Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.360
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 22(3): 347-357, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33432229

RESUMEN

Activated Vγ9Vδ2 (γδ2) T lymphocytes that sense parasite-produced phosphoantigens are expanded in Plasmodium falciparum-infected patients. Although previous studies suggested that γδ2 T cells help control erythrocytic malaria, whether γδ2 T cells recognize infected red blood cells (iRBCs) was uncertain. Here we show that iRBCs stained for the phosphoantigen sensor butyrophilin 3A1 (BTN3A1). γδ2 T cells formed immune synapses and lysed iRBCs in a contact, phosphoantigen, BTN3A1 and degranulation-dependent manner, killing intracellular parasites. Granulysin released into the synapse lysed iRBCs and delivered death-inducing granzymes to the parasite. All intra-erythrocytic parasites were susceptible, but schizonts were most sensitive. A second protective γδ2 T cell mechanism was identified. In the presence of patient serum, γδ2 T cells phagocytosed and degraded opsonized iRBCs in a CD16-dependent manner, decreasing parasite multiplication. Thus, γδ2 T cells have two ways to control blood-stage malaria-γδ T cell antigen receptor (TCR)-mediated degranulation and phagocytosis of antibody-coated iRBCs.


Asunto(s)
Antígenos de Protozoos/inmunología , Citotoxicidad Inmunológica , Eritrocitos/inmunología , Linfocitos Intraepiteliales/inmunología , Activación de Linfocitos , Malaria Falciparum/inmunología , Fagocitosis , Plasmodium falciparum/microbiología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígenos de Protozoos/sangre , Boston , Brasil , Butirofilinas/metabolismo , Células Cultivadas , Eritrocitos/metabolismo , Eritrocitos/parasitología , Femenino , Granzimas/metabolismo , Interacciones Huésped-Parásitos , Humanos , Sinapsis Inmunológicas/metabolismo , Sinapsis Inmunológicas/parasitología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Masculino , Plasmodium falciparum/crecimiento & desarrollo
2.
Nat Immunol ; 22(5): 654-665, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33888898

RESUMEN

Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.


Asunto(s)
Inmunidad Adaptativa/inmunología , Susceptibilidad a Enfermedades/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Inmunidad Adaptativa/genética , Adolescente , Adulto , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Población Negra/genética , Células Dendríticas/inmunología , Susceptibilidad a Enfermedades/sangre , Susceptibilidad a Enfermedades/parasitología , Femenino , Voluntarios Sanos , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interferón gamma/metabolismo , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Masculino , RNA-Seq , Análisis de Sistemas , Linfocitos T/inmunología , Linfocitos T/metabolismo , Población Blanca/genética , Adulto Joven
3.
Immunity ; 57(8): 1769-1779.e4, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-38901428

RESUMEN

Many infections, including malaria, are associated with an increase in autoantibodies (AAbs). Prior studies have reported an association between genetic markers of susceptibility to autoimmune disease and resistance to malaria, but the underlying mechanisms are unclear. Here, we performed a longitudinal study of children and adults (n = 602) in Mali and found that high levels of plasma AAbs before the malaria season independently predicted a reduced risk of clinical malaria in children during the ensuing malaria season. Baseline AAb seroprevalence increased with age and asymptomatic Plasmodium falciparum infection. We found that AAbs purified from the plasma of protected individuals inhibit the growth of blood-stage parasites and bind P. falciparum proteins that mediate parasite invasion. Protected individuals had higher plasma immunoglobulin G (IgG) reactivity against 33 of the 123 antigens assessed in an autoantigen microarray. This study provides evidence in support of the hypothesis that a propensity toward autoimmunity offers a survival advantage against malaria.


Asunto(s)
Autoanticuerpos , Inmunoglobulina G , Malaria Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/inmunología , Autoanticuerpos/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Niño , Preescolar , Adulto , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Femenino , Malí , Masculino , Adolescente , Anticuerpos Antiprotozoarios/inmunología , Estudios Longitudinales , Lactante , Antígenos de Protozoos/inmunología , Adulto Joven , Autoantígenos/inmunología , Estudios Seroepidemiológicos , Persona de Mediana Edad
4.
Immunity ; 56(2): 234-236, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36792569

RESUMEN

The development of a transmission-blocking vaccine (TBV) against malaria is hampered by poor understanding of functional antibody responses. In this issue of Immunity, Fabra-Garcia et al., Ivanochko et al., and Tang et al. isolate human monoclonal antibodies against the two most promising TBV candidates, Pfs48/45 and Pfs230, and map the epitopes responsible for potent transmission-reducing activity.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Humanos , Malaria Falciparum/prevención & control , Proteínas Protozoarias , Anticuerpos Antiprotozoarios , Malaria/prevención & control , Plasmodium falciparum , Antígenos de Protozoos
5.
Immunity ; 56(2): 420-432.e7, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36792575

RESUMEN

Pfs230 is essential for Plasmodium falciparum transmission to mosquitoes and is the protein targeted by the most advanced malaria-transmission-blocking vaccine candidate. Prior understanding of functional epitopes on Pfs230 is based on two monoclonal antibodies (mAbs) with moderate transmission-reducing activity (TRA), elicited from subunit immunization. Here, we screened the B cell repertoire of two naturally exposed individuals possessing serum TRA and identified five potent mAbs from sixteen Pfs230 domain-1-specific mAbs. Structures of three potent and three low-activity antibodies bound to Pfs230 domain 1 revealed four distinct epitopes. Highly potent mAbs from natural infection recognized a common conformational epitope that is highly conserved across P. falciparum field isolates, while antibodies with negligible TRA derived from natural infection or immunization recognized three distinct sites. Our study provides molecular blueprints describing P. falciparum TRA, informed by contrasting potent and non-functional epitopes elicited by natural exposure and vaccination.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Humanos , Animales , Plasmodium falciparum , Epítopos , Proteínas Protozoarias , Antígenos de Protozoos , Anticuerpos Monoclonales , Anticuerpos Antiprotozoarios , Malaria Falciparum/prevención & control
6.
Immunity ; 55(9): 1680-1692.e8, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35977542

RESUMEN

Malaria transmission-blocking vaccines (TBVs) aim to elicit human antibodies that inhibit sporogonic development of Plasmodium falciparum in mosquitoes, thereby preventing onward transmission. Pfs48/45 is a leading clinical TBV candidate antigen and is recognized by the most potent transmission-blocking monoclonal antibody (mAb) yet described; still, clinical development of Pfs48/45 antigens has been hindered, largely by its poor biochemical characteristics. Here, we used structure-based computational approaches to design Pfs48/45 antigens stabilized in the conformation recognized by the most potently inhibitory mAb, achieving >25°C higher thermostability compared with the wild-type protein. Antibodies elicited in mice immunized with these engineered antigens displayed on liposome-based or protein nanoparticle-based vaccine platforms exhibited 1-2 orders of magnitude superior transmission-reducing activity, compared with immunogens bearing the wild-type antigen, driven by improved antibody quality. Our data provide the founding principles for using molecular stabilization solely from antibody structure-function information to drive improved immune responses against a parasitic vaccine target.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Animales , Anticuerpos Bloqueadores , Anticuerpos Monoclonales , Anticuerpos Antiprotozoarios , Formación de Anticuerpos , Antígenos de Protozoos , Humanos , Malaria Falciparum/prevención & control , Glicoproteínas de Membrana , Ratones , Plasmodium falciparum , Proteínas Protozoarias , Vacunación
7.
Cell ; 164(4): 601-2, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26871626

RESUMEN

While searching for new therapeutics against malaria, Lanzavecchia and colleagues discovered that antibodies can be diversified by DNA sequences encoded outside of antibody genes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Variación Antigénica/inmunología , Antígenos de Protozoos/inmunología , Malaria/inmunología , Mutagénesis Insercional/genética , Plasmodium falciparum/inmunología , Receptores Inmunológicos/inmunología , Humanos
8.
Nature ; 625(7995): 578-584, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38123677

RESUMEN

The symptoms of malaria occur during the blood stage of infection, when parasites invade and replicate within human erythrocytes. The PfPCRCR complex1, containing PfRH5 (refs. 2,3), PfCyRPA, PfRIPR, PfCSS and PfPTRAMP, is essential for erythrocyte invasion by the deadliest human malaria parasite, Plasmodium falciparum. Invasion can be prevented by antibodies3-6 or nanobodies1 against each of these conserved proteins, making them the leading blood-stage malaria vaccine candidates. However, little is known about how PfPCRCR functions during invasion. Here we present the structure of the PfRCR complex7,8, containing PfRH5, PfCyRPA and PfRIPR, determined by cryogenic-electron microscopy. We test the hypothesis that PfRH5 opens to insert into the membrane9, instead showing that a rigid, disulfide-locked PfRH5 can mediate efficient erythrocyte invasion. We show, through modelling and an erythrocyte-binding assay, that PfCyRPA-binding antibodies5 neutralize invasion through a steric mechanism. We determine the structure of PfRIPR, showing that it consists of an ordered, multidomain core flexibly linked to an elongated tail. We also show that the elongated tail of PfRIPR, which is the target of growth-neutralizing antibodies6, binds to the PfCSS-PfPTRAMP complex on the parasite membrane. A modular PfRIPR is therefore linked to the merozoite membrane through an elongated tail, and its structured core presents PfCyRPA and PfRH5 to interact with erythrocyte receptors. This provides fresh insight into the molecular mechanism of erythrocyte invasion and opens the way to new approaches in rational vaccine design.


Asunto(s)
Eritrocitos , Malaria Falciparum , Complejos Multiproteicos , Parásitos , Plasmodium falciparum , Proteínas Protozoarias , Animales , Humanos , Anticuerpos Neutralizantes/inmunología , Antígenos de Protozoos/química , Antígenos de Protozoos/inmunología , Microscopía por Crioelectrón , Disulfuros/química , Disulfuros/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Merozoítos/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Parásitos/metabolismo , Parásitos/patogenicidad , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/ultraestructura
9.
Cell ; 158(4): 916-928, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126794

RESUMEN

A central problem in biology is to identify gene function. One approach is to infer function in large supergenomic networks of interactions and ancestral relationships among genes; however, their analysis can be computationally prohibitive. We show here that these biological networks are compressible. They can be shrunk dramatically by eliminating redundant evolutionary relationships, and this process is efficient because in these networks the number of compressible elements rises linearly rather than exponentially as in other complex networks. Compression enables global network analysis to computationally harness hundreds of interconnected genomes and to produce functional predictions. As a demonstration, we show that the essential, but functionally uncharacterized Plasmodium falciparum antigen EXP1 is a membrane glutathione S-transferase. EXP1 efficiently degrades cytotoxic hematin, is potently inhibited by artesunate, and is associated with artesunate metabolism and susceptibility in drug-pressured malaria parasites. These data implicate EXP1 in the mode of action of a frontline antimalarial drug.


Asunto(s)
Antígenos de Protozoos/aislamiento & purificación , Compresión de Datos , Genómica/métodos , Plasmodium falciparum/enzimología , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Antimaláricos/farmacología , Artemisininas/farmacología , Artesunato , Dominio Catalítico , Hemina/metabolismo , Modelos Genéticos , Plasmodium falciparum/genética
10.
Cell ; 148(1-2): 201-12, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22265412

RESUMEN

Hundreds of effector proteins of the human malaria parasite Plasmodium falciparum constitute a "secretome" carrying a host-targeting (HT) signal, which predicts their export from the intracellular pathogen into the surrounding erythrocyte. Cleavage of the HT signal by a parasite endoplasmic reticulum (ER) protease, plasmepsin V, is the proposed export mechanism. Here, we show that the HT signal facilitates export by recognition of the lipid phosphatidylinositol-3-phosphate (PI(3)P) in the ER, prior to and independent of protease action. Secretome HT signals, including those of major virulence determinants, bind PI(3)P with nanomolar affinity and amino acid specificities displayed by HT-mediated export. PI(3)P-enriched regions are detected within the parasite's ER and colocalize with endogenous HT signal on ER precursors, which also display high-affinity binding to PI(3)P. A related pathogenic oomycete's HT signal export is dependent on PI(3)P binding, without cleavage by plasmepsin V. Thus, PI(3)P in the ER functions in mechanisms of secretion and pathogenesis.


Asunto(s)
Eritrocitos/parasitología , Malaria Falciparum/parasitología , Fosfatos de Fosfatidilinositol/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Antígenos de Protozoos/química , Antígenos de Protozoos/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Retículo Endoplásmico/metabolismo , Eritrocitos/metabolismo , Humanos , Malaria Falciparum/patología , Datos de Secuencia Molecular , Plasmodium falciparum/citología , Señales de Clasificación de Proteína , Transporte de Proteínas , Proteínas Protozoarias/química
11.
Nature ; 592(7855): 639-643, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33790470

RESUMEN

Some Plasmodium falciparum repetitive interspersed families of polypeptides (RIFINs)-variant surface antigens that are expressed on infected erythrocytes1-bind to the inhibitory receptor LAIR1, and insertion of DNA that encodes LAIR1 into immunoglobulin genes generates RIFIN-specific antibodies2,3. Here we address the general relevance of this finding by searching for antibodies that incorporate LILRB1, another inhibitory receptor that binds to ß2 microglobulin and RIFINs through their apical domains4,5. By screening plasma from a cohort of donors from Mali, we identified individuals with LILRB1-containing antibodies. B cell clones isolated from three donors showed large DNA insertions in the switch region that encodes non-apical LILRB1 extracellular domain 3 and 4 (D3D4) or D3 alone in the variable-constant (VH-CH1) elbow. Through mass spectrometry and binding assays, we identified a large set of RIFINs that bind to LILRB1 D3. Crystal and cryo-electron microscopy structures of a RIFIN in complex with either LILRB1 D3D4 or a D3D4-containing antibody Fab revealed a mode of RIFIN-LILRB1 D3 interaction that is similar to that of RIFIN-LAIR1. The Fab showed an unconventional triangular architecture with the inserted LILRB1 domains opening up the VH-CH1 elbow without affecting VH-VL or CH1-CL pairing. Collectively, these findings show that RIFINs bind to LILRB1 through D3 and illustrate, with a naturally selected example, the general principle of creating novel antibodies by inserting receptor domains into the VH-CH1 elbow.


Asunto(s)
Anticuerpos/química , Anticuerpos/inmunología , Antígenos de Protozoos/química , Antígenos de Protozoos/inmunología , Microscopía por Crioelectrón , Receptor Leucocitario Tipo Inmunoglobulina B1/química , Plasmodium falciparum/química , Plasmodium falciparum/inmunología , Adolescente , Adulto , Secuencia de Aminoácidos , Anticuerpos/ultraestructura , Especificidad de Anticuerpos , Antígenos de Protozoos/ultraestructura , Sitios de Unión de Anticuerpos , Niño , Preescolar , Estudios de Cohortes , Humanos , Lactante , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Malí , Modelos Moleculares , Plasmodium falciparum/genética , Plasmodium falciparum/ultraestructura , Dominios Proteicos , Adulto Joven
12.
Nature ; 595(7865): 96-100, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34040257

RESUMEN

Trypanosomes are protozoan parasites that cause infectious diseases, including African trypanosomiasis (sleeping sickness) in humans and nagana in economically important livestock1,2. An effective vaccine against trypanosomes would be an important control tool, but the parasite has evolved sophisticated immunoprotective mechanisms-including antigenic variation3-that present an apparently insurmountable barrier to vaccination. Here we show, using a systematic genome-led vaccinology approach and a mouse model of Trypanosoma vivax infection4, that protective invariant subunit vaccine antigens can be identified. Vaccination with a single recombinant protein comprising the extracellular region of a conserved cell-surface protein that is localized to the flagellum membrane (which we term 'invariant flagellum antigen from T. vivax') induced long-lasting protection. Immunity was passively transferred with immune serum, and recombinant monoclonal antibodies to this protein could induce sterile protection and revealed several mechanisms of antibody-mediated immunity, including a major role for complement. Our discovery identifies a vaccine candidate for an important parasitic disease that has constrained socioeconomic development in countries in sub-Saharan Africa5, and provides evidence that highly protective vaccines against trypanosome infections can be achieved.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas Antiprotozoos/inmunología , Trypanosoma vivax/inmunología , Tripanosomiasis Africana/inmunología , Tripanosomiasis Africana/prevención & control , Animales , Antígenos de Protozoos/química , Proteínas del Sistema Complemento/inmunología , Secuencia Conservada/inmunología , Modelos Animales de Enfermedad , Femenino , Flagelos/química , Flagelos/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunas Antiprotozoos/química , Factores de Tiempo , Trypanosoma vivax/química , Trypanosoma vivax/citología , Tripanosomiasis Africana/parasitología , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
13.
PLoS Pathog ; 20(3): e1011879, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38437239

RESUMEN

Placental accumulation of Plasmodium falciparum infected erythrocytes results in maternal anemia, low birth weight, and pregnancy loss. The parasite protein VAR2CSA facilitates the accumulation of infected erythrocytes in the placenta through interaction with the host receptor chondroitin sulfate A (CSA). Antibodies that prevent the VAR2CSA-CSA interaction correlate with protection from placental malaria, and VAR2CSA is a high-priority placental malaria vaccine antigen. Here, structure-guided design leveraging the full-length structures of VAR2CSA produced a stable immunogen that retains the critical conserved functional elements of VAR2CSA. The design expressed with a six-fold greater yield than the full-length protein and elicited antibodies that prevent adhesion of infected erythrocytes to CSA. The reduced size and adaptability of the designed immunogen enable efficient production of multiple variants of VAR2CSA for use in a cocktail vaccination strategy to increase the breadth of protection. These designs form strong foundations for the development of potent broadly protective placental malaria vaccines.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Humanos , Embarazo , Femenino , Placenta/metabolismo , Malaria Falciparum/parasitología , Anticuerpos Antiprotozoarios , Plasmodium falciparum/metabolismo , Antígenos de Protozoos , Sulfatos de Condroitina/metabolismo , Eritrocitos/parasitología
14.
PLoS Pathog ; 20(6): e1012334, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38941356

RESUMEN

Plasmodium vivax serological exposure markers (SEMs) have emerged as promising tools for the actionable surveillance and implementation of targeted interventions to accelerate malaria elimination. To determine the dynamic profiles of SEMs in current and past P. vivax infections, we screened and selected 11 P. vivax proteins from 210 putative proteins using protein arrays, with a set of serum samples obtained from patients with acute P. vivax and documented past P. vivax infections. Then we used a murine protein immune model to initially investigate the humoral and memory B cell response involved in the generation of long-lived antibodies. We show that of the 11 proteins, especially C-terminal 42-kDa region of P. vivax merozoite surface protein 1 (PvMSP1-42) induced longer-lasting long-lived antibodies, as these antibodies were detected in individuals infected with P. vivax in the 1960-1970s who were not re-infected until 2012. In addition, we provide a potential mechanism for the maintenance of long-lived antibodies after the induction of PvMSP1-42. The results indicate that PvMSP1-42 induces more CD73+CD80+ memory B cells (MBCs) compared to P. vivax GPI-anchored micronemal antigen (PvGAMA), allowing IgG anti-PvMSP1-42 antibodies to be maintained for a long time.


Asunto(s)
Anticuerpos Antiprotozoarios , Malaria Vivax , Células B de Memoria , Proteína 1 de Superficie de Merozoito , Plasmodium vivax , Plasmodium vivax/inmunología , Humanos , Malaria Vivax/inmunología , Anticuerpos Antiprotozoarios/inmunología , Animales , Proteína 1 de Superficie de Merozoito/inmunología , Ratones , Células B de Memoria/inmunología , Inmunidad Humoral/inmunología , Biomarcadores/sangre , Femenino , Memoria Inmunológica/inmunología , Linfocitos B/inmunología , Antígenos de Protozoos/inmunología
15.
Immunity ; 47(6): 1197-1209.e10, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29195810

RESUMEN

Antibodies against the NANP repeat of circumsporozoite protein (CSP), the major surface antigen of Plasmodium falciparum (Pf) sporozoites, can protect from malaria in animal models but protective humoral immunity is difficult to induce in humans. Here we cloned and characterized rare affinity-matured human NANP-reactive memory B cell antibodies elicited by natural Pf exposure that potently inhibited parasite transmission and development in vivo. We unveiled the molecular details of antibody binding to two distinct protective epitopes within the NANP repeat. NANP repeat recognition was largely mediated by germline encoded and immunoglobulin (Ig) heavy-chain complementarity determining region 3 (HCDR3) residues, whereas affinity maturation contributed predominantly to stabilizing the antigen-binding site conformation. Combined, our findings illustrate the power of exploring human anti-CSP antibody responses to develop tools for malaria control in the mammalian and the mosquito vector and provide a molecular basis for the structure-based design of next-generation CSP malaria vaccines.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Inmunidad Humoral , Cadenas Pesadas de Inmunoglobulina/inmunología , Malaria Falciparum/prevención & control , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/química , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Linfocitos B/inmunología , Linfocitos B/parasitología , Cristalografía por Rayos X , Epítopos/química , Epítopos/inmunología , Femenino , Expresión Génica , Humanos , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/química , Memoria Inmunológica , Malaria/inmunología , Malaria/parasitología , Malaria/prevención & control , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Masculino , Ratones , Modelos Moleculares , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esporozoítos/química , Esporozoítos/inmunología
16.
Nature ; 582(7810): 104-108, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32427965

RESUMEN

Malaria caused by Plasmodium falciparum remains the leading single-agent cause of mortality in children1, yet the promise of an effective vaccine has not been fulfilled. Here, using our previously described differential screening method to analyse the proteome of blood-stage P. falciparum parasites2, we identify P. falciparum glutamic-acid-rich protein (PfGARP) as a parasite antigen that is recognized by antibodies in the plasma of children who are relatively resistant-but not those who are susceptible-to malaria caused by P. falciparum. PfGARP is a parasite antigen of 80 kDa that is expressed on the exofacial surface of erythrocytes infected by early-to-late-trophozoite-stage parasites. We demonstrate that antibodies against PfGARP kill trophozoite-infected erythrocytes in culture by inducing programmed cell death in the parasites, and that vaccinating non-human primates with PfGARP partially protects against a challenge with P. falciparum. Furthermore, our longitudinal cohort studies showed that, compared to individuals who had naturally occurring anti-PfGARP antibodies, Tanzanian children without anti-PfGARP antibodies had a 2.5-fold-higher risk of severe malaria and Kenyan adolescents and adults without these antibodies had a twofold-higher parasite density. By killing trophozoite-infected erythrocytes, PfGARP could synergize with other vaccines that target parasite invasion of hepatocytes or the invasion of and egress from erythrocytes.


Asunto(s)
Apoptosis/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Parásitos/inmunología , Plasmodium falciparum/citología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Adolescente , Adulto , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/química , Antígenos de Protozoos/inmunología , Aotidae/inmunología , Aotidae/parasitología , Caspasas/metabolismo , Niño , Estudios de Cohortes , ADN Protozoario/química , ADN Protozoario/metabolismo , Activación Enzimática , Eritrocitos/parasitología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Kenia , Vacunas contra la Malaria/inmunología , Malaria Falciparum/parasitología , Masculino , Ratones , Parásitos/citología , Parásitos/crecimiento & desarrollo , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/química , Tanzanía , Trofozoítos/citología , Trofozoítos/crecimiento & desarrollo , Trofozoítos/inmunología , Vacuolas/inmunología
17.
Proc Natl Acad Sci U S A ; 120(1): e2215003120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36577076

RESUMEN

We used a transgenic parasite in which Plasmodium falciparum parasites were genetically modified to express Plasmodium vivax apical membrane antigen 1 (PvAMA1) protein in place of PfAMA1 to study PvAMA1-mediated invasion. In P. falciparum, AMA1 interaction with rhoptry neck protein 2 (RON2) is known to be crucial for invasion, and PfRON2 peptides (PfRON2p) blocked the invasion of PfAMA1 wild-type parasites. However, PfRON2p has no effect on the invasion of transgenic parasites expressing PvAMA1 indicating that PfRON2 had no role in the invasion of PvAMA1 transgenic parasites. Interestingly, PvRON2p blocked the invasion of PvAMA1 transgenic parasites in a dose-dependent manner. We found that recombinant PvAMA1 domains 1 and 2 (rPvAMA1) bound to reticulocytes and normocytes indicating that PvAMA1 directly interacts with erythrocytes during the invasion, and invasion blocking of PvRON2p may result from it interfering with PvAMA1 binding to erythrocytes. It was previously shown that the peptide containing Loop1a of PvAMA1 (PvAMA1 Loop1a) is also bound to reticulocytes. We found that the Loop1a peptide blocked the binding of PvAMA1 to erythrocytes. PvAMA1 Loop1a has no polymorphisms in contrast to other PvAMA1 loops and may be an attractive vaccine target. We thus present the evidence that PvAMA1 binds to erythrocytes in addition to interacting with PvRON2 suggesting that the P. vivax merozoites may exploit complex pathways during the invasion process.


Asunto(s)
Malaria Falciparum , Plasmodium vivax , Humanos , Proteínas Protozoarias/química , Antígenos de Protozoos , Eritrocitos/metabolismo , Plasmodium falciparum/metabolismo , Reticulocitos/metabolismo
18.
PLoS Pathog ; 19(6): e1011442, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37307293

RESUMEN

A signature remains elusive of naturally-acquired immunity against Plasmodium falciparum. We identified P. falciparum in a 14-month cohort of 239 people in Kenya, genotyped at immunogenic parasite targets expressed in the pre-erythrocytic (circumsporozoite protein, CSP) and blood (apical membrane antigen 1, AMA-1) stages, and classified into epitope type based on variants in the DV10, Th2R, and Th3R epitopes in CSP and the c1L region of AMA-1. Compared to asymptomatic index infections, symptomatic malaria was associated with reduced reinfection by parasites bearing homologous CSP-Th2R (adjusted hazard ratio [aHR]:0.63; 95% CI:0.45-0.89; p = 0.008) CSP-Th3R (aHR:0.71; 95% CI:0.52-0.97; p = 0.033), and AMA-1 c1L (aHR:0.63; 95% CI:0.43-0.94; p = 0.022) epitope types. The association of symptomatic malaria with reduced hazard of homologous reinfection was strongest for rare epitope types. Symptomatic malaria provides more durable protection against reinfection with parasites bearing homologous epitope types. The phenotype represents a legible molecular epidemiologic signature of naturally-acquired immunity by which to identify new antigen targets.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Parásitos , Animales , Plasmodium falciparum/metabolismo , Reinfección , Proteínas Protozoarias/metabolismo , Malaria/parasitología , Malaria Falciparum/parasitología , Antígenos de Protozoos , Epítopos/genética , Anticuerpos Antiprotozoarios/metabolismo
19.
PLoS Pathog ; 19(5): e1011370, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37228009

RESUMEN

VAR2CSA is the Plasmodium falciparum variant surface antigen that mediates binding of infected erythrocytes to chondroitin sulfate A (CSA) and their sequestration in intervillous spaces of the placenta, leading to placental malaria (PM). Relatively high polymorphism in VAR2CSA sequences has hindered development of a vaccine that induces broadly neutralizing immunity. Recent research has highlighted that a broadly reactive human monoclonal antibody, called PAM1.4, binds to multiple conserved residues of different subfragments of VAR2CSA, forming a conformational epitope. In this short perspective, we describe evidence that residues located in the interdomain-1 fragment of VAR2CSA within the PAM1.4 binding epitope might be critical to broad reactivity of the antibody. Future investigation into broadly reactive anti-VAR2CSA antibodies may be important for the following: (1) identification of similar conformation epitopes targeted by broadly neutralizing antibodies; and (2) understanding different immune evasion mechanisms used by placenta-binding parasites through VAR2CSA polymorphism in critical epitopes.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Femenino , Embarazo , Humanos , Placenta/metabolismo , Epítopos/genética , Malaria Falciparum/prevención & control , Plasmodium falciparum/metabolismo , Antígenos de Protozoos , Anticuerpos Antiprotozoarios , Sulfatos de Condroitina/metabolismo , Eritrocitos/parasitología
20.
Blood ; 142(23): 2016-2028, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-37832027

RESUMEN

The malaria parasite Plasmodium falciparum invades and replicates asexually within human erythrocytes. CD44 expressed on erythrocytes was previously identified as an important host factor for P falciparum infection through a forward genetic screen, but little is known about its regulation or function in these cells, nor how it may be used by the parasite. We found that CD44 can be efficiently deleted from primary human hematopoietic stem cells using CRISPR/Cas9 genome editing, and that the efficiency of ex vivo erythropoiesis to enucleated cultured red blood cells (cRBCs) is not affected by lack of CD44. However, the rate of P falciparum invasion was reduced in CD44-null cRBCs relative to isogenic wild-type control cells, validating CD44 as an important host factor for this parasite. We identified 2 P falciparum invasion ligands as binding partners for CD44, erythrocyte binding antigen 175 (EBA-175) and EBA-140 and demonstrated that their ability to bind to human erythrocytes relies primarily on their canonical receptors, glycophorin A and glycophorin C, respectively. We further show that EBA-175 induces phosphorylation of erythrocyte cytoskeletal proteins in a CD44-dependent manner. Our findings support a model in which P falciparum exploits CD44 as a coreceptor during invasion of human erythrocytes, stimulating CD44-dependent phosphorylation of host cytoskeletal proteins that alter host cell deformability and facilitate parasite entry.


Asunto(s)
Eritrocitos , Malaria Falciparum , Plasmodium falciparum , Humanos , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Proteínas del Citoesqueleto , Eritrocitos/metabolismo , Eritrocitos/parasitología , Receptores de Hialuranos/metabolismo , Malaria Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Unión Proteica , Proteínas Protozoarias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA