Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Res Sq ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38645126

RESUMEN

Malaria is a major public health problem, but many of the factors underlying the pathogenesis of this disease are not well understood. Here, we demonstrate in Malian children that susceptibility to febrile malaria following infection with Plasmodium falciparum is associated with the composition of the gut microbiome prior to the malaria season. Gnotobiotic mice colonized with the fecal samples of malaria-susceptible children had a significantly higher parasite burden following Plasmodium infection compared to gnotobiotic mice colonized with the fecal samples of malaria-resistant children. The fecal microbiome of the susceptible children was enriched for bacteria associated with inflammation, mucin degradation, gut permeability and inflammatory bowel disorders (e.g., Ruminococcus gauvreauii, Ruminococcus torques, Dorea formicigenerans, Dorea longicatena, Lachnoclostridium phocaeense and Lachnoclostridium sp. YL32). However, the susceptible children also had a greater abundance of bacteria known to produce anti-inflammatory short-chain fatty acids and those associated with favorable prognosis and remission following dysbiotic intestinal events (e.g., Anaerobutyricum hallii, Blautia producta and Sellimonas intestinalis). Metabolomics analysis of the human fecal samples corroborated the existence of inflammatory and recovery-associated features within the gut microbiome of the susceptible children. There was an enrichment of nitric oxide-derived DNA adducts (deoxyinosine and deoxyuridine) and long-chain fatty acids, the absorption of which has been shown to be inhibited by inflamed intestinal epithelial cells, and a decrease in the abundance of mucus phospholipids. Nevertheless, there were also increased levels of pseudouridine and hypoxanthine, which have been shown to be regulated in response to cellular stress and to promote recovery following injury or hypoxia. Overall, these results indicate that the gut microbiome may contribute malaria pathogenesis and suggest that therapies targeting intestinal inflammation could decrease malaria susceptibility.

2.
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
3.
Microbiol Spectr ; 11(6): e0155423, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37819130

RESUMEN

IMPORTANCE: There is increasing evidence that microbes residing within the intestines (gut microbiota) play important roles in the well-being of humans. Yet, there are considerable challenges in determining the specific role of gut microbiota in human diseases owing to the complexity of diverse internal and environmental factors that can contribute to diseases. Mice devoid of all microorganisms (germ-free mice) can be colonized with human stool samples to examine the specific contribution of the gut microbiota to a disease. These approaches have been primarily focused on stool samples obtained from individuals in Western countries. Thus, there is limited understanding as to whether the same methods used to colonize germ-free mice with stool from Western individuals would apply to the colonization of germ-free mice with stool from non-Western individuals. Here, we report the results from colonizing germ-free mice with stool samples of Malian children.


Asunto(s)
Microbioma Gastrointestinal , Intestinos , Niño , Humanos , Animales , Ratones , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Heces
4.
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
5.
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
6.
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
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
9.
Sci Immunol ; 7(71): eabn1250, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35559666

RESUMEN

Several infectious and autoimmune diseases are associated with an expansion of CD21-CD27- atypical B cells (atBCs) that up-regulate inhibitory receptors and exhibit altered B cell receptor (BCR) signaling. The function of atBCs remains unclear, and few studies have investigated the biology of pathogen-specific atBCs during acute infection. Here, we performed longitudinal flow cytometry analyses and RNA sequencing of Plasmodium falciparum (Pf)-specific B cells isolated from study participants before and shortly after febrile malaria, with simultaneous analysis of influenza hemagglutinin (HA)-specific B cells as a comparator. At the healthy baseline before the malaria season, individuals had similar frequencies of Pf- and HA-specific atBCs that did not differ proportionally from atBCs within the total B cell population. BCR sequencing identified clonal relationships between Pf-specific atBCs, activated B cells (actBCs), and classical memory B cells (MBCs) and revealed comparable degrees of somatic hypermutation. At the healthy baseline, Pf-specific atBCs were transcriptionally distinct from Pf-specific actBCs and classical MBCs. In response to acute febrile malaria, Pf-specific atBCs and actBCs up-regulated similar intracellular signaling cascades. Pf-specific atBCs showed activation of pathways involved in differentiation into antibody-secreting cells and up-regulation of molecules that mediate B-T cell interactions, suggesting that atBCs respond to T follicular helper (TFH) cells. In the presence of TFH cells and staphylococcal enterotoxin B, atBCs of malaria-exposed individuals differentiated into CD38+ antibody-secreting cells in vitro, suggesting that atBCs may actively contribute to humoral immunity to infectious pathogens.


Asunto(s)
Gripe Humana , Malaria , Humanos , Inmunoglobulina M , Memoria Inmunológica , Plasmodium falciparum , Células T Auxiliares Foliculares
10.
Nat Commun ; 12(1): 4711, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330920

RESUMEN

Following Plasmodium falciparum infection, individuals can remain asymptomatic, present with mild fever in uncomplicated malaria cases, or show one or more severe malaria symptoms. Several studies have investigated associations between parasite transcription and clinical severity, but no broad conclusions have yet been drawn. Here, we apply a series of bioinformatic approaches based on P. falciparum's tightly regulated transcriptional pattern during its ~48-hour intraerythrocytic developmental cycle (IDC) to publicly available transcriptomes of parasites obtained from malaria cases of differing clinical severity across multiple studies. Our analysis shows that within each IDC, the circulation time of infected erythrocytes without sequestering to endothelial cells decreases with increasing parasitaemia or disease severity. Accordingly, we find that the size of circulating infected erythrocytes is inversely related to parasite density and disease severity. We propose that enhanced adhesiveness of infected erythrocytes leads to a rapid increase in parasite burden, promoting higher parasitaemia and increased disease severity.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación Bacteriana de la Expresión Génica , Malaria Falciparum/sangre , Parasitemia/sangre , Plasmodium falciparum/genética , Tiempo de Circulación Sanguínea , Eritrocitos/parasitología , Ontología de Genes , Genes Bacterianos/genética , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/fisiopatología , Parasitemia/parasitología , Parasitemia/fisiopatología , Plasmodium falciparum/fisiología
11.
Sci Transl Med ; 13(599)2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162751

RESUMEN

Immunoglobulin (Ig)A antibodies play a critical role in protection against mucosal pathogens. However, the role of serum IgA in immunity to nonmucosal pathogens, such as Plasmodium falciparum, is poorly characterized, despite being the second most abundant isotype in blood after IgG. Here, we investigated the circulating IgA response in humans to P. falciparum sporozoites that are injected into the skin by mosquitoes and migrate to the liver via the bloodstream to initiate malaria infection. We found that circulating IgA was induced in three independent sporozoite-exposed cohorts: individuals living in an endemic region in Mali, malaria-naïve individuals immunized intravenously with three large doses of irradiated sporozoites, and malaria-naïve individuals exposed to a single controlled mosquito bite infection. Mechanistically, we found evidence in an animal model that IgA responses were induced by sporozoites at dermal inoculation sites. From malaria-resistant individuals, we isolated several IgA monoclonal antibodies that reduced liver parasite burden in mice. One antibody, MAD2-6, bound to a conserved epitope in the amino terminus of the P. falciparum circumsporozoite protein, the dominant protein on the sporozoite surface. Crystal structures of this antibody revealed a unique mode of binding whereby two Fabs simultaneously bound either side of the target peptide. This study reveals a role for circulating IgA in malaria and identifies the amino terminus of the circumsporozoite protein as a target of functional antibodies.


Asunto(s)
Anticuerpos Antiprotozoarios , Inmunoglobulina A , Malaria , Animales , Anticuerpos Antiprotozoarios/inmunología , Humanos , Inmunoglobulina A/inmunología , Malaria/inmunología , Ratones , Plasmodium falciparum , Proteínas Protozoarias , Esporozoítos
12.
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
13.
Clin Transl Immunology ; 10(3): e1265, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763229

RESUMEN

OBJECTIVES: Study of individuals with protection from Plasmodium falciparum (Pf) infection and clinical malaria, including individuals affected by the sickle-cell trait (HbAS), offers the potential to identify cellular targets that could be translated for therapeutic development. We previously reported the first involvement of cellular immunity in HbAS-associated relative protection and identified a novel subset of memory-activated NK cells that was enriched in HbAS children and associated with parasite control. We hypothesised that other memory cell subsets might distinguish the baseline profile of HbAS children and children with normal haemoglobin (HbAA). METHODS: Subsets of memory T cells and NK cells were analysed by flow cytometry in paired samples collected from HbAS and HbAA children, at baseline and during the first malaria episode of the ensuing transmission season. Correlations between cell frequencies and features of HbAS-mediated protection from malaria were determined. RESULTS: HbAS children displayed significantly higher frequency of memory CD8+ T cells at baseline than HbAA children. Baseline frequency of memory CD8+ T cells correlated with features of HbAS-mediated protection from malaria. Exploration of memory CD8+ T cell subsets revealed that central memory CD8+ T cell frequency was higher in HbAS children than in HbAA children. CONCLUSION: This study shows that HbAS children develop a larger memory CD8+ T cell compartment than HbAA children, and associates this compartment with better control of subsequent onset of infection and parasite density. Our data suggest that central memory CD8+ T cells may play an important role in the relative protection against malaria experienced by HbAS individuals, and further work to investigate this is warranted.

14.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33661303

RESUMEN

IgG antibodies play a role in malaria immunity, but whether and how IgM protects from malaria and the biology of Plasmodium falciparum (Pf)-specific IgM B cells is unclear. In a Mali cohort spanning infants to adults, we conducted longitudinal analyses of Pf- and influenza-specific B cells. We found that Pf-specific memory B cells (MBCs) are disproportionally IgM+ and only gradually shift to IgG+ with age, in contrast to influenza-specific MBCs that are predominantly IgG+ from infancy to adulthood. B cell receptor analysis showed Pf-specific IgM MBCs are somatically hypermutated at levels comparable to influenza-specific IgG B cells. During acute malaria, Pf-specific IgM B cells expand and upregulate activation/costimulatory markers. Finally, plasma IgM was comparable to IgG in inhibiting Pf growth and enhancing phagocytosis of Pf by monocytes in vitro. Thus, somatically hypermutated Pf-specific IgM MBCs dominate in children, expand and activate during malaria, and produce IgM that inhibits Pf through neutralization and opsonic phagocytosis.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Linfocitos B/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Malaria Falciparum/inmunología , Malaria/inmunología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Memoria Inmunológica , Lactante , Recién Nacido , Estudios Longitudinales , Malaria/sangre , Malaria/epidemiología , Malaria/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Masculino , Malí/epidemiología , Fagocitosis/inmunología , Adulto Joven
15.
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
16.
Nat Med ; 26(12): 1929-1940, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33106664

RESUMEN

The dry season is a major challenge for Plasmodium falciparum parasites in many malaria endemic regions, where water availability limits mosquito vectors to only part of the year. How P. falciparum bridges two transmission seasons months apart, without being cleared by the human host or compromising host survival, is poorly understood. Here we show that low levels of P. falciparum parasites persist in the blood of asymptomatic Malian individuals during the 5- to 6-month dry season, rarely causing symptoms and minimally affecting the host immune response. Parasites isolated during the dry season are transcriptionally distinct from those of individuals with febrile malaria in the transmission season, coinciding with longer circulation within each replicative cycle of parasitized erythrocytes without adhering to the vascular endothelium. Low parasite levels during the dry season are not due to impaired replication but rather to increased splenic clearance of longer-circulating infected erythrocytes, which likely maintain parasitemias below clinical and immunological radar. We propose that P. falciparum virulence in areas of seasonal malaria transmission is regulated so that the parasite decreases its endothelial binding capacity, allowing increased splenic clearance and enabling several months of subclinical parasite persistence.


Asunto(s)
Infecciones Asintomáticas/epidemiología , Interacciones Huésped-Parásitos/genética , Malaria Falciparum/epidemiología , Plasmodium falciparum/patogenicidad , Adolescente , Adulto , Animales , Niño , Preescolar , Enfermedades Endémicas/prevención & control , Eritrocitos/parasitología , Femenino , Genotipo , Humanos , Lactante , Malaria Falciparum/genética , Malaria Falciparum/parasitología , Masculino , Malí/epidemiología , Persona de Mediana Edad , Plasmodium falciparum/genética , Estaciones del Año , Adulto Joven
17.
bioRxiv ; 2020 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-33106806

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 monocytes collected from uninfected, asymptomatic Malian individuals before the malaria season revealed an inverse relationship between age and Pf -iRBC-inducible inflammatory cytokine (IL-1ß, IL-6 and TNF) production, whereas Malian infants and malaria-naïve U.S. adults produced similarly high levels of inflammatory cytokines. 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. These findings also suggest that past malaria exposure could mitigate monocyte-associated immunopathology induced by other pathogens such as SARS-CoV-2. AUTHOR SUMMARY: The malaria parasite is mosquito-transmitted and causes fever and other inflammatory symptoms while circulating in the bloodstream. However, in regions of high malaria transmission the parasite is less likely to cause fever as children age and enter adulthood, even though adults commonly have malaria parasites in their blood. Monocytes are cells of the innate immune system that secrete molecules that cause fever and inflammation when encountering microorganisms like malaria. Although inflammation is critical to initiating normal immune responses, too much inflammation can harm infected individuals. In Mali, we conducted a study of a malaria-exposed population from infants to adults and found that participants' monocytes produced less inflammation as age increases, whereas monocytes of Malian infants and U.S. adults, who had never been exposed to malaria, both produced high levels of inflammatory molecules. Accordingly, monocytes exposed to malaria in the laboratory became less inflammatory when re-exposed to malaria again later, and these monocytes 'turned down' their inflammatory genes. This study helps us understand how people become immune to inflammatory symptoms of malaria and may also help explain why people in malaria-endemic areas appear to be less susceptible to the harmful effects of inflammation caused by other pathogens such as SARS-CoV-2.

18.
JCI Insight ; 5(12)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32427581

RESUMEN

BACKGROUNDMalaria pathogenicity is determined, in part, by the adherence of Plasmodium falciparum-infected erythrocytes to the microvasculature mediated via specific interactions between P. falciparum erythrocyte membrane protein (PfEMP1) variant domains and host endothelial receptors. Naturally acquired antibodies against specific PfEMP1 variants can play an important role in clinical protection against malaria.METHODSWe evaluated IgG responses against a repertoire of PfEMP1 CIDR domain variants to determine the rate and order of variant-specific antibody acquisition and their association with protection against febrile malaria in a prospective cohort study conducted in an area of intense, seasonal malaria transmission.RESULTSUsing longitudinal data, we found that IgG antibodies against the pathogenic domain variants CIDRα1.7 and CIDRα1.8 were acquired the earliest. Furthermore, IgG antibodies against CIDRγ3 were associated with reduced prospective risk of febrile malaria and recurrent malaria episodes.CONCLUSIONThis study provides evidence that acquisition of IgG antibodies against PfEMP1 variants is ordered and demonstrates that antibodies against CIDRα1 domains are acquired the earliest in children residing in an area of intense, seasonal malaria transmission. Future studies will need to validate these findings in other transmission settings and determine the functional activity of these naturally acquired CIDR variant-specific antibodies.TRIAL REGISTRATIONClinicalTrials.gov NCT01322581.FUNDINGDivision of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH.


Asunto(s)
Inmunoglobulina G/sangre , Malaria Falciparum/inmunología , Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Anticuerpos Antiprotozoarios/sangre , Eritrocitos/inmunología , Humanos , Estudios Prospectivos
19.
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
20.
Mali Med ; 33(1): 10-15, 2018.
Artículo en Francés | MEDLINE | ID: mdl-30484584

RESUMEN

Soil transmitted helminthiasis and schistosomiasis are neglected tropical diseases (NTD), affecting the health status of endemic Malian populations. Mali has a national NTD elimination program using the mass drug administration (MDA) strategy combining Albendazole, Ivermectinand Praziquantel. Malaria still remains a public health problem in Mali. The Community health Center (CSCOM) in Kalifabougouvillage in the Kati health district has benefited from such MDA program since 2010. AIM: To evaluate the prevalence rate of malaria, intestinal and urinary parasite infections in the local population. MATERIEL AND METHODS: We conducted a nested cross sectional and cohort study in May 2011 on volunteers aged three months old to 25 years old. Blood smear (blood), Kato-Katz (Stools) and urine filtration techniques were used to evaluate parasite prevalence. Informed consent and assentment were obtained from the volunteers before their inclusion. All volunteers received treatment against the parasite diseases of interest according to the guidelines of national disease control programs. RESULTS: A total of 688 volunteers were included. The prevalence rates of parasitic infections were 22.1% [95% CI= 22.06 - 22.12] for Plasmodium falciparum, 9% [95% CI: 8.9-9.034] for Schistosoma haematobium; 3.5% [95% CI: 3.48-3.513] for Hymenolepis nana and 0.1% [95% CI : 0.093-0.107] for Schistoso mamansoni. The prevalence rate of the co-infection Plasmodium falciparum - Schistosoma haematobium was 2.18% [95% CI= 2.17 - 2.19] in Kalifabougou. CONCLUSION: Praziquantel and Albendazole-based MDA and Artemisinin based combined therapy (ACTs) could explain theobserved low prevalence of helminthiasis and malaria in Kalifabougou, Mali.


Les géohelminthes et les schistosomoses sont des maladies tropicales négligées, impactant sur l'état de santé des populations maliennes endémiques. Le Mali dispose d'un programme national d'élimination qui utilise la stratégie du Traitement de Masse à base Communautaire (TMC) combinant l'Albendazole, Ivermectine et le Praziquantel (PZQ). Quant au paludisme, il reste un problème majeur de santé publique au Mali. L'aire de santé du Centre de Santé Communautaire (CSCOM) de Kalifabougou, dans le district sanitaire de Kati, bénéficie de tel programme TMC depuis 2010. OBJECTIF: Evaluer les taux de prévalences du paludisme infestation, des parasitoses intestinales et urinaires dans la population du village de Kalifabougou, Mali. MATERIELS ET MÉTHODES: Nous avons réalisé une étude transversale en mai 2011 nichée dans une cohorte de volontaires âgés de 3 mois à 25 ans. Les techniques de la goutte épaisse (sang), de Kato-Katz, (selles), et de la filtration des urines; ont été utilisées pour l'évaluation des prévalences parasitaires. Le consentement éclairé et libre ainsi l'assentiment ont été obtenus de tous les volontaires avant leur inclusion. Tous les volontaires ont reçu des traitements contre les parasitoses selon les recommandations des programmes nationaux de lutte contre ces maladies. RÉSULTATS: Un total de 688 volontaires a été inclus dans notre étude. Les taux de prévalences des infections parasitaires étaient de 22,1%[95% CI: 22,06­22,12] pour le Plasmodium falciparum, 9%[95% CI: 8,9­9,034] pour Schistosomahaematobium, 3,5%[95% CI:3,48­3,513] pour Hymenolepis nana et 0,1%[95% CI : 0,093­0,107] pour Schistosomamansoni. Le taux de prévalence de la co-infection Plasmodium falciparum - Schistosomahaematobium était de 2,18%[95% CI :2,17­2,19] a Kalifabougou. CONCLUSION: Le traitement de masse à base de Praziquantel (PZQ) et d'Albendazole et le traitement systématique des cas de fièvres par les combinaisons thérapeutiques à base d'artemisinine (CTAs), auraient pu contribuer à la baisse de la prévalence des helminthes et du paludisme à Kalifabougou, Mali.


Asunto(s)
Parasitosis Intestinales/epidemiología , Infecciones Urinarias/epidemiología , Infecciones Urinarias/parasitología , Adolescente , Niño , Preescolar , Estudios de Cohortes , Estudios Transversales , Femenino , Humanos , Lactante , Masculino , Malí/epidemiología , Prevalencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...