Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 32: 157-87, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24655294

RESUMEN

Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the world's most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasite's complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.


Asunto(s)
Culicidae/inmunología , Interacciones Huésped-Patógeno/inmunología , Malaria/inmunología , Plasmodium/inmunología , Animales , Culicidae/parasitología , Humanos , Estadios del Ciclo de Vida , Malaria/parasitología , Malaria/prevención & control , Plasmodium/crecimiento & desarrollo , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología
2.
Immunity ; 2024 Jun 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.

3.
Cell ; 159(6): 1277-89, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25480293

RESUMEN

Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:ß-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galß1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans.


Asunto(s)
Escherichia coli/fisiología , Inmunoglobulina M/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/transmisión , Plasmodium/fisiología , Polisacáridos/inmunología , Adulto , Animales , Anopheles/parasitología , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Autoantígenos/inmunología , Línea Celular Tumoral , Niño , Escherichia coli/clasificación , Escherichia coli/inmunología , Femenino , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Humanos , Inmunoglobulina M/sangre , Malaria Falciparum/microbiología , Malaria Falciparum/parasitología , Ratones , Plasmodium/clasificación , Plasmodium/crecimiento & desarrollo , Plasmodium/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/fisiología , Esporozoítos/inmunología , Receptor Toll-Like 9/agonistas
4.
Immunity ; 51(4): 750-765.e10, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31492649

RESUMEN

Immunity that controls parasitemia and inflammation during Plasmodium falciparum (Pf) malaria can be acquired with repeated infections. A limited understanding of this complex immune response impedes the development of vaccines and adjunctive therapies. We conducted a prospective systems biology study of children who differed in their ability to control parasitemia and fever following Pf infection. By integrating whole-blood transcriptomics, flow-cytometric analysis, and plasma cytokine and antibody profiles, we demonstrate that a pre-infection signature of B cell enrichment, upregulation of T helper type 1 (Th1) and Th2 cell-associated pathways, including interferon responses, and p53 activation associated with control of malarial fever and coordinated with Pf-specific immunoglobulin G (IgG) and Fc receptor activation to control parasitemia. Our hypothesis-generating approach identified host molecules that may contribute to differential clinical outcomes during Pf infection. As a proof of concept, we have shown that enhanced p53 expression in monocytes attenuated Plasmodium-induced inflammation and predicted protection from fever.


Asunto(s)
Linfocitos B/inmunología , Proteínas Sanguíneas/metabolismo , Inflamación/metabolismo , Malaria Falciparum/metabolismo , Plasmodium falciparum/fisiología , Células TH1/inmunología , Células Th2/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Adulto , Animales , Anticuerpos Antiprotozoarios/metabolismo , Niño , Preescolar , Resistencia a la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Lactante , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estudios Prospectivos , Receptores Fc/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Adulto Joven
5.
N Engl J Med ; 390(17): 1549-1559, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38669354

RESUMEN

BACKGROUND: Subcutaneous administration of the monoclonal antibody L9LS protected adults against controlled Plasmodium falciparum infection in a phase 1 trial. Whether a monoclonal antibody administered subcutaneously can protect children from P. falciparum infection in a region where this organism is endemic is unclear. METHODS: We conducted a phase 2 trial in Mali to assess the safety and efficacy of subcutaneous administration of L9LS in children 6 to 10 years of age over a 6-month malaria season. In part A of the trial, safety was assessed at three dose levels in adults, followed by assessment at two dose levels in children. In part B of the trial, children were randomly assigned, in a 1:1:1 ratio, to receive 150 mg of L9LS, 300 mg of L9LS, or placebo. The primary efficacy end point, assessed in a time-to-event analysis, was the first P. falciparum infection, as detected on blood smear performed at least every 2 weeks for 24 weeks. A secondary efficacy end point was the first episode of clinical malaria, as assessed in a time-to-event analysis. RESULTS: No safety concerns were identified in the dose-escalation part of the trial (part A). In part B, 225 children underwent randomization, with 75 children assigned to each group. No safety concerns were identified in part B. P. falciparum infection occurred in 36 participants (48%) in the 150-mg group, in 30 (40%) in the 300-mg group, and in 61 (81%) in the placebo group. The efficacy of L9LS against P. falciparum infection, as compared with placebo, was 66% (adjusted confidence interval [95% CI], 45 to 79) with the 150-mg dose and 70% (adjusted 95% CI, 50 to 82) with the 300-mg dose (P<0.001 for both comparisons). Efficacy against clinical malaria was 67% (adjusted 95% CI, 39 to 82) with the 150-mg dose and 77% (adjusted 95% CI, 55 to 89) with the 300-mg dose (P<0.001 for both comparisons). CONCLUSIONS: Subcutaneous administration of L9LS to children was protective against P. falciparum infection and clinical malaria over a period of 6 months. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT05304611.).


Asunto(s)
Anticuerpos Monoclonales Humanizados , Malaria Falciparum , Adulto , Niño , Femenino , Humanos , Masculino , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Enfermedades Endémicas/prevención & control , Inyecciones Subcutáneas , Estimación de Kaplan-Meier , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/epidemiología , Malaria Falciparum/prevención & control , Malí/epidemiología , Plasmodium falciparum , Resultado del Tratamiento , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Terapia por Observación Directa , Combinación Arteméter y Lumefantrina/administración & dosificación , Combinación Arteméter y Lumefantrina/uso terapéutico , Adulto Joven , Persona de Mediana Edad
6.
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
7.
N Engl J Med ; 387(20): 1833-1842, 2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-36317783

RESUMEN

BACKGROUND: CIS43LS is a monoclonal antibody that was shown to protect against controlled Plasmodium falciparum infection in a phase 1 clinical trial. Whether a monoclonal antibody can prevent P. falciparum infection in a region in which the infection is endemic is unknown. METHODS: We conducted a phase 2 trial to assess the safety and efficacy of a single intravenous infusion of CIS43LS against P. falciparum infection in healthy adults in Mali over a 6-month malaria season. In Part A, safety was assessed at three escalating dose levels. In Part B, participants were randomly assigned (in a 1:1:1 ratio) to receive 10 mg of CIS43LS per kilogram of body weight, 40 mg of CIS43LS per kilogram, or placebo. The primary efficacy end point, assessed in a time-to-event analysis, was the first P. falciparum infection detected on blood-smear examination, which was performed at least every 2 weeks for 24 weeks. At enrollment, all the participants received artemether-lumefantrine to clear possible P. falciparum infection. RESULTS: In Part B, 330 adults underwent randomization; 110 were assigned to each trial group. The risk of moderate headache was 3.3 times as high with 40 mg of CIS43LS per kilogram as with placebo. P. falciparum infections were detected on blood-smear examination in 39 participants (35.5%) who received 10 mg of CIS43LS per kilogram, 20 (18.2%) who received 40 mg of CIS43LS per kilogram, and 86 (78.2%) who received placebo. At 6 months, the efficacy of 40 mg of CIS43LS per kilogram as compared with placebo was 88.2% (adjusted 95% confidence interval [CI], 79.3 to 93.3; P<0.001), and the efficacy of 10 mg of CIS43LS per kilogram as compared with placebo was 75.0% (adjusted 95% CI, 61.0 to 84.0; P<0.001). CONCLUSIONS: CIS43LS was protective against P. falciparum infection over a 6-month malaria season in Mali without evident safety concerns. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT04329104.).


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antimaláricos , Malaria Falciparum , Adulto , Humanos , Antimaláricos/efectos adversos , Antimaláricos/uso terapéutico , Combinación Arteméter y Lumefantrina/uso terapéutico , Malaria Falciparum/diagnóstico , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/prevención & control , Malí , Plasmodium falciparum , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Cefalea/inducido químicamente
8.
PLoS Pathog ; 19(11): e1011585, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37939134

RESUMEN

Natural killer (NK) cells lyse virus-infected cells and transformed cells through polarized delivery of lytic effector molecules into target cells. We have shown that NK cells lyse Plasmodium falciparum-infected red blood cells (iRBC) via antibody-dependent cellular cytotoxicity (ADCC). A high frequency of adaptive NK cells, with elevated intrinsic ADCC activity, in people chronically exposed to malaria transmission is associated with reduced parasitemia and resistance to disease. How NK cells bind to iRBC and the outcome of iRBC lysis by NK cells has not been investigated. We applied gene ablation in inducible erythrocyte precursors and antibody-blocking experiments with iRBC to demonstrate a central role of CD58 and ICAM-4 as ligands for adhesion by NK cells via CD2 and integrin αMß2, respectively. Adhesion was dependent on opsonization of iRBC by IgG. Live imaging and quantitative flow cytometry of NK-mediated ADCC toward iRBC revealed that damage to the iRBC plasma membrane preceded damage to P. falciparum within parasitophorous vacuoles (PV). PV were identified and tracked with a P.falciparum strain that expresses the PV membrane-associated protein EXP2 tagged with GFP. After NK-mediated ADCC, PV were either found inside iRBC ghosts or released intact and devoid of RBC plasma membrane. Electron microscopy images of ADCC cultures revealed tight NK-iRBC synapses and free vesicles similar in size to GFP+ PV isolated from iRBC lysates by cell sorting. The titer of IgG in plasma of malaria-exposed individuals that bound PV was two orders of magnitude higher than IgG that bound iRBC. This immune IgG stimulated efficient phagocytosis of PV by primary monocytes. The selective NK-mediated damage to iRBC, resulting in release of PV, and subsequent phagocytosis of PV by monocytes may combine for efficient killing and removal of intra-erythrocytic P.falciparum parasite. This mechanism may mitigate the inflammation and malaria symptoms during blood-stage P. falciparum infection.


Asunto(s)
Malaria Falciparum , Malaria , Humanos , Monocitos , Ligandos , Vacuolas , Malaria Falciparum/parasitología , Eritrocitos/parasitología , Células Asesinas Naturales , Plasmodium falciparum , Malaria/metabolismo , Fagocitosis , Inmunoglobulina G/metabolismo
9.
Immunity ; 45(2): 231-3, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27533008

RESUMEN

How early interactions between innate and adaptive immune cells influence outcomes of acute infections is incompletely understood. In this issue of Immunity, Karunarathne et al. (2016) show that dendritic cells help CD4(+) T helper 1 cell immunity against malaria through PD-L2's competition with PD-L1.


Asunto(s)
Antígeno B7-H1/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1 , Células Dendríticas/inmunología , Humanos , Activación de Linfocitos/inmunología , Malaria
10.
Immunity ; 45(2): 402-14, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27473412

RESUMEN

Humoral immunity consists of pre-existing antibodies expressed by long-lived plasma cells and rapidly reactive memory B cells (MBC). Recent studies of MBC development and function after protein immunization have uncovered significant MBC heterogeneity. To clarify functional roles for distinct MBC subsets during malaria infection, we generated tetramers that identify Plasmodium-specific MBCs in both humans and mice. Long-lived murine Plasmodium-specific MBCs consisted of three populations: somatically hypermutated immunoglobulin M(+) (IgM(+)) and IgG(+) MBC subsets and an unmutated IgD(+) MBC population. Rechallenge experiments revealed that high affinity, somatically hypermutated Plasmodium-specific IgM(+) MBCs proliferated and gave rise to antibody-secreting cells that dominated the early secondary response to parasite rechallenge. IgM(+) MBCs also gave rise to T cell-dependent IgM(+) and IgG(+)B220(+)CD138(+) plasmablasts or T cell-independent B220(-)CD138(+) IgM(+) plasma cells. Thus, even in competition with IgG(+) MBCs, IgM(+) MBCs are rapid, plastic, early responders to a secondary Plasmodium rechallenge and should be targeted by vaccine strategies.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Plasmodium/inmunología , Animales , Afinidad de Anticuerpos , Antígenos de Protozoos/inmunología , Células Cultivadas , Epítopos , Femenino , Humanos , Inmunidad Humoral , Inmunización Secundaria , Inmunoglobulina M/metabolismo , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hipermutación Somática de Inmunoglobulina , Linfocitos T/inmunología
11.
Metabolomics ; 20(4): 67, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38940866

RESUMEN

INTRODUCTION: Burkitt lymphoma (BL) is an aggressive non-Hodgkin lymphoma associated with Plasmodium falciparum and Epstein-Barr virus, both of which affect metabolic pathways. The metabolomic patterns of BL is unknown. MATERIALS AND METHODS: We measured 627 metabolites in pre-chemotherapy treatment plasma samples from 25 male children (6-11 years) with BL and 25 cancer-free area- and age-frequency-matched male controls from the Epidemiology of Burkitt Lymphoma in East African Children and Minors study in Uganda using liquid chromatography-tandem mass spectrometry. Unconditional, age-adjusted logistic regression analysis was used to estimate odds ratios (ORs) and their 95% confidence intervals (CIs) for the BL association with 1-standard deviation increase in the log-metabolite concentration, adjusting for multiple comparisons using false discovery rate (FDR) thresholds and Bonferroni correction. RESULTS: Compared to controls, levels for 42 metabolite concentrations differed in BL cases (FDR < 0.001), including triacylglyceride (18:0_38:6), alpha-aminobutyric acid (AABA), ceramide (d18:1/20:0), phosphatidylcholine ae C40:6 and phosphatidylcholine C38:6 as the top signals associated with BL (ORs = 6.9 to 14.7, P < 2.4✕10- 4). Two metabolites (triacylglyceride (18:0_38:6) and AABA) selected using stepwise logistic regression discriminated BL cases from controls with an area under the curve of 0.97 (95% CI: 0.94, 1.00). CONCLUSION: Our findings warrant further examination of plasma metabolites as potential biomarkers for BL risk/diagnosis.


Asunto(s)
Linfoma de Burkitt , Metabolómica , Humanos , Linfoma de Burkitt/sangre , Linfoma de Burkitt/metabolismo , Niño , Uganda/epidemiología , Masculino , Estudios de Casos y Controles , Metabolómica/métodos , Metaboloma , Femenino
12.
Immunol Rev ; 293(1): 144-162, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31680289

RESUMEN

The complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against malaria. Studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines. Systems immunology can complement conventional approaches to facilitate our understanding of the complex immune response to the highly dynamic malaria parasite. In this review, recent studies that used systems-based approaches to evaluate human immune responses during natural and experimental Plasmodium falciparum and Plasmodium vivax infections as well as during immunization with candidate malaria vaccines are summarized and related to each other. The potential for next-generation technologies to address the current limitations of systems-based studies of human malaria are discussed.


Asunto(s)
Interacciones Huésped-Parásitos/inmunología , Malaria/inmunología , Malaria/parasitología , Plasmodium/inmunología , Biología de Sistemas , Biomarcadores , Perfilación de la Expresión Génica , Genómica/métodos , Interacciones Huésped-Parásitos/genética , Humanos , Inmunidad , Malaria/genética , Malaria/metabolismo , Vacunas contra la Malaria/inmunología , Biología de Sistemas/métodos
13.
J Infect Dis ; 228(2): 202-211, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-36961831

RESUMEN

BACKGROUND: TP53 has been shown to play a role in inflammatory processes, including malaria. We previously found that p53 attenuates parasite-induced inflammation and predicts clinical protection to Plasmodium falciparum infection in Malian children. Here, we investigated whether p53 codon 47 and 72 polymorphisms are associated with differential risk of P. falciparum infection and uncomplicated malaria in a prospective cohort study of malaria immunity. METHODS: p53 codon 47 and 72 polymorphisms were determined by sequencing TP53 exon 4 in 631 Malian children and adults enrolled in the Kalifabougou cohort study. The effects of these polymorphisms on the prospective risk of febrile malaria, incident parasitemia, and time to fever after incident parasitemia over 6 months of intense malaria transmission were assessed using Cox proportional hazards models. RESULTS: Confounders of malaria risk, including age and hemoglobin S or C, were similar between individuals with or without p53 S47 and R72 polymorphisms. Relative to their respective common variants, neither S47 nor R72 was associated with differences in prospective risk of febrile malaria, incident parasitemia, or febrile malaria after parasitemia. CONCLUSIONS: These findings indicate that p53 codon 47 and 72 polymorphisms are not associated with protection against incident P. falciparum parasitemia or uncomplicated febrile malaria.


Asunto(s)
Malaria Falciparum , Malaria , Niño , Adulto , Humanos , Estudios de Cohortes , Estudios Prospectivos , Parasitemia/genética , Proteína p53 Supresora de Tumor/genética , Plasmodium falciparum/genética , Malaria/complicaciones , Malaria Falciparum/epidemiología , Malaria Falciparum/genética , Malaria Falciparum/complicaciones , Fiebre/etiología
14.
J Infect Dis ; 228(6): 759-768, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37150885

RESUMEN

BACKGROUND: Studies have demonstrated the protective role of antibodies against malaria. Young children are known to be particularly vulnerable to malaria, pointing to the evolution of naturally acquired clinical immunity over time. However, whether changes in antibody functionality track with the acquisition of naturally acquired malaria immunity remains incompletely understood. METHODS: Using systems serology, we characterized sporozoite- and merozoite-specific antibody profiles of uninfected Malian children before the malaria season who differed in their ability to control parasitemia and fever following Plasmodium falciparum (Pf) infection. We then assessed the contributions of individual traits to overall clinical outcomes, focusing on the immunodominant sporozoite CSP and merozoite AMA1 and MSP1 antigens. RESULTS: Humoral immunity evolved with age, with an expansion of both magnitude and functional quality, particularly within blood-stage phagocytic antibody activity. Moreover, concerning clinical outcomes postinfection, protected children had higher antibody-dependent neutrophil activity along with higher levels of MSP1-specific IgG3 and IgA and CSP-specific IgG3 and IgG4 prior to the malaria season. CONCLUSIONS: These data point to the natural evolution of functional humoral immunity to Pf with age and highlight particular antibody Fc-effector profiles associated with the control of malaria in children, providing clues for the design of next-generation vaccines or therapeutics.


Asunto(s)
Malaria Falciparum , Malaria , Animales , Humanos , Niño , Preescolar , Plasmodium falciparum , Proteína 1 de Superficie de Merozoito , Neutrófilos , Antígenos de Protozoos , Anticuerpos Antiprotozoarios , Inmunidad Adaptativa , Merozoítos , Inmunoglobulina G , Autoanticuerpos
15.
PLoS Pathog ; 17(4): e1009430, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33822828

RESUMEN

In malaria-naïve children and adults, Plasmodium falciparum-infected red blood cells (Pf-iRBCs) trigger fever and other symptoms of systemic inflammation. However, in endemic areas where individuals experience repeated Pf infections over many years, the risk of Pf-iRBC-triggered inflammatory symptoms decreases with cumulative Pf exposure. The molecular mechanisms underlying these clinical observations remain unclear. Age-stratified analyses of uninfected, asymptomatic Malian individuals before the malaria season revealed that monocytes of adults produced lower levels of inflammatory cytokines (IL-1ß, IL-6 and TNF) in response to Pf-iRBC stimulation compared to monocytes of Malian children and malaria-naïve U.S. adults. Moreover, monocytes of Malian children produced lower levels of IL-1ß and IL-6 following Pf-iRBC stimulation compared to 4-6-month-old infants. Accordingly, monocytes of Malian adults produced more IL-10 and expressed higher levels of the regulatory molecules CD163, CD206, Arginase-1 and TGM2. These observations were recapitulated in an in vitro system of monocyte to macrophage differentiation wherein macrophages re-exposed to Pf-iRBCs exhibited attenuated inflammatory cytokine responses and a corresponding decrease in the epigenetic marker of active gene transcription, H3K4me3, at inflammatory cytokine gene loci. Together these data indicate that Pf induces epigenetic reprogramming of monocytes/macrophages toward a regulatory phenotype that attenuates inflammatory responses during subsequent Pf exposure. Trial Registration: ClinicalTrials.gov NCT01322581.


Asunto(s)
Malaria Falciparum/inmunología , Malaria/inmunología , Monocitos/metabolismo , Fenotipo , Adulto , Niño , Preescolar , Citocinas/metabolismo , Eritrocitos/metabolismo , Humanos , Lactante , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/metabolismo , Malaria/sangre , Malaria Falciparum/sangre , Monocitos/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/metabolismo
16.
Nat Immunol ; 13(2): 188-95, 2011 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22157630

RESUMEN

Infection of erythrocytes with Plasmodium species induces clinical malaria. Parasite-specific CD4(+) T cells correlate with lower parasite burdens and severity of human malaria and are needed to control blood-stage infection in mice. However, the characteristics of CD4(+) T cells that determine protection or parasite persistence remain unknown. Here we show that infection of humans with Plasmodium falciparum resulted in higher expression of the inhibitory receptor PD-1 associated with T cell dysfunction. In vivo blockade of the PD-1 ligand PD-L1 and the inhibitory receptor LAG-3 restored CD4(+) T cell function, amplified the number of follicular helper T cells and germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. Thus, chronic malaria drives specific T cell dysfunction, and proper function can be restored by inhibitory therapies to enhance parasite control.


Asunto(s)
Antígenos CD/efectos de los fármacos , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD4-Positivos/efectos de los fármacos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Enfermedad Aguda , Animales , Antígenos CD/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/parasitología , Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Niño , Preescolar , Enfermedad Crónica , Eritrocitos/inmunología , Eritrocitos/parasitología , Femenino , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Centro Germinal/parasitología , Humanos , Malaria Falciparum/inmunología , Malí , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Estados Unidos , Regulación hacia Arriba/efectos de los fármacos , Proteína del Gen 3 de Activación de Linfocitos
17.
Malar J ; 22(1): 42, 2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36737743

RESUMEN

BACKGROUND: In malaria endemic regions, transmission of Plasmodium falciparum parasites is often seasonal with very low transmission during the dry season and high transmission in the wet season. Parasites survive the dry season within some individuals who experience prolonged carriage of parasites and are thought to 'seed' infection in the next transmission season. METHODS: Dry season carriers and their role in the subsequent transmission season are characterized using a combination of mathematical simulations and data analysis of previously described data from a longitudinal study in Mali of individuals aged 3 months-12 years (n = 579). RESULTS: Simulating the life-history of individuals experiencing repeated exposure to infection predicts that dry season carriage is more likely in the oldest, most exposed and most immune individuals. This hypothesis is supported by the data from Mali, which shows that carriers are significantly older, experience a higher biting rate at the beginning of the transmission season and develop clinical malaria later than non-carriers. Further, since the most exposed individuals in a community are most likely to be dry season carriers, this is predicted to enable a more than twofold faster spread of parasites into the mosquito population at the start of the subsequent wet season. CONCLUSIONS: Carriage of malaria parasites over the months-long dry season in Mali is most likely in the older, more exposed and more immune children. These children may act as super-spreaders facilitating the fast spread of parasites at the beginning of the next transmission season.


Asunto(s)
Malaria Falciparum , Malaria , Parásitos , Niño , Animales , Humanos , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Estaciones del Año , Estudios Longitudinales , Plasmodium falciparum , Malaria/epidemiología
18.
Nature ; 548(7669): 597-601, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847005

RESUMEN

In two previously described donors, the extracellular domain of LAIR1, a collagen-binding inhibitory receptor encoded on chromosome 19 (ref. 1), was inserted between the V and DJ segments of an antibody. This insertion generated, through somatic mutations, broadly reactive antibodies against RIFINs, a type of variant antigen expressed on the surface of Plasmodium falciparum-infected erythrocytes. To investigate how frequently such antibodies are produced in response to malaria infection, we screened plasma from two large cohorts of individuals living in malaria-endemic regions. Here we report that 5-10% of malaria-exposed individuals, but none of the European blood donors tested, have high levels of LAIR1-containing antibodies that dominate the response to infected erythrocytes without conferring enhanced protection against febrile malaria. By analysing the antibody-producing B cell clones at the protein, cDNA and gDNA levels, we characterized additional LAIR1 insertions between the V and DJ segments and discovered a second insertion modality whereby the LAIR1 exon encoding the extracellular domain and flanking intronic sequences are inserted into the switch region. By exon shuffling, this mechanism leads to the production of bispecific antibodies in which the LAIR1 domain is precisely positioned at the elbow between the VH and CH1 domains. Additionally, in one donor the genomic DNA encoding the VH and CH1 domains was deleted, leading to the production of a camel-like LAIR1-containing antibody. Sequencing of the switch regions of memory B cells from European blood donors revealed frequent templated inserts originating from transcribed genes that, in rare cases, comprised exons with orientations and frames compatible with expression. These results reveal different modalities of LAIR1 insertion that lead to public and dominant antibodies against infected erythrocytes and suggest that insertion of templated DNA represents an additional mechanism of antibody diversification that can be selected in the immune response against pathogens and exploited for B cell engineering.


Asunto(s)
Anticuerpos Antiprotozoarios/química , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Donantes de Sangre , Malaria/inmunología , Mutagénesis Insercional , Plasmodium falciparum/inmunología , Receptores Inmunológicos/genética , Anticuerpos Antiprotozoarios/genética , Antígenos de Protozoos/metabolismo , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Europa (Continente) , Femenino , Genes de las Cadenas Pesadas de las Inmunoglobulinas/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Memoria Inmunológica , Intrones/genética , Malaria/epidemiología , Malaria/parasitología , Masculino , Plasmodium falciparum/metabolismo , Dominios Proteicos , Receptores Inmunológicos/química , Receptores Inmunológicos/inmunología , Moldes Genéticos , Exones VDJ/genética
19.
Mol Cell Proteomics ; 19(1): 101-113, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31658979

RESUMEN

A large body of evidence supports the role of antibodies directed against the Plasmodium spp. parasite in the development of naturally acquired immunity to malaria, however an antigen signature capable of predicting protective immunity against Plasmodium remains to be identified. Key challenges for the identification of a predictive immune signature include the high dimensionality of data produced by high-throughput technologies and the limitation of standard statistical tests in accounting for synergetic interactions between immune responses to multiple targets. In this study, using samples collected from young children in Ghana at multiple time points during a longitudinal study, we adapted a predictive modeling framework which combines feature selection and machine learning techniques to identify an antigen signature of clinical immunity to malaria. Our results show that an individual's immune status can be accurately predicted by measuring antibody responses to a small defined set of 15 target antigens. We further demonstrate that the identified immune signature is highly versatile and capable of providing precise and accurate estimates of clinical protection from malaria in an independent geographic community. Our findings pave the way for the development of a robust point-of-care test to identify individuals at high risk of disease and which could be applied to monitor the impact of vaccinations and other interventions. This approach could be also translated to biomarker discovery for other infectious diseases.


Asunto(s)
Antígenos de Protozoos/inmunología , Enfermedades Endémicas , Inmunidad Innata , Malaria Falciparum/epidemiología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Biomarcadores , Preescolar , Femenino , Estudios de Seguimiento , Predicción , Ghana/epidemiología , Estado de Salud , Humanos , Inmunoglobulina G/inmunología , Lactante , Estudios Longitudinales , Aprendizaje Automático , Malaria Falciparum/diagnóstico , Malaria Falciparum/parasitología , Masculino , Pronóstico
20.
Malar J ; 20(1): 9, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407502

RESUMEN

BACKGROUND: Plasmodium falciparum causes the majority of malaria cases worldwide and children in sub-Saharan Africa are the most vulnerable group affected. Non-sterile clinical immunity that protects from symptoms develops slowly and is relatively short-lived. Moreover, current malaria vaccine candidates fail to induce durable high-level protection in endemic settings, possibly due to the immunomodulatory effects of the malaria parasite itself. Because dendritic cells play a crucial role in initiating immune responses, the aim of this study was to better understand the impact of cumulative malaria exposure as well as concurrent P. falciparum infection on dendritic cell phenotype and function. METHODS: In this cross-sectional study, the phenotype and function of dendritic cells freshly isolated from peripheral blood samples of Malian adults with a lifelong history of malaria exposure who were either uninfected (n = 27) or asymptomatically infected with P. falciparum (n = 8) was assessed. Additionally, plasma cytokine and chemokine levels were measured in these adults and in Malian children (n = 19) with acute symptomatic malaria. RESULTS: With the exception of lower plasmacytoid dendritic cell frequencies in asymptomatically infected Malian adults, peripheral blood dendritic cell subset frequencies and HLA-DR surface expression did not differ by infection status. Peripheral blood myeloid dendritic cells of uninfected Malian adults responded to in vitro stimulation with P. falciparum blood-stage parasites by up-regulating the costimulatory molecules HLA-DR, CD80, CD86 and CD40 and secreting IL-10, CXCL9 and CXCL10. In contrast, myeloid dendritic cells of asymptomatically infected Malian adults exhibited no significant responses above the uninfected red blood cell control. IL-10 and CXCL9 plasma levels were elevated in both asymptomatic adults and children with acute malaria. CONCLUSIONS: The findings of this study indicate that myeloid dendritic cells of uninfected adults with a lifelong history of malaria exposure are able to up-regulate co-stimulatory molecules and produce cytokines. Whether mDCs of malaria-exposed individuals are efficient antigen-presenting cells capable of mounting an appropriate immune response remains to be determined. The data also highlights IL-10 and CXCL9 as important factors in both asymptomatic and acute malaria and add to the understanding of asymptomatic P. falciparum infections in malaria-endemic areas.


Asunto(s)
Citocinas/sangre , Células Dendríticas/parasitología , Malaria Falciparum/sangre , Adulto , Infecciones Asintomáticas , Quimiocinas/sangre , Niño , Preescolar , Estudios Transversales , Eritrocitos/parasitología , Femenino , Humanos , Malaria/sangre , Masculino , Malí , Persona de Mediana Edad , Fenotipo , Plasmodium falciparum/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA