Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Malar J ; 15: 28, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26772184

RESUMO

BACKGROUND: Rosetting, namely the capacity of the Plasmodium falciparum-infected red blood cells to bind uninfected RBCs, is commonly observed in African children with severe malaria. Rosetting results from specific interactions between a subset of variant P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesins encoded by var genes, serum components and RBC receptors. Rosette formation is a redundant phenotype, as there exists more than one var gene encoding a rosette-mediating PfEMP1 in each genome and hence a diverse array of underlying interactions. Moreover, field diversity creates a large panel of rosetting-associated serotypes and studies with human immune sera indicate that surface-reacting antibodies are essentially variant-specific. To gain better insight into the interactions involved in rosetting and map surface epitopes, a panel of monoclonal antibodies (mAbs) was investigated. METHODS: Monoclonal antibodies were isolated from mice immunized with PfEMP1-VarO recombinant domains. They were characterized using ELISA and reactivity with the native PfEMP1-VarO adhesin on immunoblots of reduced and unreduced extracts, as well as SDS-extracts of Palo Alto 89F5 VarO schizonts. Functionality was assessed using inhibition of Palo Alto 89F5 VarO rosette formation and disruption of Palo Alto 89F5 VarO rosettes. Competition ELISAs were performed with biotinylated antibodies against DBL1 to identify reactivity groups. Specificity of mAbs reacting with the DBL1 adhesion domain was explored using recombinant proteins carrying mutations abolishing RBC binding or binding to heparin, a potent inhibitor of rosette formation. RESULTS: Domain-specific, surface-reacting mAbs were obtained for four individual domains (DBL1, CIDR1, DBL2, DBL4). Monoclonal antibodies reacting with DBL1 potently inhibited the formation of rosettes and disrupted Palo Alto 89F5 VarO rosettes. Most surface-reactive mAbs and all mAbs interfering with rosetting reacted on parasite immunoblots with disulfide bond-dependent PfEMP1 epitopes. Based on competition ELISA and binding to mutant DBL1 domains, two distinct binding sites for rosette-disrupting mAbs were identified in close proximity to the RBC-binding site. CONCLUSIONS: Rosette-inhibitory antibodies bind to conformation-dependent epitopes located close to the RBC-binding site and distant from the heparin-binding site. These results provide novel clues for a rational intervention strategy that targets rosetting.


Assuntos
Anticorpos Monoclonais/metabolismo , Moléculas de Adesão Celular/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Ensaio de Imunoadsorção Enzimática , Camundongos , Plasmodium falciparum/efeitos dos fármacos , Ligação Proteica
2.
PLoS Pathog ; 8(6): e1002755, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22737069

RESUMO

Members of the phylum Apicomplexa, which include the malaria parasite Plasmodium, share many features in their invasion mechanism in spite of their diverse host cell specificities and life cycle characteristics. The formation of a moving junction (MJ) between the membranes of the invading apicomplexan parasite and the host cell is common to these intracellular pathogens. The MJ contains two key parasite components: the surface protein Apical Membrane Antigen 1 (AMA1) and its receptor, the Rhoptry Neck Protein (RON) complex, which is targeted to the host cell membrane during invasion. In particular, RON2, a transmembrane component of the RON complex, interacts directly with AMA1. Here, we report the crystal structure of AMA1 from Plasmodium falciparum in complex with a peptide derived from the extracellular region of PfRON2, highlighting clear specificities of the P. falciparum RON2-AMA1 interaction. The receptor-binding site of PfAMA1 comprises the hydrophobic groove and a region that becomes exposed by displacement of the flexible Domain II loop. Mutations of key contact residues of PfRON2 and PfAMA1 abrogate binding between the recombinant proteins. Although PfRON2 contacts some polymorphic residues, binding studies with PfAMA1 from different strains show that these have little effect on affinity. Moreover, we demonstrate that the PfRON2 peptide inhibits erythrocyte invasion by P. falciparum merozoites and that this strong inhibitory potency is not affected by AMA1 polymorphisms. In parallel, we have determined the crystal structure of PfAMA1 in complex with the invasion-inhibitory peptide R1 derived by phage display, revealing an unexpected structural mimicry of the PfRON2 peptide. These results identify the key residues governing the interactions between AMA1 and RON2 in P. falciparum and suggest novel approaches to antimalarial therapeutics.


Assuntos
Antígenos de Protozoários/química , Interações Hospedeiro-Parasita/fisiologia , Proteínas de Membrana/química , Plasmodium falciparum/química , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/metabolismo , Membrana Celular/metabolismo , Cristalização , Proteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium falciparum/metabolismo , Polimorfismo Genético , Ligação Proteica , Estrutura Quaternária de Proteína , Proteínas de Protozoários/metabolismo , Ressonância de Plasmônio de Superfície
3.
PLoS Pathog ; 8(7): e1002781, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22807674

RESUMO

The ABO blood group influences susceptibility to severe Plasmodium falciparum malaria. Recent evidence indicates that the protective effect of group O operates by virtue of reduced rosetting of infected red blood cells (iRBCs) with uninfected RBCs. Rosetting is mediated by a subgroup of PfEMP1 adhesins, with RBC binding being assigned to the N-terminal DBL1α1 domain. Here, we identify the ABO blood group as the main receptor for VarO rosetting, with a marked preference for group A over group B, which in turn is preferred to group O RBCs. We show that recombinant NTS-DBL1α1 and NTS-DBL1α1-CIDR1γ reproduce the VarO-iRBC blood group preference and document direct binding to blood group trisaccharides by surface plasmon resonance. More detailed RBC subgroup analysis showed preferred binding to group A1, weaker binding to groups A2 and B, and least binding to groups A(x) and O. The 2.8 Å resolution crystal structure of the PfEMP1-VarO Head region, NTS-DBL1α1-CIDR1γ, reveals extensive contacts between the DBL1α1 and CIDR1γ and shows that the NTS-DBL1α1 hinge region is essential for RBC binding. Computer docking of the blood group trisaccharides and subsequent site-directed mutagenesis localized the RBC-binding site to the face opposite to the heparin-binding site of NTS-DBLα1. RBC binding involves residues that are conserved between rosette-forming PfEMP1 adhesins, opening novel opportunities for intervention against severe malaria. By deciphering the structural basis of blood group preferences in rosetting, we provide a link between ABO blood grouppolymorphisms and rosette-forming adhesins, consistent with the selective role of falciparum malaria on human genetic makeup.


Assuntos
Sistema ABO de Grupos Sanguíneos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Formação de Roseta , Sistema ABO de Grupos Sanguíneos/imunologia , Sequência de Aminoácidos , Anticorpos Antiprotozoários/imunologia , Sítios de Ligação , Cristalografia por Raios X , Eritrócitos/imunologia , Eritrócitos/metabolismo , Humanos , Reação de Imunoaderência , Malária Falciparum/sangue , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Plasmodium falciparum/genética , Plasmodium falciparum/ultraestrutura , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia
4.
Int J Neuropsychopharmacol ; 17(5): 705-13, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24405657

RESUMO

The brain-derived neurotropic factor (BDNF) Val66Met polymorphism has been associated with abnormalities of synaptic plasticity in animal models, and abnormalities in motor cortical plasticity have also been described in humans using transcranial direct current stimulation. No study has yet been done on plasticity in non-motor regions, and the effect of two Met alleles (i.e. 'Met dose') is not well understood. We studied the effect of the BDNF Val66Met polymorphism on the after-effects of transcranial direct current stimulation and tetanic auditory stimulation in 65 subjects (23; Val66Val, 22; Val66Met and 20; Met66Met genotypes). In the first session, motor evoked potentials (MEP) were recorded under stereotaxic guidance for 90 min after 9 min of anodal transcranial direct current stimulation (TDCS). In the second session, auditory-evoked potentials (AEP) were recorded before and after 2 min of auditory 13 Hz tetanic stimulation. There was a difference in MEP facilitation post-TDCS comparing Met carriers with non-Met carriers, with Met carriers having a modest late facilitation at 30-90 min. There was no difference in responses between Val66Met genotype and Met66Met genotype subjects. Tetanic auditory stimulation also produced late facilitation of N1-P2 AEP at 25 min, but there was no apparent effect of genetic status. This study indicates that Met66Met carriers behave like Val66Met carriers for TDCS-induced plasticity, and produce a late facilitation of MEPs. Auditory cortical plasticity was not affected by the BDNF Val66Met polymorphism. This study sheds light on the differences between auditory and motor cortical plasticity and the role of the BDNF Val66Met polymorphism.


Assuntos
Córtex Auditivo/fisiologia , Percepção Auditiva , Fator Neurotrófico Derivado do Encéfalo/genética , Córtex Motor/fisiologia , Plasticidade Neuronal , Polimorfismo de Nucleotídeo Único , Estimulação Acústica , Adulto , Alelos , Percepção Auditiva/genética , Estimulação Elétrica , Potenciais Evocados Auditivos/genética , Potencial Evocado Motor/genética , Feminino , Técnicas de Genotipagem , Humanos , Masculino , Pessoa de Meia-Idade , Plasticidade Neuronal/genética , Estimulação Magnética Transcraniana , Adulto Jovem
5.
Proc Natl Acad Sci U S A ; 108(13): 5243-8, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21402930

