Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Electrophoresis ; 43(3): 509-515, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34679212

RESUMO

Rhoptry neck protein 2 (RON2) binds to the hydrophobic groove of apical membrane antigen 1 (AMA1), an interaction essential for invasion of red blood cells (RBCs) by Plasmodium falciparum (Pf) parasites. Vaccination with AMA1 alone has been shown to be immunogenic, but unprotective even against homologous challenge in human trials. However, the AMA1-RON2L (L is referred to as the loop region of RON2 peptide) complex is a promising candidate, as preclinical studies with Freund's adjuvant have indicated complete protection against lethal challenge in mice and superior protection against virulent infection in Aotus monkeys. To prepare for clinical trials of the AMA1-RON2L complex, identity and integrity of the candidate vaccine must be assessed, and characterization methods must be carefully designed to not dissociate the delicate complex during evaluation. In this study, we developed a native Tris-glycine gel method to separate and identify the AMA1-RON2L complex, which was further identified and confirmed by Western blotting using anti-AMA1 monoclonal antibodies (mAbs 4G2 and 2C2) and anti-RON2L polyclonal Ab coupled with mass spectrometry. The formation of complex was also confirmed by Capillary Isoelectric Focusing (cIEF). A short-term (48 h and 72 h at 4°C) stability study of AMA1-RON2L complex was also performed. The results indicate that the complex was stable for 72 h at 4°C. Our research demonstrates that the native Tris-glycine gel separation/Western blotting coupled with mass spectrometry and cIEF can fully characterize the identity and integrity of the AMA1-RON2L complex and provide useful quality control data for the subsequent clinical trials.


Assuntos
Antígenos de Protozoários , Vacinas Antimaláricas , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Glicina , Focalização Isoelétrica , Vacinas Antimaláricas/química , Proteínas de Membrana/química , Camundongos , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
2.
BMC Genomics ; 20(1): 47, 2019 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651090

RESUMO

BACKGROUND: Bloodstream malaria parasites require Ca++ for their development, but the sites and mechanisms of Ca++ utilization are not well understood. We hypothesized that there may be differences in Ca++ uptake or utilization by genetically distinct lines of P. falciparum. These differences, if identified, may provide insights into molecular mechanisms. RESULTS: Dose response studies with the Ca++ chelator EGTA (ethylene glycol-bis(ß-aminoethyl ether)-N,N,N',N'-tetraacetic acid) revealed stable differences in Ca++ requirement for six geographically divergent parasite lines used in previous genetic crosses, with the largest difference seen between the parents of the HB3 x Dd2 cross. Genetic mapping of Ca++ requirement yielded complex inheritance in 34 progeny clones with a single significant locus on chromosome 7 and possible contributions from other loci. Although encoded by a gene in the significant locus and a proposed Ca++ target, PfCRT (P. falciparum chloroquine resistance transporter), the primary determinant of clinical resistance to the antimalarial drug chloroquine, does not appear to contribute to this quantitative trait. Stage-specific application of extracellular EGTA also excluded determinants associated with merozoite egress and erythrocyte reinvasion. CONCLUSIONS: We have identified differences in Ca++ utilization amongst P. falciparum lines. These differences are under genetic regulation, segregating as a complex trait in genetic cross progeny. Ca++ uptake and utilization throughout the bloodstream asexual cycle of malaria parasites represents an unexplored target for therapeutic intervention.


Assuntos
Cálcio/metabolismo , Loci Gênicos , Malária Falciparum/parasitologia , Parasitos/genética , Plasmodium falciparum/genética , Animais , Cruzamentos Genéticos , Ácido Egtázico/farmacologia , Feminino , Estudos de Associação Genética , Haplótipos/genética , Padrões de Herança/genética , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Merozoítos/efeitos dos fármacos , Merozoítos/metabolismo , Parasitos/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/metabolismo
3.
Nature ; 499(7457): 223-7, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23823717

RESUMO

The variant antigen Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1), which is expressed on the surface of P. falciparum-infected red blood cells, is a critical virulence factor for malaria. Each parasite has 60 antigenically distinct var genes that each code for a different PfEMP1 protein. During infection the clonal parasite population expresses only one gene at a time before switching to the expression of a new variant antigen as an immune-evasion mechanism to avoid the host antibody response. The mechanism by which 59 of the 60 var genes are silenced remains largely unknown. Here we show that knocking out the P. falciparum variant-silencing SET gene (here termed PfSETvs), which encodes an orthologue of Drosophila melanogaster ASH1 and controls histone H3 lysine 36 trimethylation (H3K36me3) on var genes, results in the transcription of virtually all var genes in the single parasite nuclei and their expression as proteins on the surface of individual infected red blood cells. PfSETvs-dependent H3K36me3 is present along the entire gene body, including the transcription start site, to silence var genes. With low occupancy of PfSETvs at both the transcription start site of var genes and the intronic promoter, expression of var genes coincides with transcription of their corresponding antisense long noncoding RNA. These results uncover a previously unknown role of PfSETvs-dependent H3K36me3 in silencing var genes in P. falciparum that might provide a general mechanism by which orthologues of PfSETvs repress gene expression in other eukaryotes. PfSETvs knockout parasites expressing all PfEMP1 proteins may also be applied to the development of a malaria vaccine.


Assuntos
Inativação Gênica , Histonas/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Fatores de Virulência/genética , Proteínas de Ligação a DNA , Proteínas de Drosophila , Eritrócitos/citologia , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Genes de Protozoários/genética , Histonas/química , Íntrons/genética , Lisina/metabolismo , Vacinas Antimaláricas/genética , Metilação , Plasmodium falciparum/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas de Protozoários/genética , RNA Longo não Codificante/genética , Fatores de Transcrição , Sítio de Iniciação de Transcrição , Virulência/genética
4.
Proc Natl Acad Sci U S A ; 111(28): 10311-6, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24958881

RESUMO

An essential step in the invasion of red blood cells (RBCs) by Plasmodium falciparum (Pf) merozoites is the binding of rhoptry neck protein 2 (RON2) to the hydrophobic groove of apical membrane antigen 1 (AMA1), triggering junction formation between the apical end of the merozoite and the RBC surface to initiate invasion. Vaccination with AMA1 provided protection against homologous parasites in one of two phase 2 clinical trials; however, despite its ability to induce high-titer invasion-blocking antibodies in a controlled human challenge trial, the vaccine conferred little protection even against the homologous parasite. Here we provide evidence that immunization with an AMA1-RON2 peptide complex, but not with AMA1 alone, provided complete protection against a lethal Plasmodium yoelii challenge in mice. Significantly, IgG from mice immunized with the complex transferred protection. Furthermore, IgG from PfAMA1-RON2-immunized animals showed enhanced invasion inhibition compared with IgG elicited by AMA1 alone. Interestingly, this qualitative increase in inhibitory activity appears to be related, at least in part, to a switch in the proportion of IgG specific for certain loop regions in AMA1 surrounding the binding site of RON2. Antibodies induced by the complex were not sufficient to block the FVO strain heterologous parasite, however, reinforcing the need to include multiallele AMA1 to cover polymorphisms. Our results suggest that AMA1 subunit vaccines may be highly effective when presented to the immune system as an invasion complex with RON2.


Assuntos
Antígenos de Protozoários/farmacologia , Eritrócitos/imunologia , Imunização , Vacinas Antimaláricas/farmacologia , Malária Falciparum/imunologia , Proteínas de Membrana/farmacologia , Complexos Multiproteicos/farmacologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/farmacologia , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Eritrócitos/parasitologia , Humanos , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/genética , Malária Falciparum/prevenção & controle , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia
5.
Infect Immun ; 83(10): 3771-80, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169272

RESUMO

The extended rod-like Plasmodium falciparum circumsporozoite protein (CSP) is comprised of three primary domains: a charged N terminus that binds heparan sulfate proteoglycans, a central NANP repeat domain, and a C terminus containing a thrombospondin-like type I repeat (TSR) domain. Only the last two domains are incorporated in RTS,S, the leading malaria vaccine in phase 3 trials that, to date, protects about 50% of vaccinated children against clinical disease. A seroepidemiological study indicated that the N-terminal domain might improve the efficacy of a new CSP vaccine. Using a panel of CSP-specific monoclonal antibodies, well-characterized recombinant CSPs, label-free quantitative proteomics, and in vitro inhibition of sporozoite invasion, we show that native CSP is N-terminally processed in the mosquito host and undergoes a reversible conformational change to mask some epitopes in the N- and C-terminal domains until the sporozoite interacts with the liver hepatocyte. Our findings show the importance of understanding processing and the biophysical change in conformation, possibly due to a mechanical or molecular signal, and may aid in the development of a new CSP vaccine.


Assuntos
Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Animais , Anopheles/parasitologia , Anticorpos Antiprotozoários/imunologia , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Hepatócitos/imunologia , Hepatócitos/parasitologia , Humanos , Malária Falciparum/imunologia , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Esporozoítos/química , Esporozoítos/crescimento & desenvolvimento
6.
Proc Natl Acad Sci U S A ; 109(14): 5429-34, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22431641

RESUMO

The malaria parasite, Plasmodium falciparum, and the human immune system have coevolved to ensure that the parasite is not eliminated and reinfection is not resisted. This relationship is likely mediated through a myriad of host-parasite interactions, although surprisingly few such interactions have been identified. Here we show that the 33-kDa fragment of P. falciparum merozoite surface protein 1 (MSP1(33)), an abundant protein that is shed during red blood cell invasion, binds to the proinflammatory protein, S100P. MSP1(33) blocks S100P-induced NFκB activation in monocytes and chemotaxis in neutrophils. Remarkably, S100P binds to both dimorphic alleles of MSP1, estimated to have diverged >27 Mya, suggesting an ancient, conserved relationship between these parasite and host proteins that may serve to attenuate potentially damaging inflammatory responses.


Assuntos
Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteína 1 de Superfície de Merozoito/fisiologia , Proteínas de Neoplasias/antagonistas & inibidores , Plasmodium falciparum/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio/química , Cromatografia em Gel , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Microscopia Confocal , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
7.
Mol Microbiol ; 88(1): 20-34, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23347042

RESUMO

Malaria parasites grow within erythrocytes, but are also free in host plasma between cycles of asexual replication. As a result, the parasite is exposed to fluctuating levels of Na(+) and K(+) , ions assumed to serve important roles for the human pathogen, Plasmodium falciparum. We examined these assumptions and the parasite's ionic requirements by establishing continuous culture in novel sucrose-based media. With sucrose as the primary osmoticant and K(+) and Cl(-) as the main extracellular ions, we obtained parasite growth and propagation at rates indistinguishable from those in physiological media. These conditions abolish long-known increases in intracellular Na(+) via parasite-induced channels, excluding a requirement for erythrocyte cation remodelling. We also dissected Na(+) , K(+) and Cl(-) requirements and found that unexpectedly low concentrations of each ion meet the parasite's demands. Surprisingly, growth was not adversely affected by up to 148 mM K(+) , suggesting that low extracellular K(+) is not an essential trigger for erythrocyte invasion. At the same time, merozoite egress and invasion required a threshold ionic strength, suggesting critical electrostatic interactions between macromolecules at these stages. These findings provide insights into transmembrane signalling in malaria and reveal fundamental differences between host and parasite ionic requirements.


Assuntos
Cátions/farmacologia , Malária/parasitologia , Parasitos/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Animais , Cloretos/farmacologia , Meios de Cultura/farmacologia , Citosol/efeitos dos fármacos , Citosol/metabolismo , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Eritrócitos/ultraestrutura , Interações Hospedeiro-Parasita , Humanos , Merozoítos/efeitos dos fármacos , Merozoítos/crescimento & desenvolvimento , Concentração Osmolar , Parasitos/crescimento & desenvolvimento , Fosfatos/metabolismo , Plasmodium falciparum/crescimento & desenvolvimento , Potássio/farmacologia , Sódio/farmacologia , Sacarose/farmacologia , Trofozoítos/efeitos dos fármacos , Trofozoítos/crescimento & desenvolvimento , Trofozoítos/ultraestrutura
8.
Methods ; 59(3): 301-15, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23270813

RESUMO

Microscale thermophoresis (MST) allows for quantitative analysis of protein interactions in free solution and with low sample consumption. The technique is based on thermophoresis, the directed motion of molecules in temperature gradients. Thermophoresis is highly sensitive to all types of binding-induced changes of molecular properties, be it in size, charge, hydration shell or conformation. In an all-optical approach, an infrared laser is used for local heating, and molecule mobility in the temperature gradient is analyzed via fluorescence. In standard MST one binding partner is fluorescently labeled. However, MST can also be performed label-free by exploiting intrinsic protein UV-fluorescence. Despite the high molecular weight ratio, the interaction of small molecules and peptides with proteins is readily accessible by MST. Furthermore, MST assays are highly adaptable to fit to the diverse requirements of different biomolecules, such as membrane proteins to be stabilized in solution. The type of buffer and additives can be chosen freely. Measuring is even possible in complex bioliquids like cell lysate allowing close to in vivo conditions without sample purification. Binding modes that are quantifiable via MST include dimerization, cooperativity and competition. Thus, its flexibility in assay design qualifies MST for analysis of biomolecular interactions in complex experimental settings, which we herein demonstrate by addressing typically challenging types of binding events from various fields of life science.


Assuntos
Proteínas/química , Espectrometria de Fluorescência/métodos , Animais , Ligação Competitiva , Dimerização , Proteína Adaptadora GRB2/química , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/química , Lasers , Conformação Molecular , Ligação Proteica , Proteínas de Protozoários/química , Ratos , Receptor A2A de Adenosina/química , Receptores de Neurotensina/química , Temperatura , Termodinâmica , Inibidores de beta-Lactamases , beta-Lactamases/química
9.
Proc Natl Acad Sci U S A ; 108(32): 13275-80, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21788485

RESUMO

The commitment of Plasmodium merozoites to invade red blood cells (RBCs) is marked by the formation of a junction between the merozoite and the RBC and the coordinated induction of the parasitophorous vacuole. Despite its importance, the molecular events underlying the parasite's commitment to invasion are not well understood. Here we show that the interaction of two parasite proteins, RON2 and AMA1, known to be critical for invasion, is essential to trigger junction formation. Using antibodies (Abs) that bind near the hydrophobic pocket of AMA1 and AMA1 mutated in the pocket, we identified RON2's binding site on AMA1. Abs specific for the AMA1 pocket blocked junction formation and the induction of the parasitophorous vacuole. We also identified the critical residues in the RON2 peptide (previously shown to bind AMA1) that are required for binding to the AMA1 pocket, namely, two conserved, disulfide-linked cysteines. The RON2 peptide blocked junction formation but, unlike the AMA1-specific Ab, did not block formation of the parasitophorous vacuole, indicating that formation of the junction and parasitophorous vacuole are molecularly distinct steps in the invasion process. Collectively, these results identify the binding of RON2 to the hydrophobic pocket of AMA1 as the step that commits Plasmodium merozoites to RBC invasion and point to RON2 as a potential vaccine candidate.


Assuntos
Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/imunologia , Sítios de Ligação , Sequência Conservada/genética , Cisteína/metabolismo , Citocalasina D/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Frutose-Bifosfato Aldolase/química , Frutose-Bifosfato Aldolase/metabolismo , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Merozoítos/efeitos dos fármacos , Merozoítos/ultraestrutura , Modelos Biológicos , Dados de Sequência Molecular , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/ultraestrutura , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas de Protozoários/química , Relação Estrutura-Atividade
10.
bioRxiv ; 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38352500

RESUMO

Malaria is a global and deadly human disease caused by the apicomplexan parasites of the genus Plasmodium. Parasite proliferation within human red blood cells (RBC) is associated with the clinical manifestations of the disease. This asexual expansion within human RBCs, begins with the invasion of RBCs by P. falciparum, which is mediated by the secretion of effectors from two specialized club-shaped secretory organelles in merozoite-stage parasites known as rhoptries. We investigated the function of the Rhoptry Neck Protein 11 (RON11), which contains seven transmembrane domains and calcium-binding EF-hand domains. We generated conditional mutants of the P. falciparum RON11. Knockdown of RON11 inhibits parasite growth by preventing merozoite invasion. The loss of RON11 did not lead to any defects in processing of rhoptry proteins but instead led to a decrease in the amount of rhoptry proteins. We utilized ultrastructure expansion microscopy (U-ExM) to determine the effect of RON11 knockdown on rhoptry biogenesis. Surprisingly, in the absence of RON11, fully developed merozoites had only one rhoptry each. The single rhoptry in RON11 deficient merozoites were morphologically typical with a bulb and a neck oriented into the apical polar ring. Moreover, rhoptry proteins are trafficked accurately to the single rhoptry in RON11 deficient parasites. These data show that in the absence of RON11, the first rhoptry is generated during schizogony but upon the start of cytokinesis, the second rhoptry never forms. Interestingly, these single-rhoptry merozoites were able to attach to host RBCs but are unable to invade RBCs. Instead, RON11 deficient merozoites continue to engage with RBC for prolonged periods eventually resulting in echinocytosis, a result of secreting the contents from the single rhoptry into the RBC. Together, our data show that RON11 triggers the de novo biogenesis of the second rhoptry and functions in RBC invasion.

11.
bioRxiv ; 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38895284

RESUMO

Rodent malaria models serve as important preclinical antimalarial and vaccine testing tools. Evaluating treatment outcomes in these models often requires manually counting parasite-infected red blood cells (iRBCs), a time-consuming process, which can be inconsistent between individuals and labs. We have developed an easy-to-use machine learning (ML)-based software, Malaria Screener R, to expedite and standardize such studies by automating the counting of Plasmodium iRBCs in rodents. This software can process Giemsa-stained blood smear images captured by any camera-equipped microscope. It features an intuitive graphical user interface that facilitates image processing and visualization of the results. The software has been developed as a desktop application that processes images on standard Windows and Mac OS computers. A previous ML model created by the authors designed to count P. falciparum -infected human RBCs did not perform well counting Plasmodium -infected mouse RBCs. We leveraged that model by loading the pre-trained weights and training the algorithm with newly collected data to target P. yoelii and P. berghei mouse iRBCs. This new model reliably measured both P. yoelii and P. berghei parasitemia (R 2 = 0.9916). Additional rounds of training data to incorporate variances due to length of Giemsa staining, microscopes etc, have produced a generalizable model, meeting WHO Competency Level 1 for the sub-category of parasite counting using independent microscopes. Reliable, automated analyses of blood-stage parasitemia will facilitate rapid and consistent evaluation of novel vaccines and antimalarials across labs in an easily accessible in vivo malaria model.

12.
PLoS Pathog ; 7(3): e1001318, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21445239

RESUMO

Malaria-protective CD8+ T cells specific for the circumsporozoite (CS) protein are primed by dendritic cells (DCs) after sporozoite injection by infected mosquitoes. The primed cells then eliminate parasite liver stages after recognizing the CS epitopes presented by hepatocytes. To define the in vivo processing of CS by DCs and hepatocytes, we generated parasites carrying a mutant CS protein containing the H-2K(b) epitope SIINFEKL, and evaluated the T cell response using transgenic and mutant mice. We determined that in both DCs and hepatocytes CS epitopes must reach the cytosol and use the TAP transporters to access the ER. Furthermore, we used endosomal mutant (3d) and cytochrome c treated mice to address the role of cross-presentation in the priming and effector phases of the T cell response. We determined that in DCs, CS is cross-presented via endosomes while, conversely, in hepatocytes protein must be secreted directly into the cytosol. This suggests that the main targets of protective CD8+ T cells are parasite proteins exported to the hepatocyte cytosol. Surprisingly, however, secretion of the CS protein into hepatocytes was not dependent upon parasite-export (Pexel/VTS) motifs in this protein. Together, these results indicate that the presentation of epitopes to CD8+ T cells follows distinct pathways in DCs when the immune response is induced and in hepatocytes during the effector phase.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Protozoários/imunologia , Células Dendríticas/imunologia , Hepatócitos/imunologia , Malária/imunologia , Plasmodium berghei/imunologia , Proteínas de Protozoários/imunologia , Animais , Apresentação de Antígeno/genética , Antígenos de Protozoários/genética , Linfócitos T CD8-Positivos/imunologia , Epitopos/genética , Epitopos/imunologia , Feminino , Malária/genética , Camundongos , Camundongos Transgênicos , Plasmodium berghei/genética , Proteínas de Protozoários/genética
13.
Arterioscler Thromb Vasc Biol ; 32(3): 786-98, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22116094

RESUMO

OBJECTIVE: The coagulation-inflammation cycle has been implicated as a critical component in malaria pathogenesis. Defibrotide (DF), a mixture of DNA aptamers, displays anticoagulant, anti-inflammatory, and endothelial cell (EC)-protective activities and has been successfully used to treat comatose children with veno-occlusive disease. DF was investigated here as a drug to treat cerebral malaria. METHODS AND RESULTS: DF blocks tissue factor expression by ECs incubated with parasitized red blood cells and attenuates prothrombinase activity, platelet aggregation, and complement activation. In contrast, it does not affect nitric oxide bioavailability. We also demonstrated that Plasmodium falciparum glycosylphosphatidylinositol (Pf-GPI) induces tissue factor expression in ECs and cytokine production by dendritic cells. Notably, dendritic cells, known to modulate coagulation and inflammation systemically, were identified as a novel target for DF. Accordingly, DF inhibits Toll-like receptor ligand-dependent dendritic cells activation by a mechanism that is blocked by adenosine receptor antagonist (8-p-sulfophenyltheophylline) but not reproduced by synthetic poly-A, -C, -T, and -G. These results imply that aptameric sequences and adenosine receptor mediate dendritic cells responses to the drug. DF also prevents rosetting formation, red blood cells invasion by P. falciparum and abolishes oocysts development in Anopheles gambiae. In a murine model of cerebral malaria, DF affected parasitemia, decreased IFN-γ levels, and ameliorated clinical score (day 5) with a trend for increased survival. CONCLUSION: Therapeutic use of DF in malaria is proposed.


Assuntos
Anti-Inflamatórios/farmacologia , Anticoagulantes/farmacologia , Antimaláricos/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Malária Cerebral/tratamento farmacológico , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Polidesoxirribonucleotídeos/farmacologia , Animais , Células Cultivadas , Ativação do Complemento/efeitos dos fármacos , Citocinas/sangue , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/parasitologia , Feminino , Glicosilfosfatidilinositóis/metabolismo , Hemoglobinas/metabolismo , Humanos , Mediadores da Inflamação/sangue , Malária Cerebral/sangue , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Plasmodium berghei/patogenicidade , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Agregação Plaquetária/efeitos dos fármacos , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores Purinérgicos P1/metabolismo , Índice de Gravidade de Doença , Tromboplastina/metabolismo , Fatores de Tempo
14.
Nat Commun ; 14(1): 5879, 2023 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-37735574

RESUMO

Invasion of human erythrocytes by Plasmodium falciparum (Pf) merozoites relies on the interaction between two parasite proteins: apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2). While antibodies to AMA1 provide limited protection against Pf in non-human primate malaria models, clinical trials using recombinant AMA1 alone (apoAMA1) yielded no protection due to insufficient functional antibodies. Immunization with AMA1 bound to RON2L, a 49-amino acid peptide from its ligand RON2, has shown superior protection by increasing the proportion of neutralizing antibodies. However, this approach relies on the formation of a complex in solution between the two vaccine components. To advance vaccine development, here we engineered chimeric antigens by replacing the AMA1 DII loop, displaced upon ligand binding, with RON2L. Structural analysis confirmed that the fusion chimera (Fusion-FD12) closely mimics the binary AMA1-RON2L complex. Immunization studies in female rats demonstrated that Fusion-FD12 immune sera, but not purified IgG, neutralized vaccine-type parasites more efficiently compared to apoAMA1, despite lower overall anti-AMA1 titers. Interestingly, Fusion-FD12 immunization enhanced antibodies targeting conserved epitopes on AMA1, leading to increased neutralization of non-vaccine type parasites. Identifying these cross-neutralizing antibody epitopes holds promise for developing an effective, strain-transcending malaria vaccine.


Assuntos
Anticorpos Neutralizantes , Feminino , Animais , Ratos , Anticorpos Amplamente Neutralizantes , Ligantes , Membrana Celular , Epitopos
15.
Res Sq ; 2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37131813

RESUMO

Invasion of human red blood cells (RBCs) by Plasmodium falciparum (Pf) merozoites relies on the interaction between two parasite proteins, apical membrane antigen 1 (AMA1) and rhoptry neck protein 2 (RON2) 1,2 . Antibodies to AMA1 confer limited protection against P. falciparum in non-human primate malaria models 3,4 . However, clinical trials with recombinant AMA1 alone (apoAMA1) saw no protection, likely due to inadequate levels of functional antibodies 5-8 . Notably, immunization with AMA1 in its ligand bound conformation using RON2L, a 49 amino acid peptide from RON2, confers superior protection against P. falciparum malaria by enhancing the proportion of neutralizing antibodies 9,10 . A limitation of this approach, however, is that it requires the two vaccine components to form a complex in solution. To facilitate vaccine development, we engineered chimeric antigens by strategically replacing the AMA1 DII loop that is displaced upon ligand binding with RON2L. Structural characterization of the fusion chimera, Fusion-F D12 to 1.55 Å resolution showed that it closely mimics the binary receptor-ligand complex. Immunization studies showed that Fusion-F D12 immune sera neutralized parasites more efficiently than apoAMA1 immune sera despite having an overall lower anti-AMA1 titer, suggesting improvement in antibody quality. Furthermore, immunization with Fusion-F D12 enhanced antibodies targeting conserved epitopes on AMA1 resulting in greater neutralization of non-vaccine type parasites. Identifying epitopes of such cross-neutralizing antibodies will help in the development of an effective, strain-transcending malaria vaccine. Our fusion protein design is a robust vaccine platform that can be enhanced by incorporating polymorphisms in AMA1 to effectively neutralize all P. falciparum parasites.

16.
Artigo em Inglês | MEDLINE | ID: mdl-22871010

RESUMO

A simple, yet robust analytical method was developed to detect and quantify three sulfonamides (SA), namely sulfamethoxazole (SMO), sulfachloropyridazine (SCP), and sulfamethazine (SM), and a macrolide tylosin (TT) in aqueous (calcium chloride and leachate solutions) and solid (agricultural soils) matrices using high performance liquid chromatography and ultra violet detection at 290 nm (TT) and 275 nm (SA) respectively. Chromatography was performed using a Phenomenex Onyx Monolithic C(18) column for TT and a C(18) Luna column for sulfonamides as single analytes eluted isocratically with a mobile phase consisting of acetonitrile: trifluoroacetic acid: tetrahydrofuran in the ratio 22.5:68:9.5 for TT, 40:55:5 for SMO, 32:63:5 for SCP and 31:64:5 for SM (v/v) at 1.0 mL min(-1) and an injection volume of 20 µL. A gradient method to detect all three sulfonamides in a single run was also developed. The soil residue analysis consisted of extraction with dichloromethane and pre-concentration steps as the aqueous phase was measured directly. The limits of detection at an S/N (signal: noise) ratio of 3 were 20.0 µg L(-1) and 50 µg L(-1) for all sulfonamides and tylosin respectively. The average recoveries for all sulfonamides and tylosin in aqueous matrices ranged from 95 to 105% across the six concentrations investigated. Recoveries from the soils were slightly lower for sulfonamides and tylosin. The isocratic method was used to determine the sorption and degradation of sulfonamides in soils, while the gradient method was used to determine degradation kinetics and leachate concentrations in soils and aqueous systems.


Assuntos
Antibacterianos/análise , Cromatografia Líquida de Alta Pressão/métodos , Poluentes do Solo/análise , Poluentes da Água/análise , Sulfametazina/análise , Sulfametoxazol/análise , Sulfonamidas/análise , Tilosina/análise
17.
PLoS Pathog ; 5(1): e1000262, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19148267

RESUMO

Invasion of host cells by the malaria parasite involves recognition and interaction with cell-surface receptors. A wide variety of parasite surface proteins participate in this process, most of which are specific to the parasite's particular invasive form. Upon entry, the parasite has to dissociate itself from the host-cell receptors. One mechanism by which it does so is by shedding its surface ligands using specific enzymes. Rhomboid belongs to a family of serine proteases that cleave cell-surface proteins within their transmembrane domains. Here we identify and partially characterize a Plasmodium berghei rhomboid protease (PbROM1) that plays distinct roles during parasite development. PbROM1 localizes to the surface of sporozoites after salivary gland invasion. In blood stage merozoites, PbROM1 localizes to the apical end where proteins involved in invasion are also present. Our genetic analysis suggests that PbROM1 functions in the invasive stages of parasite development. Whereas wild-type P. berghei is lethal to mice, animals infected with PbROM1 null mutants clear the parasites efficiently and develop long-lasting protective immunity. The results indicate that P. berghei Rhomboid 1 plays a nonessential but important role during parasite development and identify rhomboid proteases as potential targets for disease control.


Assuntos
Malária/parasitologia , Plasmodium berghei/enzimologia , Plasmodium berghei/patogenicidade , Serina Endopeptidases/fisiologia , Animais , Anopheles/parasitologia , Feminino , Interações Hospedeiro-Parasita , Camundongos , Proteínas de Protozoários/fisiologia , Serina Endopeptidases/genética , Esporozoítos/metabolismo
18.
PLoS Pathog ; 5(2): e1000302, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19229315

RESUMO

Infection of red blood cells (RBC) subjects the malaria parasite to oxidative stress. Therefore, efficient antioxidant and redox systems are required to prevent damage by reactive oxygen species. Plasmodium spp. have thioredoxin and glutathione (GSH) systems that are thought to play a major role as antioxidants during blood stage infection. In this report, we analyzed a critical component of the GSH biosynthesis pathway using reverse genetics. Plasmodium berghei parasites lacking expression of gamma-glutamylcysteine synthetase (gamma-GCS), the rate limiting enzyme in de novo synthesis of GSH, were generated through targeted gene disruption thus demonstrating, quite unexpectedly, that gamma-GCS is not essential for blood stage development. Despite a significant reduction in GSH levels, blood stage forms of pbggcs(-) parasites showed only a defect in growth as compared to wild type. In contrast, a dramatic effect on development of the parasites in the mosquito was observed. Infection of mosquitoes with pbggcs(-) parasites resulted in reduced numbers of stunted oocysts that did not produce sporozoites. These results have important implications for the design of drugs aiming at interfering with the GSH redox-system in blood stages and demonstrate that de novo synthesis of GSH is pivotal for development of Plasmodium in the mosquito.


Assuntos
Glutamato-Cisteína Ligase/genética , Glutationa/metabolismo , Malária/transmissão , Plasmodium berghei/genética , Análise de Variância , Animais , Anopheles/parasitologia , Proliferação de Células , Eritrócitos/parasitologia , Feminino , Expressão Gênica , Marcação de Genes , Glutamato-Cisteína Ligase/metabolismo , Malária/parasitologia , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Mitocôndrias , Oocistos/citologia , Oocistos/crescimento & desenvolvimento , Oocistos/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/metabolismo , Esporozoítos/metabolismo , Estatísticas não Paramétricas
19.
Elife ; 102021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33393463

RESUMO

Malaria parasites use the RhopH complex for erythrocyte invasion and channel-mediated nutrient uptake. As the member proteins are unique to Plasmodium spp., how they interact and traffic through subcellular sites to serve these essential functions is unknown. We show that RhopH is synthesized as a soluble complex of CLAG3, RhopH2, and RhopH3 with 1:1:1 stoichiometry. After transfer to a new host cell, the complex crosses a vacuolar membrane surrounding the intracellular parasite and becomes integral to the erythrocyte membrane through a PTEX translocon-dependent process. We present a 2.9 Å single-particle cryo-electron microscopy structure of the trafficking complex, revealing that CLAG3 interacts with the other subunits over large surface areas. This soluble complex is tightly assembled with extensive disulfide bonding and predicted transmembrane helices shielded. We propose a large protein complex stabilized for trafficking but poised for host membrane insertion through large-scale rearrangements, paralleling smaller two-state pore-forming proteins in other organisms.


Malaria is an infectious disease caused by the family of Plasmodium parasites, which pass between mosquitoes and animals to complete their life cycle. With one bite, mosquitoes can deposit up to one hundred malaria parasites into the human skin, from where they enter the bloodstream. After increasing their numbers in liver cells, the parasites hijack, invade and remodel red blood cells to create a safe space to grow and mature. This includes inserting holes in the membrane of red blood cells to take up nutrients from the bloodstream. A complex of three tightly bound RhopH proteins plays an important role in these processes. These proteins are unique to malaria parasites, and so far, it has been unclear how they collaborate to perform these specialist roles. Here, Schureck et al. have purified the RhopH complex from Plasmodium-infected human blood to determine its structure and reveal how it moves within an infected red blood cell. Using cryo-electron microscopy to visualise the assembly in fine detail, Schureck et al. showed that the three proteins bind tightly to each other over large areas using multiple anchor points. As the three proteins are produced, they assemble into a complex that remains dissolved and free of parasite membranes until the proteins have been delivered to their target red blood cells. Some hours after delivery, specific sections of the RhopH complex are inserted into the red blood cell membrane to produce pores that allow them to take up nutrients and to grow. The study of Schureck et al. provides important new insights into how the RhopH complex serves multiple roles during Plasmodium infection of human red blood cells. The findings provide a framework for the development of effective antimalarial treatments that target RhopH proteins to block red blood cell invasion and nutrient uptake.


Assuntos
Eritrócitos/parasitologia , Genes de Protozoários/fisiologia , Plasmodium falciparum/fisiologia , Família Multigênica/fisiologia , Nutrientes/metabolismo , Plasmodium falciparum/genética
20.
Cell Microbiol ; 10(6): 1304-12, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18248630

RESUMO

An essential requisite for transmission of Plasmodium, the causative agent of malaria, is the successful completion of a complex developmental cycle in its mosquito vector. Of hundreds of ookinetes that form in the mosquito midgut, only few transform into oocysts, a loss attributed to the action of the mosquito immune system. However, once oocysts form, they appear to be resistant to mosquito defences. During oocyst development, a thick capsule forms around the parasite and appears to function as a protective cover. Little information is available about the composition of this capsule. Here we report on the identification and partial characterization of the first Plasmodium oocyst capsule protein (PbCap380). Genetic analysis indicates that the gene is essential and that PbCap380(-) mutant parasites form oocysts in normal numbers but are gradually eliminated. As a result, mosquitoes infected with PbCap380(-) parasites do not transmit malaria. Targeting of the oocyst capsule may provide a new strategy for malaria control.


Assuntos
Culicidae/parasitologia , Insetos Vetores/parasitologia , Malária/parasitologia , Oocistos/crescimento & desenvolvimento , Plasmodium/fisiologia , Proteínas de Protozoários/fisiologia , Animais , Genes de Protozoários/genética , Interações Hospedeiro-Parasita , Imuno-Histoquímica , Estágios do Ciclo de Vida , Microscopia Imunoeletrônica , Oocistos/metabolismo , Plasmodium/patogenicidade , Mutação Puntual , Reação em Cadeia da Polimerase , Proteínas de Protozoários/isolamento & purificação , Esporozoítos/crescimento & desenvolvimento , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA