961, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33956974

ABSTRACT

Macrolide antibiotics are categorized by the WHO as Highest Priority, Critically Important Antimicrobials due to their recommendation as treatment for severe cases of campylobacteriosis in humans; a self-limiting, rarely life-threatening, zoonotic foodborne infection. Low rates of macrolide resistance in Campylobacter jejuni and the availability of alternative treatments have prompted some regulatory schemes to assign macrolides to a lower importance category. Apart from rare, specific infections, macrolides largely play a supportive role to other drug classes in human medicine. By contrast, although the advent of alternative control methods has seen significant reductions in macrolide use in intensive livestock, they still have a crucial role in the treatment/control of respiratory infections and liver abscesses in cattle. Whilst acknowledging that ongoing surveillance is required to reduce the spread of recently emerged, transferable macrolide resistance among Campylobacter, this article recommends that macrolides should be moved to the WHO Highly Important category.


Subject(s)
Anti-Infective Agents , Campylobacter Infections , Campylobacter jejuni , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Campylobacter Infections/drug therapy , Campylobacter Infections/veterinary , Cattle , Drug Resistance, Bacterial , Humans , Macrolides/therapeutic use , Microbial Sensitivity Tests , World Health Organization
8.
Bioorg Chem ; 117: 105359, 2021 12.
Article in English | MEDLINE | ID: mdl-34689083

ABSTRACT

Malaria is a devastating disease caused by Plasmodium parasites. Emerging resistance against current antimalarial therapeutics has engendered the need to develop antimalarials with novel structural classes. We recently described the identification and initial optimization of the 2-anilino quinazoline antimalarial class. Here, we refine the physicochemical properties of this antimalarial class with the aim to improve aqueous solubility and metabolism and to reduce adverse promiscuity. We show the physicochemical properties of this class are intricately balanced with asexual parasite activity and human cell cytotoxicity. Structural modifications we have implemented improved LipE, aqueous solubility and in vitro metabolism while preserving fast acting P. falciparum asexual stage activity. The lead compounds demonstrated equipotent activity against P. knowlesi parasites and were not predisposed to resistance mechanisms of clinically used antimalarials. The optimized compounds exhibited modest activity against early-stage gametocytes, but no activity against pre-erythrocytic liver parasites. Confoundingly, the refined physicochemical properties installed in the compounds did not engender improved oral efficacy in a P. berghei mouse model of malaria compared to earlier studies on the 2-anilino quinazoline class. This study provides the framework for further development of this antimalarial class.


Subject(s)
Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Malaria/drug therapy , Plasmodium/drug effects , Quinazolines/chemistry , Quinazolines/pharmacology , Amination , Aniline Compounds/therapeutic use , Animals , Antimalarials/therapeutic use , Female , Humans , Malaria/parasitology , Mice , Plasmodium/physiology , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Quinazolines/therapeutic use
9.
Bioorg Chem ; 115: 105244, 2021 10.
Article in English | MEDLINE | ID: mdl-34452759

ABSTRACT

Malaria is a devastating parasitic disease caused by parasites from the genus Plasmodium. Therapeutic resistance has been reported against all clinically available antimalarials, threatening our ability to control the disease and therefore there is an ongoing need for the development of novel antimalarials. Towards this goal, we identified the 2-(N-phenyl carboxamide) triazolopyrimidine class from a high throughput screen of the Janssen Jumpstarter library against the asexual stages of the P. falciparum parasite. Here we describe the structure activity relationship of the identified class and the optimisation of asexual stage activity while maintaining selectivity against the human HepG2 cell line. The most potent analogues from this study were shown to exhibit equipotent activity against P. falciparum multidrug resistant strains and P. knowlesi asexual parasites. Asexual stage phenotyping studies determined the triazolopyrimidine class arrests parasites at the trophozoite stage, but it is likely these parasites are still metabolically active until the second asexual cycle, and thus have a moderate to slow onset of action. Non-NADPH dependent degradation of the central carboxamide and low aqueous solubility was observed in in vitro ADME profiling. A significant challenge remains to correct these liabilities for further advancement of the 2-(N-phenyl carboxamide) triazolopyrimidine scaffold as a potential moderate to slow acting partner in a curative or prophylactic antimalarial treatment.


Subject(s)
Antimalarials/pharmacology , Erythrocytes/drug effects , Plasmodium falciparum/drug effects , Plasmodium knowlesi/drug effects , Purines/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Erythrocytes/parasitology , Humans , Molecular Structure , Parasitic Sensitivity Tests , Purines/chemical synthesis , Purines/chemistry , Structure-Activity Relationship
10.
BMC Biol ; 18(1): 133, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32993629

ABSTRACT

BACKGROUND: Resistance to front-line antimalarials (artemisinin combination therapies) is spreading, and development of new drug treatment strategies to rapidly kill Plasmodium spp. malaria parasites is urgently needed. Azithromycin is a clinically used macrolide antibiotic proposed as a partner drug for combination therapy in malaria, which has also been tested as monotherapy. However, its slow-killing 'delayed-death' activity against the parasite's apicoplast organelle and suboptimal activity as monotherapy limit its application as a potential malaria treatment. Here, we explore a panel of azithromycin analogues and demonstrate that chemical modifications can be used to greatly improve the speed and potency of antimalarial action. RESULTS: Investigation of 84 azithromycin analogues revealed nanomolar quick-killing potency directed against the very earliest stage of parasite development within red blood cells. Indeed, the best analogue exhibited 1600-fold higher potency than azithromycin with less than 48 hrs treatment in vitro. Analogues were effective against zoonotic Plasmodium knowlesi malaria parasites and against both multi-drug and artemisinin-resistant Plasmodium falciparum lines. Metabolomic profiles of azithromycin analogue-treated parasites suggested activity in the parasite food vacuole and mitochondria were disrupted. Moreover, unlike the food vacuole-targeting drug chloroquine, azithromycin and analogues were active across blood-stage development, including merozoite invasion, suggesting that these macrolides have a multi-factorial mechanism of quick-killing activity. The positioning of functional groups added to azithromycin and its quick-killing analogues altered their activity against bacterial-like ribosomes but had minimal change on 'quick-killing' activity. Apicoplast minus parasites remained susceptible to both azithromycin and its analogues, further demonstrating that quick-killing is independent of apicoplast-targeting, delayed-death activity. CONCLUSION: We show that azithromycin and analogues can rapidly kill malaria parasite asexual blood stages via a fast action mechanism. Development of azithromycin and analogues as antimalarials offers the possibility of targeting parasites through both a quick-killing and delayed-death mechanism of action in a single, multifactorial chemotype.


Subject(s)
Antimalarials/pharmacology , Azithromycin/analogs & derivatives , Azithromycin/pharmacology , Malaria/prevention & control , Plasmodium falciparum/drug effects , Plasmodium knowlesi/drug effects , Plasmodium vivax/drug effects , Malaria, Falciparum/prevention & control , Malaria, Vivax/prevention & control
11.
J Infect Dis ; 220(12): 1950-1961, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31419296

ABSTRACT

BACKGROUND: Complement-fixing antibodies are important mediators of protection against Plasmodium falciparum malaria. However, complement-fixing antibodies remain uncharacterized for Plasmodium vivax malaria. P. vivax merozoite surface protein 3α (PvMSP3α) is a target of acquired immunity and a potential vaccine candidate. METHODS: Plasma from children and adults with P. vivax malaria in Sabah, Malaysia, were collected during acute infection, 7 and 28 days after drug treatment. Complement-fixing antibodies and immunoglobulin M and G (IgM and IgG), targeting 3 distinctive regions of PvMSP3α, were measured by means of enzyme-linked immunosorbent assay. RESULTS: The seroprevalence of complement-fixing antibodies was highest against the PvMSP3α central region (77.6%). IgG1, IgG3, and IgM were significantly correlated with C1q fixation, and both purified IgG and IgM were capable of mediating C1q fixation to PvMSP3α. Complement-fixing antibody levels were similar between age groups, but IgM was predominant in children and IgG3 more prevalent in adults. Levels of functional antibodies increased after acute infection through 7 days after treatment but rapidly waned by day 28. CONCLUSION: Our study demonstrates that PvMSP3α antibodies acquired during P. vivax infection can mediate complement fixation and shows the important influence of age in shaping these specific antibody responses. Further studies are warranted to understand the role of these functional antibodies in protective immunity against P. vivax malaria.


Subject(s)
Antigens, Protozoan/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Aged , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant, Newborn , Kinetics , Malaria, Vivax/drug therapy , Male , Merozoites/immunology , Middle Aged , Plasmodium vivax/classification , Plasmodium vivax/genetics , Seroepidemiologic Studies , Young Adult
12.
Bioorg Med Chem ; 27(2): 436-441, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30581047

ABSTRACT

The 26S proteasome and calpain are linked to a number of important human diseases. Here, we report a series of analogues of the prototypical tripeptide aldehyde inhibitor MG132 that show a unique combination of high activity and selectivity for calpains over proteasome. Tripeptide aldehydes (1-3) with an aromatic P3 substituent show enhanced activity and selectivity against ovine calpain 2 relative to chymotrypsin-like activity of proteasome. Docking studies reveal the key contacts between inhibitors and calpain to confirm the importance of the S3 pocket with respect to selectivity between calpains 1 and 2 and the proteasome.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Proteasome Inhibitors/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/pharmacology , Calpain/chemistry , Catalytic Domain , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Leupeptins/chemical synthesis , Leupeptins/chemistry , Molecular Docking Simulation , Molecular Structure , Plasmodium falciparum/enzymology , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry , Protein Conformation , Rats , Sheep , Swine
13.
Infect Immun ; 84(8): 2175-2184, 2016 08.
Article in English | MEDLINE | ID: mdl-27185785

ABSTRACT

It is unclear whether naturally acquired immunity to Plasmodium falciparum results from the acquisition of antibodies to multiple, diverse antigens or to fewer, highly conserved antigens. Moreover, the specific antibody functions required for malaria immunity are unknown, and hence informative immunological assays are urgently needed to address these knowledge gaps and guide vaccine development. In this study, we investigated whether merozoite-opsonizing antibodies are associated with protection from malaria in a strain-specific or strain-transcending manner by using a novel field isolate and an immune plasma-matched cohort from Papua New Guinea with our validated assay of merozoite phagocytosis. Highly correlated opsonization responses were observed across the 15 parasite strains tested, as were strong associations with protection (composite phagocytosis score across all strains in children uninfected at baseline: hazard ratio of 0.15, 95% confidence interval of 0.04 to 0.63). Opsonizing antibodies had a strong strain-transcending component, and the opsonization of transgenic parasites deficient for MSP3, MSP6, MSPDBL1, or P. falciparum MSP1-19 (PfMSP1-19) was similar to that of wild-type parasites. We have provided the first evidence that merozoite opsonization is predominantly strain transcending, and the highly consistent associations with protection against diverse parasite strains strongly supports the use of merozoite opsonization as a correlate of immunity for field studies and vaccine trials. These results demonstrate that conserved domains within merozoite antigens targeted by opsonization generate strain-transcending immune responses and represent promising vaccine candidates.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Merozoites/immunology , Opsonin Proteins/immunology , Plasmodium falciparum/immunology , Adolescent , Antibodies, Protozoan/blood , Child , Child, Preschool , Humans , Malaria, Falciparum/blood , Patient Outcome Assessment , Phagocytosis/immunology
14.
BMC Med ; 14(1): 144, 2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27658419

ABSTRACT

BACKGROUND: The polymorphic nature of many malaria vaccine candidates presents major challenges to achieving highly efficacious vaccines. Presently, there is very little knowledge on the prevalence and patterns of functional immune responses to polymorphic vaccine candidates in populations to guide vaccine design. A leading polymorphic vaccine candidate against blood-stage Plasmodium falciparum is apical membrane antigen 1 (AMA1), which is essential for erythrocyte invasion. The importance of AMA1 as a target of acquired human inhibitory antibodies, their allele specificity and prevalence in populations is unknown, but crucial for vaccine design. METHODS: P. falciparum lines expressing different AMA1 alleles were genetically engineered and used to quantify functional antibodies from two malaria-exposed populations of adults and children. The acquisition of AMA1 antibodies was also detected using enzyme-linked immunosorbent assay (ELISA) and competition ELISA (using different AMA1 alleles) from the same populations. RESULTS: We found that AMA1 was a major target of naturally acquired invasion-inhibitory antibodies that were highly prevalent in malaria-endemic populations and showed a high degree of allele specificity. Significantly, the prevalence of inhibitory antibodies to different alleles varied substantially within populations and between geographic locations. Inhibitory antibodies to three specific alleles were highly prevalent (FVO and W2mef in Papua New Guinea; FVO and XIE in Kenya), identifying them for potential vaccine inclusion. Measurement of antibodies by standard or competition ELISA was not strongly predictive of allele-specific inhibitory antibodies. The patterns of allele-specific functional antibody responses detected with our novel assays may indicate that acquired immunity is elicited towards serotypes that are prevalent in each geographic location. CONCLUSIONS: These findings provide new insights into the nature and acquisition of functional immunity to a polymorphic vaccine candidate and strategies to quantify functional immunity in populations to guide rational vaccine design.

15.
BMC Biol ; 13: 52, 2015 Jul 18.
Article in English | MEDLINE | ID: mdl-26187647

ABSTRACT

BACKGROUND: Malaria invasion of red blood cells involves multiple parasite-specific targets that are easily accessible to inhibitory compounds, making it an attractive target for antimalarial development. However, no current antimalarial agents act against host cell invasion. RESULTS: Here, we demonstrate that the clinically used macrolide antibiotic azithromycin, which is known to kill human malaria asexual blood-stage parasites by blocking protein synthesis in their apicoplast, is also a rapid inhibitor of red blood cell invasion in human (Plasmodium falciparum) and rodent (P. berghei) malarias. Multiple lines of evidence demonstrate that the action of azithromycin in inhibiting parasite invasion of red blood cells is independent of its inhibition of protein synthesis in the parasite apicoplast, opening up a new strategy to develop a single drug with multiple parasite targets. We identified derivatives of azithromycin and erythromycin that are better invasion inhibitors than parent compounds, offering promise for development of this novel antimalarial strategy. CONCLUSIONS: Safe and effective macrolide antibiotics with dual modalities could be developed to combat malaria and reduce the parasite's options for resistance.


Subject(s)
Antimalarials/pharmacology , Azithromycin/pharmacology , Erythrocytes/parasitology , Erythromycin/pharmacology , Malaria/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Animals , Anopheles , Antimalarials/chemistry , Azithromycin/chemistry , Erythromycin/chemistry , Host-Parasite Interactions/drug effects , Humans , Malaria/parasitology , Mice , Plasmodium berghei/physiology , Plasmodium falciparum/physiology
16.
Cell Microbiol ; 16(5): 642-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24571085

ABSTRACT

Malaria is caused by obligate intracellular parasites, of which Plasmodium falciparum is the most lethal species. In humans, P. falciparum merozoites (invasive forms of the parasite) employ a host of parasite proteins to rapidly invade erythrocytes. One of these is the P. falciparum apical membrane antigen 1 (PfAMA1) which forms a complex with rhoptry neck proteins at the tight junction. Here, we have placed the Pfama1 gene under conditional control using dimerizable Cre recombinase (DiCre) in P. falciparum. DiCre-mediated excision of the loxP-flanked Pfama1 gene results in approximately 80% decreased expression of the protein within one intraerythrocytic growth cycle. This reduces growth by 40%, due to decreased invasion efficiency characterized by a post-invasion defect in sealing of the parasitophorous vacuole. These results show that PfAMA1 is an essential protein for merozoite invasion in P. falciparum and either directly or indirectly plays a role in resealing of the red blood cell at the posterior end of the invasion event.


Subject(s)
Antigens, Protozoan/metabolism , Endocytosis , Erythrocytes/parasitology , Membrane Proteins/metabolism , Merozoites/physiology , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Vacuoles/parasitology , Antigens, Protozoan/genetics , Gene Expression , Membrane Proteins/genetics , Molecular Biology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Recombination, Genetic
17.
J Immunol ; 191(2): 785-94, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23776178

ABSTRACT

Abs that inhibit Plasmodium falciparum invasion of erythrocytes form an important component of human immunity against malaria, but key target Ags are largely unknown. Phenotypic variation by P. falciparum mediates the evasion of inhibitory Abs, contributing to the capacity of P. falciparum to cause repeat and chronic infections. However, Ags involved in mediating immune evasion have not been defined, and studies of the function of human Abs are limited. In this study, we used novel approaches to determine the importance of P. falciparum erythrocyte-binding Ags (EBAs), which are important invasion ligands, as targets of human invasion-inhibitory Abs and define their role in contributing to immune evasion through variation in function. We evaluated the invasion-inhibitory activity of acquired Abs from malaria-exposed children and adults from Kenya, using P. falciparum with disruption of genes encoding EBA140, EBA175, and EBA181, either individually or combined as EBA140/EBA175 or EBA175/EBA181 double knockouts. Our findings provide important new evidence that variation in the expression and function of the EBAs plays an important role in evasion of acquired Abs and that a substantial amount of phenotypic diversity results from variation in expression of different EBAs that contributes to immune evasion by P. falciparum. All three EBAs were identified as important targets of naturally acquired inhibitory Abs demonstrated by differential inhibition of parental parasites greater than EBA knockout lines. This knowledge will help to advance malaria vaccine development and suggests that multiple invasion ligands need to be targeted to overcome the capacity of P. falciparum for immune evasion.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Carrier Proteins/immunology , Immune Evasion , Protozoan Proteins/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Carrier Proteins/genetics , Child , Child, Preschool , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Gene Knockout Techniques , Genetic Variation , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Membrane Proteins , Middle Aged , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Young Adult
18.
Traffic ; 13(11): 1457-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22844982

ABSTRACT

The establishment of parasite infection within the human erythrocyte is an essential stage in the development of malaria disease. As such, significant interest has focused on the mechanics that underpin invasion and on characterization of parasite molecules involved. Previous evidence has implicated a presenilin-like signal peptide peptidase (SPP) from the most virulent human malaria parasite, Plasmodium falciparum, in the process of invasion where it has been proposed to function in the cleavage of the erythrocyte cytoskeletal protein Band 3. The role of a traditionally endoplasmic reticulum (ER) protease in the process of red blood cell invasion is unexpected. Here, using a combination of molecular, cellular and chemical approaches we provide evidence that PfSPP is, instead, a bona fide ER-resident peptidase that remains intracellular throughout the invasion process. Furthermore, SPP-specific drug inhibition has no effect on erythrocyte invasion whilst having low micromolar potency against intra-erythrocytic development. Contrary to previous reports, these results show that PfSPP plays no role in erythrocyte invasion. Nonetheless, PfSPP clearly represents a potential chemotherapeutic target to block parasite growth, supporting ongoing efforts to develop antimalarial-targeting protein maturation and trafficking during intra-erythrocytic development.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Endoplasmic Reticulum/enzymology , Plasmodium falciparum/enzymology , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Erythrocytes/enzymology , Erythrocytes/parasitology , Humans , Merozoites/enzymology , Plasmodium falciparum/growth & development , Plasmodium falciparum/pathogenicity , Protease Inhibitors/pharmacology
19.
Nat Commun ; 15(1): 7206, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174515

ABSTRACT

Apical membrane antigen-1 (AMA1) is a conserved malarial vaccine candidate essential for the formation of tight junctions with the rhoptry neck protein (RON) complex, enabling Plasmodium parasites to invade human erythrocytes, hepatocytes, and mosquito salivary glands. Despite its critical role, extensive surface polymorphisms in AMA1 have led to strain-specific protection, limiting the success of AMA1-based interventions beyond initial clinical trials. Here, we identify an i-body, a humanised single-domain antibody-like molecule that recognises a conserved pan-species conformational epitope in AMA1 with low nanomolar affinity and inhibits the binding of the RON2 ligand to AMA1. Structural characterisation indicates that the WD34 i-body epitope spans the centre of the conserved hydrophobic cleft in AMA1, where interacting residues are highly conserved among all Plasmodium species. Furthermore, we show that WD34 inhibits merozoite invasion of erythrocytes by multiple Plasmodium species and hepatocyte invasion by P. falciparum sporozoites. Despite a short half-life in mouse serum, we demonstrate that WD34 transiently suppressed P. berghei infections in female BALB/c mice. Our work describes the first pan-species AMA1 biologic with inhibitory activity against multiple life-cycle stages of Plasmodium. With improved pharmacokinetic characteristics, WD34 could be a potential immunotherapy against multiple species of Plasmodium.


Subject(s)
Antigens, Protozoan , Erythrocytes , Liver , Membrane Proteins , Mice, Inbred BALB C , Protozoan Proteins , Animals , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Female , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Humans , Erythrocytes/parasitology , Erythrocytes/immunology , Liver/parasitology , Liver/immunology , Liver/metabolism , Malaria Vaccines/immunology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Cross Reactions/immunology , Plasmodium falciparum/immunology , Plasmodium berghei/immunology , Epitopes/immunology , Hepatocytes/parasitology , Hepatocytes/immunology , Hepatocytes/metabolism , Plasmodium/immunology , Merozoites/immunology , Merozoites/metabolism
20.
ChemMedChem ; : e202400549, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39210733

ABSTRACT

The emergence of resistance against current antimalarial treatments has necessitated the need for the development of novel antimalarial chemotypes. Toward this goal, we recently optimised the antimalarial activity of the dihydroquinazolinone scaffold and showed it targeted PfATP4. Here, we deconstruct the lactam moiety of the tricyclic dihydroquinazolinone scaffold and investigate the structure-activity relationship of the truncated scaffold. It was shown that SAR between scaffolds was largely transferrable and generated analogues with potent asexual stage activity. Evaluation of the truncated analogues against PfATP4 mutant drug resistant parasite strains and in assays measuring PfATP4-associated ATPase activity demonstrated retention of PfATP4 as the molecular target. Analogues exhibited activity against both male and female gametes and multidrug resistant parasites. Limited efficacy of analogues in a P. berghei asexual stage mouse model was attributed to their moderate metabolic stability and low aqueous stability. Further development is required to address these attributes toward the potential use of the dihydroquinazolinone class in a curative and transmission blocking combination antimalarial therapy.

SELECTION OF CITATIONS
SEARCH DETAIL