RESUMO

The human malaria parasite Plasmodium falciparum can cause infected red blood cells (iRBC) to form rosettes with uninfected RBC, a phenotype associated with severe malaria. Rosetting is mediated by a subset of the Plasmodium falciparum membrane protein 1 (PfEMP1) variant adhesins expressed on the infected host-cell surface. Heparin and other sulfated oligosaccharides, however, can disrupt rosettes, suggesting that therapeutic approaches to this form of severe malaria are feasible. We present a structural and functional study of the N-terminal domain of PfEMP1 from the VarO variant comprising the N-terminal segment (NTS) and the first DBL domain (DBL1α(1)), which is directly implicated in rosetting. We demonstrate that NTS-DBL1α(1)-VarO binds to RBC and that heparin inhibits this interaction in a dose-dependent manner, thus mimicking heparin-mediated rosette disruption. We have determined the crystal structure of NTS-DBL1α(1), showing that NTS, previously thought to be a structurally independent component of PfEMP1, forms an integral part of the DBL1α domain. Using mutagenesis and docking studies, we have located the heparin-binding site, which includes NTS. NTS, unique to the DBL α-class domain, is thus an intrinsic structural and functional component of the N-terminal VarO domain. The specific interaction observed with heparin opens the way for developing antirosetting therapeutic strategies.


Assuntos
Eritrócitos/parasitologia , Heparina/metabolismo , Plasmodium falciparum/metabolismo , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Formação de Roseta , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
6.
PLoS Pathog ; 7(2): e1001276, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21347343

RESUMO

Obligate intracellular Apicomplexa parasites share a unique invasion mechanism involving a tight interaction between the host cell and the parasite surfaces called the moving junction (MJ). The MJ, which is the anchoring structure for the invasion process, is formed by secretion of a macromolecular complex (RON2/4/5/8), derived from secretory organelles called rhoptries, into the host cell membrane. AMA1, a protein secreted from micronemes and associated with the parasite surface during invasion, has been shown in vitro to bind the MJ complex through a direct association with RON2. Here we show that RON2 is inserted as an integral membrane protein in the host cell and, using several interaction assays with native or recombinant proteins, we define the region that binds AMA1. Our studies were performed both in Toxoplasma gondii and Plasmodium falciparum and although AMA1 and RON2 proteins have diverged between Apicomplexa species, we show an intra-species conservation of their interaction. More importantly, invasion inhibition assays using recombinant proteins demonstrate that the RON2-AMA1 interaction is crucial for both T. gondii and P. falciparum entry into their host cells. This work provides the first evidence that AMA1 uses the rhoptry neck protein RON2 as a receptor to promote invasion by Apicomplexa parasites.


Assuntos
Antígenos de Protozoários/metabolismo , Apicomplexa/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Proteínas de Protozoários/metabolismo , Internalização do Vírus , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Apicomplexa/genética , Apicomplexa/metabolismo , Células Cultivadas , Chlorocebus aethiops , Conexinas/metabolismo , Sequência Conservada , Interações Hospedeiro-Parasita/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Modelos Biológicos , Modelos Moleculares , Parasitos/genética , Parasitos/metabolismo , Parasitos/fisiologia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Ligação Proteica/genética , Domínios e Motivos de Interação entre Proteínas/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasma/fisiologia , Células Vero
7.
Int J Neuropsychopharmacol ; 16(4): 721-31, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22932339

RESUMO

Episodic memory deficits are a core feature of neurodegenerative disorders. Muscarinic M(1) receptors play a critical role in modulating learning and memory and are highly expressed in the hippocampus. We examined the effect of GSK1034702, a potent M(1) receptor allosteric agonist, on cognitive function, and in particular episodic memory, in healthy smokers using the nicotine abstinence model of cognitive dysfunction. The study utilized a randomized, double-blind, placebo-controlled, cross-over design in which 20 male nicotine abstained smokers were tested following single doses of placebo, 4 and 8 mg GSK1034702. Compared to the baseline (nicotine on-state), nicotine abstinence showed statistical significance in reducing immediate (p=0.019) and delayed (p=0.02) recall. GSK1034702 (8 mg) significantly attenuated (i.e. improved) immediate recall (p=0.014) but not delayed recall. None of the other cognitive domains was modulated by either nicotine abstinence or GSK1034702. These findings suggest that stimulating M(1) receptor mediated neurotransmission in humans with GSK1034702 improves memory encoding potentially by modulating hippocampal function. Hence, selective M(1) receptor allosteric agonists may have therapeutic benefits in disorders of impaired learning including Alzheimer's disease.


Assuntos
Comportamento Aditivo/psicologia , Benzimidazóis/uso terapêutico , Transtornos Cognitivos/psicologia , Memória Episódica , Receptor Muscarínico M1/agonistas , Abandono do Hábito de Fumar/psicologia , Adulto , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Comportamento Aditivo/tratamento farmacológico , Benzimidazóis/farmacologia , Transtornos Cognitivos/tratamento farmacológico , Estudos Cross-Over , Método Duplo-Cego , Humanos , Masculino , Pessoa de Meia-Idade , Nicotina , Receptor Muscarínico M1/fisiologia , Fumar/psicologia , Adulto Jovem
8.
Proc Natl Acad Sci U S A ; 107(11): 4884-9, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20194779

RESUMO

Pregnancy-associated malaria (PAM) is a serious consequence of sequestration of Plasmodium falciparum-parasitized erythrocytes (PE) in the placenta through adhesion to chondroitin sulfate A (CSA) present on placental proteoglycans. Recent work implicates var2CSA, a member of the PfEMP1 family, as the mediator of placental sequestration and as a key target for PAM vaccine development. Var2CSA is a 350 kDa transmembrane protein, whose extracellular region includes six Duffy-binding-like (DBL) domains. Due to its size and high cysteine content, the full-length var2CSA extracellular region has not hitherto been expressed in heterologous systems, thus limiting investigations to individual recombinant domains. Here we report for the first time the expression of the full-length var2CSA extracellular region (domains DBL1X to DBL6epsilon) from the 3D7 parasite strain using the human embryonic kidney 293 cell line. We show that the recombinant extracellular var2CSA region is correctly folded and that, unlike the individual DBL domains, it binds with high affinity and specificity to CSA (K(D) = 61 nM) and efficiently inhibits PE from binding to CSA. Structural characterization by analytical ultracentrifugation and small-angle x-ray scattering reveals a compact organization of the full-length protein, most likely governed by specific interdomain interactions, rather than an extended structure. Collectively, these data suggest that a high-affinity, CSA-specific binding site is formed by the higher-order structure of the var2CSA extracellular region. These results have important consequences for the development of an effective vaccine and therapeutic inhibitors.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Sulfatos de Condroitina/metabolismo , Espaço Extracelular/química , Plasmodium falciparum/metabolismo , Animais , Linhagem Celular , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Dicroísmo Circular , Decorina , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Humanos , Cinética , Modelos Moleculares , Parasitos/metabolismo , Placenta/metabolismo , Gravidez , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
9.
Malar J ; 9: 267, 2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-20923548

RESUMO

BACKGROUND: The capacity of Plasmodium falciparum-infected erythrocytes to bind uninfected erythrocytes (rosetting) is associated with severe malaria in African children. Rosetting is mediated by a subset of the variant surface antigens PfEMP1 targeted by protective antibody responses. Analysis of the response to rosette-forming parasites and their PfEMP1 adhesive domains is essential for understanding the acquisition of protection against severe malaria. To this end, the antibody response to a rosetting variant was analysed in children recruited with severe or uncomplicated malaria or asymptomatic P. falciparum infection. METHODS: Serum was collected from Beninese children with severe malaria, uncomplicated malaria or P. falciparum asymptomatic infection (N = 65, 37 and 52, respectively) and from immune adults (N = 30) living in the area. Infected erythrocyte surface-reactive IgG, rosette disrupting antibodies and IgG to the parasite crude extract were analysed using the single variant Palo Alto VarO-infected line. IgG, IgG1 and IgG3 to PfEMP1-varO-derived NTS-DBL1α1, CIDRγ and DBL2ßC2 recombinant domains were analysed by ELISA. Antibody responses were compared in the clinical groups. Stability of the response was studied using a blood sampling collected 14 months later from asymptomatic children. RESULTS: Seroprevalence of erythrocyte surface-reactive IgG was high in adults (100%) and asymptomatic children (92.3%) but low in children with severe or uncomplicated malaria (26.1% and 37.8%, respectively). The IgG, IgG1 and IgG3 antibody responses to the varO-derived PfEMP1 domains were significantly higher in asymptomatic children than in children with clinical malaria in a multivariate analysis correcting for age and parasite density at enrolment. They were essentially stable, although levels tended to decrease with time. VarO-surface reactivity correlated positively with IgG reactivity to the rosetting domain varO-NTS-DBL1α1. None of the children sera, including those with surface-reactive antibodies possessed anti-VarO-rosetting activity, and few adults had rosette-disrupting antibodies. CONCLUSIONS: Children with severe and uncomplicated malaria had similar responses. The higher prevalence and level of VarO-reactive antibodies in asymptomatic children compared to children with malaria is consistent with a protective role for anti-VarO antibodies against clinical falciparum malaria. The mechanism of such protection seems independent of rosette-disruption, suggesting that the cytophilic properties of antibodies come into play.


Assuntos
Anticorpos Antiprotozoários/sangue , Portador Sadio/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adulto , Benin , Portador Sadio/patologia , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Humanos , Imunoglobulina G/sangue , Malária Falciparum/patologia , Masculino , Parasitologia/métodos
10.
Infect Immun ; 76(12): 5565-80, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18809668

RESUMO

In the Saimiri sciureus monkey, erythrocytes infected with the varO antigenic variant of the Plasmodium falciparum Palo Alto 89F5 clone bind uninfected red blood cells (rosetting), form autoagglutinates, and have a high multiplication rate, three phenotypic characteristics that are associated with severe malaria in human patients. We report here that varO parasites express a var gene having the characteristics of group A var genes, and we show that the varO Duffy binding-like 1alpha(1) (DBL1alpha(1)) domain is implicated in the rosetting of both S. sciureus and human erythrocytes. The soluble varO N-terminal sequence (NTS)-DBL1alpha(1) recombinant domain, produced in a baculovirus-insect cell system, induced high titers of antibodies that reacted with varO-infected red blood cells and disrupted varO rosettes. varO parasites were culture adapted in vitro using human erythrocytes. They formed rosettes and autoagglutinates, and they had the same surface serotype and expressed the same varO gene as the monkey-propagated parasites. To develop an in vitro model with highly homogeneous varO parasites, rosette purification was combined with positive selection by panning with a varO NTS-DBL1alpha(1)-specific mouse monoclonal antibody. The single-variant, clonal parasites were used to analyze seroprevalence for varO at the village level in a setting where malaria is holoendemic (Dielmo, Senegal). We found 93.6% (95% confidence interval, 89.7 to 96.4%) seroprevalence for varO surface-reacting antibodies and 86.7% (95% confidence interval, 82.8 to 91.6%) seroprevalence for the recombinant NTS-DBL1alpha(1) domain, and virtually all permanent residents had seroconverted by the age of 5 years. These data imply that the varO model is a relevant in vivo and in vitro model for rosetting and autoagglutination that can be used for rational development of vaccine candidates and therapeutic strategies aimed at preventing malaria pathology.


Assuntos
Anticorpos Antiprotozoários , Eritrócitos/metabolismo , Hemaglutinação , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Formação de Roseta/métodos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Sequência de Bases , Técnicas de Cultura de Células , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Malária Falciparum/sangue , Malária Falciparum/diagnóstico , Malária Falciparum/epidemiologia , Camundongos , Dados de Sequência Molecular , Doenças dos Macacos/diagnóstico , Plasmodium falciparum/imunologia , Plasmodium falciparum/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saimiri , Homologia de Sequência do Ácido Nucleico , Sorotipagem/métodos
11.
J Mol Biol ; 366(5): 1523-37, 2007 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-17229439

RESUMO

Apical membrane antigen 1 (AMA1) has an important, but as yet uncharacterised, role in host cell invasion by the malaria parasite, Plasmodium. The protein, which is quite conserved between Plasmodium species, comprises an ectoplasmic region, a single transmembrane segment and a small cytoplasmic domain. The ectoplasmic region, which can induce protective immunity in animal models of human malaria, is a leading vaccine candidate that has entered clinical trials. The monoclonal antibody F8.12.19, raised against the recombinant ectoplasmic region of AMA1 from Plasmodium vivax, cross-reacts with homologues from Plasmodium knowlesi, Plasmodium cynomolgi, Plasmodium berghei and Plasmodium falciparum, as shown by immunofluorescence assays on mature schizonts. The binding of F8.12.19 to recombinant AMA1 from both P. vivax and P. falciparum was measured by surface plasmon resonance, revealing an apparent affinity constant that is about 100-fold weaker for the cross-reacting antigen when compared to the cognate antigen. Crystal structure analysis of Fab F8.12.19 complexed to AMA1 from P. vivax and P. falciparum shows that the monoclonal antibody recognises a discontinuous epitope located on domain III of the ectoplasmic region, the major component being a loop containing a cystine knot. The structures provide a basis for understanding the cross-reactivity. Antibody contacts are made mainly to main-chain and invariant side-chain atoms of AMA1; contact antigen residues that differ in sequence are located at the periphery of the antigen-binding site and can be accommodated at the interface between the two components of the complex. The implications for AMA1 vaccine development are discussed.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Protozoários/imunologia , Proteínas de Membrana/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Sequência de Bases , Reações Cruzadas , Cristalografia por Raios X , Cisteína/química , Epitopos , Técnica Indireta de Fluorescência para Anticorpo , Ligação de Hidrogênio , Proteínas de Membrana/química , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium vivax/química , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
13.
Curr Opin Microbiol ; 9(4): 395-400, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16797221

RESUMO

Malaria is a major global health problem and is caused by the unicellular parasite Plasmodium. Plasmodial surface proteins have important roles in host cell invasion and are responsible for antigenic diversity in this organism. Knowledge of the three-dimensional structure of surface proteins can facilitate our understanding their biological function, and contribute to the development of therapeutic and vaccine strategies against malaria. Structural studies allow rational drug design when ligand- or receptor-binding sites are identified and characterized. Analysis of the three-dimensional distribution of protective antibody epitopes and polymorphic residues can facilitate vaccine candidate optimization. With this in mind, some Plasmodium surface-protein structures have determined by X-ray crystallography or nuclear magnetic resonance.


Assuntos
Proteína 1 de Superfície de Merozoito/química , Plasmodium/química , Proteínas de Protozoários/química , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/fisiologia , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Proteínas de Membrana/fisiologia , Proteína 1 de Superfície de Merozoito/imunologia , Proteína 1 de Superfície de Merozoito/fisiologia , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/fisiologia , Receptores de Superfície Celular/química , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/fisiologia
14.
Mol Biochem Parasitol ; 151(1): 89-99, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17118469

RESUMO

We have previously identified a number of DBLgamma domains in Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) transcripts obtained from placental parasite isolates, showing that they bind specifically to chondroitin sulfate A (CSA) (Khattab A, Kun J, Deloron P, Kremsner PG, Klinkert MQ. Variants of Plasmodium falciparum erythrocyte membrane protein 1 expressed by different placental parasites are closely related and adhere to chondroitin sulfate A. J Infect Dis 2001;183:1165-9). Here we give a more detailed physico-chemical and binding characterisation of the soluble, recombinant DBLgamma domain derived from one of these isolates. Results from circular dichroism and limited proteolysis experiments are consistent with the recombinant domain being expressed with the native fold. Specific binding of DBLgamma to placental cryosections was demonstrated by labeling with antibodies raised against the recombinant domain; binding was diminished after treatment of the cryosections with chondroitinase or by blocking with anti-CSA antibody, showing that CSA mediates the interaction. Binding of the DBLgamma domain to purified placental chondroitin sulfate proteoglycan (CSPG) was also studied using surface plasmon resonance techniques, with DBLgamma as analyte and CSPG immobilised on the sensor chip; these quantitative measurements gave an affinity constant in the mu-molar range under the conditions used. The native conformation of the DBLgamma domain is essential for CSPG recognition since binding to the sensor chip is abolished when the protein is irreversibly reduced. As with the placental cryosections, association was significantly reduced after treating the immobilised CSPG with chondroitinase. Together, these results demonstrate specific interaction between the DBLgamma domain and the placental receptor.


Assuntos
Placenta/metabolismo , Placenta/parasitologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Fenômenos Químicos , Físico-Química , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Dicroísmo Circular , Sequência Conservada , Humanos , Cinética , Dados de Sequência Molecular , Plasmodium falciparum/genética , Plasmodium falciparum/isolamento & purificação , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas de Protozoários/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Cloreto de Sódio , Ressonância de Plasmônio de Superfície , Tripsina/metabolismo
15.
PLoS One ; 12(8): e0183198, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28817634

RESUMO

Malaria, a disease endemic in many tropical and subtropical regions, is caused by infection of the erythrocyte by the apicomplexan parasite Plasmodium. Host-cell invasion is a complex process but two Plasmodium proteins, Apical Membrane Antigen 1 (AMA1) and the Rhoptry Neck protein complex (RON), play a key role. AMA1, present on the surface of the parasite, binds tightly to the RON2 component of the RON protein complex, which is inserted into the erythrocyte membrane during invasion. Blocking the AMA1-RON2 interaction with antibodies or peptides inhibits invasion, underlining its importance in the Plasmodium life cycle and as a target for therapeutic strategies. We describe the crystal structure of the complex formed between AMA1 from P. vivax (PvAMA1) and a peptide derived from the externally exposed region of P. vivax RON2 (PvRON2sp1), and of the heterocomplex formed between P. falciparum AMA1 (PfAMA1) and PvRON2sp1. Binding studies show that the affinity of PvRON2sp1 for PvAMA1 is weaker than that previously reported for the PfRON2sp1-PfAMA1 association. Moreover, while PvRON2sp1 shows strong cross-reactivity with PfAMA1, PfRON2sp1 displays no detectable interaction with PvAMA1. The structures show that the equivalent residues PvRON2-Thr2055 and PfRON2-Arg2041 largely account for this pattern of reactivity.


Assuntos
Reações Cruzadas , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Animais , Ligantes , Ligação Proteica , Proteínas de Protozoários/metabolismo
16.
Mol Biochem Parasitol ; 144(1): 55-67, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16154214

RESUMO

Apical membrane antigen 1 (AMA1) is a membrane protein present in Plasmodium species and is probably common to all apicomplexan parasites. The recent crystal structure of the complete ectoplasmic region of AMA1 from Plasmodium vivax has shown that it comprises three structural domains and that the first two domains are based on the PAN folding motif. Here, we discuss the consequences of this analysis for the three-dimensional structure of AMA1 from other Plasmodium species and other apicomplexan parasites, and for the Plasmodium paralogue MAEBL. Many polar and apolar interactions observed in the PvAMA1 crystal structure are made by residues that are invariant or highly conserved throughout all Plasmodium orthologues; a subgroup of these residues is also present in other apicomplexan orthologues and in MAEBL. These interactions presumably play a key role in defining the protein fold. Previous studies have shown that the ectoplasmic region of AMA1 must be cleaved from the parasite surface for host-cell invasion to proceed. The cleavage site in the crystal structure is not readily accessible to proteases and we discuss possible consequences of this observation. The three-dimensional distribution of polymorphic sites in PfAMA1 shows that these are all on the surface and that their positions are significantly biased to one side of the ectoplasmic region. Of particular note, a flexible segment in domain II, comprising about 40 residues and devoid of polymorphism, carries an epitope recognized by an invasion-inhibitory monoclonal antibody and a T-cell epitope implicated in the human immune response to AMA1.


Assuntos
Antígenos de Protozoários/química , Antígenos de Superfície/química , Proteínas de Membrana/química , Plasmodium/química , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Epitopos de Linfócito T , Modelos Moleculares , Dados de Sequência Molecular , Polimorfismo Genético , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína/genética , Alinhamento de Sequência
17.
Br J Pharmacol ; 144(3): 357-66, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15655523

RESUMO

Alpha3IA (6-(4-pyridyl)-5-(4-methoxyphenyl)-3-carbomethoxy-1-methyl-1H-pyridin-2-one) is a pyridone with higher binding and functional affinity and greater inverse agonist efficacy for GABA(A) receptors containing an alpha3 rather than an alpha1, alpha2 or alpha5 subunit. If doses are selected that minimise the occupancy at these latter subtypes, then the in vivo effects of alpha3IA are most probably mediated by the alpha3 subtype. Alpha3IA has good CNS penetration in rats and mice as measured using a [(3)H]Ro 15-1788 in vivo binding assay. At doses in rats that produce relatively low levels of occupancy (12%) in the cerebellum (i.e. alpha1-containing receptors), alpha3IA (30 mg kg(-1) i.p.), like the nonselective partial inverse agonist N-methyl-beta-carboline-3-carboxamide (FG 7142), not only caused behavioural disruption in an operant, chain-pulling assay but was also anxiogenic in the elevated plus maze, an anxiogenic-like effect that could be blocked with the benzodiazepine antagonist Ro 15-1788 (flumazenil). Neurochemically, alpha3IA (30 mg kg(-1) i.p.) as well as FG 7142 (15 mg kg(-1) i.p.) increased the concentration of the dopamine metabolite 3,4-dihydroxyphenylacetic acid in rat medial prefrontal cortex by 74 and 68%, respectively, relative to vehicle-treated animals, a response that mimicked that seen following immobilisation stress. Taken together, these data demonstrate that an inverse agonist selective for GABA(A) receptors containing an alpha3 subunit is anxiogenic, and suggest that since alpha3-containing GABA(A) receptors play a role in anxiety, then agonists selective for this subtype should be anxiolytic.


Assuntos
Ansiedade/induzido quimicamente , Agonistas GABAérgicos/farmacologia , Receptores de GABA-A/efeitos dos fármacos , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Ansiedade/psicologia , Química Encefálica/efeitos dos fármacos , Carbolinas/farmacologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Fibroblastos/metabolismo , Agonistas GABAérgicos/metabolismo , Humanos , Masculino , Camundongos , Técnicas de Patch-Clamp , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
18.
Sci Rep ; 5: 14868, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26450557

RESUMO

The human malaria parasite, Plasmodium falciparum, is able to evade spleen-mediated clearing from blood stream by sequestering in peripheral organs. This is due to the adhesive properties conferred by the P. falciparum Erythrocyte Membrane Protein 1 (PfEMP1) family exported by the parasite to the surface of infected erythrocytes. Expression of the VAR2CSA variant of PfEMP1 leads to pregnancy-associated malaria, which occurs when infected erythrocytes massively sequester in the placenta by binding to low-sulfated Chondroitin Sulfate A (CSA) present in the intervillous spaces. VAR2CSA is a 350 kDa protein that carries six Duffy-Binding Like (DBL) domains, one Cysteine-rich Inter-Domain Regions (CIDR) and several inter-domain regions. In the present paper, we report for the first time the crystal structure at 2.9 Šof a VAR2CSA double domain, DBL3X-DBL4ε, from the FCR3 strain. DBL3X and DBL4ε share a large contact interface formed by residues that are invariant or highly conserved in VAR2CSA variants, which suggests that these two central DBL domains (DBL3X-DBL4ε) contribute significantly to the structuring of the functional VAR2CSA extracellular region. We have also examined the antigenicity of peptides corresponding to exposed loop regions of the DBL4ε structure.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Placenta/imunologia , Plasmodium falciparum/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Sulfatos de Condroitina/imunologia , Sulfatos de Condroitina/metabolismo , Cristalografia por Raios X , Eritrócitos/imunologia , Eritrócitos/parasitologia , Feminino , Interações Hospedeiro-Parasita/imunologia , Humanos , Soros Imunes/imunologia , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Placenta/metabolismo , Placenta/parasitologia , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Gravidez , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Coelhos , Homologia de Sequência de Aminoácidos
19.
PLoS One ; 10(4): e0123567, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25886591

RESUMO

The malaria parasite Plasmodium knowlesi, previously associated only with infection of macaques, is now known to infect humans as well and has become a significant public health problem in Southeast Asia. This species should therefore be targeted in vaccine and therapeutic strategies against human malaria. Apical Membrane Antigen 1 (AMA1), which plays a role in Plasmodium merozoite invasion of the erythrocyte, is currently being pursued in human vaccine trials against P. falciparum. Recent vaccine trials in macaques using the P. knowlesi orthologue PkAMA1 have shown that it protects against infection by this parasite species and thus should be developed for human vaccination as well. Here, we present the crystal structure of Domains 1 and 2 of the PkAMA1 ectodomain, and of its complex with the invasion-inhibitory monoclonal antibody R31C2. The Domain 2 (D2) loop, which is displaced upon binding the Rhoptry Neck Protein 2 (RON2) receptor, makes significant contacts with the antibody. R31C2 inhibits binding of the Rhoptry Neck Protein 2 (RON2) receptor by steric blocking of the hydrophobic groove and by preventing the displacement of the D2 loop which is essential for exposing the complete binding site on AMA1. R31C2 recognizes a non-polymorphic epitope and should thus be cross-strain reactive. PkAMA1 is much less polymorphic than the P. falciparum and P. vivax orthologues. Unlike these two latter species, there are no polymorphic sites close to the RON2-binding site of PkAMA1, suggesting that P. knowlesi has not developed a mechanism of immune escape from the host's humoral response to AMA1.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Protozoários/química , Proteínas de Membrana/química , Plasmodium knowlesi/imunologia , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/imunologia , Sequência de Bases , Cristalografia por Raios X , Proteínas de Membrana/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Proteínas de Protozoários/imunologia
20.
PLoS One ; 10(7): e0134292, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26222304

RESUMO

Adhesion of Plasmodium falciparum-infected red blood cells (iRBC) to human erythrocytes (i.e. rosetting) is associated with severe malaria. Rosetting results from interactions between a subset of variant PfEMP1 (Plasmodium falciparum erythrocyte membrane protein 1) adhesins and specific erythrocyte receptors. Interfering with such interactions is considered a promising intervention against severe malaria. To evaluate the feasibility of a vaccine strategy targetting rosetting, we have used here the Palo Alto 89F5 VarO rosetting model. PfEMP1-VarO consists of five Duffy-Binding Like domains (DBL1-5) and one Cysteine-rich Interdomain Region (CIDR1). The binding domain has been mapped to DBL1 and the ABO blood group was identified as the erythrocyte receptor. Here, we study the immunogenicity of all six recombinant PfEMP1-VarO domains and the DBL1- CIDR1 Head domain in BALB/c and outbred OF1 mice. Five readouts of antibody responses are explored: ELISA titres on the recombinant antigen, VarO-iRBC immunoblot reactivity, VarO-iRBC surface-reactivity, capacity to disrupt VarO rosettes and the capacity to prevent VarO rosette formation. For three domains, we explore influence of the expression system on antigenicity and immunogenicity. We show that correctly folded PfEMP1 domains elicit high antibody titres and induce a homogeneous response in outbred and BALB/c mice after three injections. High levels of rosette-disrupting and rosette-preventing antibodies are induced by DBL1 and the Head domain. Reduced-alkylated or denatured proteins fail to induce surface-reacting and rosette-disrupting antibodies, indicating that surface epitopes are conformational. We also report limited cross-reactivity between some PfEMP1 VarO domains. These results highlight the high immunogenicity of the individual domains in outbred animals and provide a strong basis for a rational vaccination strategy targeting rosetting.


Assuntos
Adesinas Bacterianas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Reações Cruzadas , Epitopos/química , Epitopos/genética , Eritrócitos/parasitologia , Feminino , Humanos , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Formação de Roseta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA