Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 242
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 602(7895): 106-111, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34883497

RESUMEN

Host genetic factors can confer resistance against malaria1, raising the question of whether this has led to evolutionary adaptation of parasite populations. Here we searched for association between candidate host and parasite genetic variants in 3,346 Gambian and Kenyan children with severe malaria caused by Plasmodium falciparum. We identified a strong association between sickle haemoglobin (HbS) in the host and three regions of the parasite genome, which is not explained by population structure or other covariates, and which is replicated in additional samples. The HbS-associated alleles include nonsynonymous variants in the gene for the acyl-CoA synthetase family member2-4 PfACS8 on chromosome 2, in a second region of chromosome 2, and in a region containing structural variation on chromosome 11. The alleles are in strong linkage disequilibrium and have frequencies that covary with the frequency of HbS across populations, in particular being much more common in Africa than other parts of the world. The estimated protective effect of HbS against severe malaria, as determined by comparison of cases with population controls, varies greatly according to the parasite genotype at these three loci. These findings open up a new avenue of enquiry into the biological and epidemiological significance of the HbS-associated polymorphisms in the parasite genome and the evolutionary forces that have led to their high frequency and strong linkage disequilibrium in African P. falciparum populations.


Asunto(s)
Genotipo , Hemoglobina Falciforme/genética , Adaptación al Huésped/genética , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Parásitos/genética , Plasmodium falciparum/genética , Alelos , Animales , Niño , Femenino , Gambia/epidemiología , Genes Protozoarios/genética , Humanos , Kenia/epidemiología , Desequilibrio de Ligamiento , Malaria Falciparum/epidemiología , Masculino , Polimorfismo Genético
2.
Nature ; 600(7889): 506-511, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34649268

RESUMEN

The evolution of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus leads to new variants that warrant timely epidemiological characterization. Here we use the dense genomic surveillance data generated by the COVID-19 Genomics UK Consortium to reconstruct the dynamics of 71 different lineages in each of 315 English local authorities between September 2020 and June 2021. This analysis reveals a series of subepidemics that peaked in early autumn 2020, followed by a jump in transmissibility of the B.1.1.7/Alpha lineage. The Alpha variant grew when other lineages declined during the second national lockdown and regionally tiered restrictions between November and December 2020. A third more stringent national lockdown suppressed the Alpha variant and eliminated nearly all other lineages in early 2021. Yet a series of variants (most of which contained the spike E484K mutation) defied these trends and persisted at moderately increasing proportions. However, by accounting for sustained introductions, we found that the transmissibility of these variants is unlikely to have exceeded the transmissibility of the Alpha variant. Finally, B.1.617.2/Delta was repeatedly introduced in England and grew rapidly in early summer 2021, constituting approximately 98% of sampled SARS-CoV-2 genomes on 26 June 2021.


Asunto(s)
COVID-19/epidemiología , COVID-19/virología , Genoma Viral/genética , Genómica , SARS-CoV-2/genética , Sustitución de Aminoácidos , COVID-19/transmisión , Inglaterra/epidemiología , Monitoreo Epidemiológico , Humanos , Epidemiología Molecular , Mutación , Cuarentena/estadística & datos numéricos , SARS-CoV-2/clasificación , Análisis Espacio-Temporal , Glicoproteína de la Espiga del Coronavirus/genética
3.
Nature ; 593(7858): 266-269, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33767447

RESUMEN

The SARS-CoV-2 lineage B.1.1.7, designated variant of concern (VOC) 202012/01 by Public Health England1, was first identified in the UK in late summer to early autumn 20202. Whole-genome SARS-CoV-2 sequence data collected from community-based diagnostic testing for COVID-19 show an extremely rapid expansion of the B.1.1.7 lineage during autumn 2020, suggesting that it has a selective advantage. Here we show that changes in VOC frequency inferred from genetic data correspond closely to changes inferred by S gene target failures (SGTF) in community-based diagnostic PCR testing. Analysis of trends in SGTF and non-SGTF case numbers in local areas across England shows that B.1.1.7 has higher transmissibility than non-VOC lineages, even if it has a different latent period or generation time. The SGTF data indicate a transient shift in the age composition of reported cases, with cases of B.1.1.7 including a larger share of under 20-year-olds than non-VOC cases. We estimated time-varying reproduction numbers for B.1.1.7 and co-circulating lineages using SGTF and genomic data. The best-supported models did not indicate a substantial difference in VOC transmissibility among different age groups, but all analyses agreed that B.1.1.7 has a substantial transmission advantage over other lineages, with a 50% to 100% higher reproduction number.


Asunto(s)
COVID-19/transmisión , COVID-19/virología , Filogenia , SARS-CoV-2/clasificación , SARS-CoV-2/patogenicidad , Adolescente , Adulto , Distribución por Edad , Anciano , Anciano de 80 o más Años , Número Básico de Reproducción , COVID-19/diagnóstico , COVID-19/epidemiología , Niño , Preescolar , Inglaterra/epidemiología , Evolución Molecular , Genoma Viral/genética , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/análisis , Glicoproteína de la Espiga del Coronavirus/genética , Factores de Tiempo , Adulto Joven
4.
Nature ; 585(7826): 579-583, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32939086

RESUMEN

Malaria has had a major effect on the human genome, with many protective polymorphisms-such as the sickle-cell trait-having been selected to high frequencies in malaria-endemic regions1,2. The blood group variant Dantu provides 74% protection against all forms of severe malaria in homozygous individuals3-5, a similar degree of protection to that afforded by the sickle-cell trait and considerably greater than that offered by the best malaria vaccine. Until now, however, the protective mechanism has been unknown. Here we demonstrate the effect of Dantu on the ability of the merozoite form of the malaria parasite Plasmodium falciparum to invade red blood cells (RBCs). We find that Dantu is associated with extensive changes to the repertoire of proteins found on the RBC surface, but, unexpectedly, inhibition of invasion does not correlate with specific RBC-parasite receptor-ligand interactions. By following invasion using video microscopy, we find a strong link between RBC tension and merozoite invasion, and identify a tension threshold above which invasion rarely occurs, even in non-Dantu RBCs. Dantu RBCs have higher average tension than non-Dantu RBCs, meaning that a greater proportion resist invasion. These findings provide both an explanation for the protective effect of Dantu, and fresh insight into why the efficiency of P. falciparum invasion might vary across the heterogenous populations of RBCs found both within and between individuals.


Asunto(s)
Antígenos de Grupos Sanguíneos/genética , Eritrocitos/citología , Eritrocitos/parasitología , Malaria Falciparum/patología , Malaria Falciparum/prevención & control , Plasmodium falciparum/metabolismo , Polimorfismo Genético , Antígenos de Grupos Sanguíneos/clasificación , Antígenos de Grupos Sanguíneos/metabolismo , Niño , Eritrocitos/metabolismo , Eritrocitos/patología , Femenino , Genotipo , Humanos , Kenia , Ligandos , Masculino , Merozoítos/metabolismo , Merozoítos/patogenicidad , Microscopía por Video , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad
5.
PLoS Genet ; 19(9): e1010910, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37708213

RESUMEN

Blood group O is associated with protection against severe malaria and reduced size and stability of P. falciparum-host red blood cell (RBC) rosettes compared to non-O blood groups. Whether the non-O blood groups encoded by the specific ABO genotypes AO, BO, AA, BB and AB differ in their associations with severe malaria and rosetting is unknown. The A and B antigens are host RBC receptors for rosetting, hence we hypothesized that the higher levels of A and/or B antigen on RBCs from AA, BB and AB genotypes compared to AO/BO genotypes could lead to larger rosettes, increased microvascular obstruction and higher risk of malaria pathology. We used a case-control study of Kenyan children and in vitro adhesion assays to test the hypothesis that "double dose" non-O genotypes (AA, BB, AB) are associated with increased risk of severe malaria and larger rosettes than "single dose" heterozygotes (AO, BO). In the case-control study, compared to OO, the double dose genotypes consistently had higher odds ratios (OR) for severe malaria than single dose genotypes, with AB (OR 1.93) and AO (OR 1.27) showing most marked difference (p = 0.02, Wald test). In vitro experiments with blood group A-preferring P. falciparum parasites showed that significantly larger rosettes were formed with AA and AB host RBCs compared to OO, whereas AO and BO genotypes rosettes were indistinguishable from OO. Overall, the data show that ABO genotype influences P. falciparum rosetting and support the hypothesis that double dose non-O genotypes confer a greater risk of severe malaria than AO/BO heterozygosity.


Asunto(s)
Malaria Falciparum , Malaria , Niño , Humanos , Sistema del Grupo Sanguíneo ABO/genética , Plasmodium falciparum/genética , Estudios de Casos y Controles , Kenia , Genotipo , Malaria Falciparum/genética
6.
PLoS Genet ; 17(1): e1009253, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33476334

RESUMEN

Vector population control using insecticides is a key element of current strategies to prevent malaria transmission in Africa. The introduction of effective insecticides, such as the organophosphate pirimiphos-methyl, is essential to overcome the recurrent emergence of resistance driven by the highly diverse Anopheles genomes. Here, we use a population genomic approach to investigate the basis of pirimiphos-methyl resistance in the major malaria vectors Anopheles gambiae and A. coluzzii. A combination of copy number variation and a single non-synonymous substitution in the acetylcholinesterase gene, Ace1, provides the key resistance diagnostic in an A. coluzzii population from Côte d'Ivoire that we used for sequence-based association mapping, with replication in other West African populations. The Ace1 substitution and duplications occur on a unique resistance haplotype that evolved in A. gambiae and introgressed into A. coluzzii, and is now common in West Africa primarily due to selection imposed by other organophosphate or carbamate insecticides. Our findings highlight the predictive value of this complex resistance haplotype for phenotypic resistance and clarify its evolutionary history, providing tools to for molecular surveillance of the current and future effectiveness of pirimiphos-methyl based interventions.


Asunto(s)
Acetilcolinesterasa/genética , Resistencia a los Insecticidas/genética , Malaria/genética , Malaria/transmisión , África Occidental , Animales , Anopheles/efectos de los fármacos , Anopheles/genética , Anopheles/parasitología , Variaciones en el Número de Copia de ADN/genética , Genes Duplicados/genética , Introgresión Genética/genética , Humanos , Insecticidas/efectos adversos , Malaria/parasitología , Malaria/prevención & control , Mosquitos Vectores/genética , Compuestos Organotiofosforados/efectos adversos , Compuestos Organotiofosforados/farmacología
7.
Genome Res ; 30(8): 1154-1169, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32817236

RESUMEN

The characterization of de novo mutations in regions of high sequence and structural diversity from whole-genome sequencing data remains highly challenging. Complex structural variants tend to arise in regions of high repetitiveness and low complexity, challenging both de novo assembly, in which short reads do not capture the long-range context required for resolution, and mapping approaches, in which improper alignment of reads to a reference genome that is highly diverged from that of the sample can lead to false or partial calls. Long-read technologies can potentially solve such problems but are currently unfeasible to use at scale. Here we present Corticall, a graph-based method that combines the advantages of multiple technologies and prior data sources to detect arbitrary classes of genetic variant. We construct multisample, colored de Bruijn graphs from short-read data for all samples, align long-read-derived haplotypes and multiple reference data sources to restore graph connectivity information, and call variants using graph path-finding algorithms and a model for simultaneous alignment and recombination. We validate and evaluate the approach using extensive simulations and use it to characterize the rate and spectrum of de novo mutation events in 119 progeny from four Plasmodium falciparum experimental crosses, using long-read data on the parents to inform reconstructions of the progeny and to detect several known and novel nonallelic homologous recombination events.


Asunto(s)
Genoma de Protozoos/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación/genética , Plasmodium falciparum/genética , Secuenciación Completa del Genoma/métodos , Algoritmos , Secuencia de Bases , Variación Genética/genética , Alineación de Secuencia , Análisis de Secuencia de ADN/métodos , Programas Informáticos
8.
PLoS Comput Biol ; 18(2): e1009801, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35108259

RESUMEN

Investigation of the diversity of malaria parasite antigens can help prioritize and validate them as vaccine candidates and identify the most common variants for inclusion in vaccine formulations. Studies of vaccine candidates of the most virulent human malaria parasite, Plasmodium falciparum, have focused on a handful of well-known antigens, while several others have never been studied. Here we examine the global diversity and population structure of leading vaccine candidate antigens of P. falciparum using the MalariaGEN Pf3K (version 5.1) resource, comprising more than 2600 genomes from 15 malaria endemic countries. A stringent variant calling pipeline was used to extract high quality antigen gene 'haplotypes' from the global dataset and a new R-package named VaxPack was used to streamline population genetic analyses. In addition, a newly developed algorithm that enables spatial averaging of selection pressure on 3D protein structures was applied to the dataset. We analysed the genes encoding 23 leading and novel candidate malaria vaccine antigens including csp, trap, eba175, ama1, rh5, and CelTOS. Our analysis shows that current malaria vaccine formulations are based on rare haplotypes and thus may have limited efficacy against natural parasite populations. High levels of diversity with evidence of balancing selection was detected for most of the erythrocytic and pre-erythrocytic antigens. Measures of natural selection were then mapped to 3D protein structures to predict targets of functional antibodies. For some antigens, geographical variation in the intensity and distribution of these signals on the 3D structure suggests adaptation to different human host or mosquito vector populations. This study provides an essential framework for the diversity of P. falciparum antigens to be considered in the design of the next generation of malaria vaccines.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Animales , Humanos
10.
Nature ; 542(7639): 101-104, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28117441

RESUMEN

Elucidation of the evolutionary history and interrelatedness of Plasmodium species that infect humans has been hampered by a lack of genetic information for three human-infective species: P. malariae and two P. ovale species (P. o. curtisi and P. o. wallikeri). These species are prevalent across most regions in which malaria is endemic and are often undetectable by light microscopy, rendering their study in human populations difficult. The exact evolutionary relationship of these species to the other human-infective species has been contested. Using a new reference genome for P. malariae and a manually curated draft P. o. curtisi genome, we are now able to accurately place these species within the Plasmodium phylogeny. Sequencing of a P. malariae relative that infects chimpanzees reveals similar signatures of selection in the P. malariae lineage to another Plasmodium lineage shown to be capable of colonization of both human and chimpanzee hosts. Molecular dating suggests that these host adaptations occurred over similar evolutionary timescales. In addition to the core genome that is conserved between species, differences in gene content can be linked to their specific biology. The genome suggests that P. malariae expresses a family of heterodimeric proteins on its surface that have structural similarities to a protein crucial for invasion of red blood cells. The data presented here provide insight into the evolution of the Plasmodium genus as a whole.


Asunto(s)
Evolución Molecular , Genoma/genética , Malaria/parasitología , Plasmodium malariae/genética , Plasmodium ovale/genética , Animales , Eritrocitos/parasitología , Femenino , Genómica , Humanos , Pan troglodytes/parasitología , Filogenia
11.
Genome Res ; 29(8): 1250-1261, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31345938

RESUMEN

Polymorphisms in genetic copy number can influence gene expression, coding sequence, and zygosity, making them powerful actors in the evolutionary process. Copy number variants (CNVs) are however understudied, being more difficult to detect than single-nucleotide polymorphisms. We take advantage of the intense selective pressures on the major malaria vector Anopheles gambiae, caused by the widespread use of insecticides for malaria control, to investigate the role of CNVs in the evolution of insecticide resistance. Using the whole-genome sequencing data from 1142 samples in the An. gambiae 1000 genomes project, we identified 250 gene-containing CNVs, encompassing a total of 267 genes of which 28 were in gene families linked to metabolic insecticide resistance, representing significant enrichment of these families. The five major gene clusters for metabolic resistance all contained CNVs, with 44 different CNVs being found across these clusters and multiple CNVs frequently covering the same genes. These 44 CNVs are widespread (45% of individuals carry at least one of them) and have been spreading through positive selection, indicated by their high local frequencies and extended haplotype homozygosity. Our results demonstrate the importance of CNVs in the response to selection, highlighting the urgent need to identify the contribution of each CNV to insecticide resistance and to track their spread as the use of insecticides in malaria endemic countries intensifies and as the operational deployment of next-generation bed nets targeting metabolic resistance gathers pace. Our detailed descriptions of CNVs found across the species range provide the tools to do so.


Asunto(s)
Anopheles/genética , Sistema Enzimático del Citocromo P-450/genética , Variaciones en el Número de Copia de ADN , Genoma de los Insectos , Resistencia a los Insecticidas/genética , Mosquitos Vectores/genética , Animales , Anopheles/parasitología , Evolución Biológica , Mapeo Cromosómico , Sistema Enzimático del Citocromo P-450/metabolismo , Dosificación de Gen , Sitios Genéticos , Haplotipos , Homocigoto , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Insecticidas , Malaria/prevención & control , Malaria/transmisión , Mosquitos Vectores/parasitología , Familia de Multigenes , Piretrinas , Selección Genética , Secuenciación Completa del Genoma
12.
PLoS Pathog ; 16(12): e1009133, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33320907

RESUMEN

The rapid and aggressive spread of artemisinin-resistant Plasmodium falciparum carrying the C580Y mutation in the kelch13 gene is a growing threat to malaria elimination in Southeast Asia, but there is no evidence of their spread to other regions. We conducted cross-sectional surveys in 2016 and 2017 at two clinics in Wewak, Papua New Guinea (PNG) where we identified three infections caused by C580Y mutants among 239 genotyped clinical samples. One of these mutants exhibited the highest survival rate (6.8%) among all parasites surveyed in ring-stage survival assays (RSA) for artemisinin. Analyses of kelch13 flanking regions, and comparisons of deep sequencing data from 389 clinical samples from PNG, Indonesian Papua and Western Cambodia, suggested an independent origin of the Wewak C580Y mutation, showing that the mutants possess several distinctive genetic features. Identity by descent (IBD) showed that multiple portions of the mutants' genomes share a common origin with parasites found in Indonesian Papua, comprising several mutations within genes previously associated with drug resistance, such as mdr1, ferredoxin, atg18 and pnp. These findings suggest that a P. falciparum lineage circulating on the island of New Guinea has gradually acquired a complex ensemble of variants, including kelch13 C580Y, which have affected the parasites' drug sensitivity. This worrying development reinforces the need for increased surveillance of the evolving parasite populations on the island, to contain the spread of resistance.


Asunto(s)
Antiinfecciosos , Artemisininas , Resistencia a Medicamentos/genética , Genes Protozoarios/genética , Plasmodium falciparum/genética , Antiinfecciosos/uso terapéutico , Artemisininas/uso terapéutico , Estudios Transversales , Humanos , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Mutación , Papúa Nueva Guinea
13.
PLoS Comput Biol ; 17(8): e1009287, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34411093

RESUMEN

There is an abundance of malaria genetic data being collected from the field, yet using these data to understand the drivers of regional epidemiology remains a challenge. A key issue is the lack of models that relate parasite genetic diversity to epidemiological parameters. Classical models in population genetics characterize changes in genetic diversity in relation to demographic parameters, but fail to account for the unique features of the malaria life cycle. In contrast, epidemiological models, such as the Ross-Macdonald model, capture malaria transmission dynamics but do not consider genetics. Here, we have developed an integrated model encompassing both parasite evolution and regional epidemiology. We achieve this by combining the Ross-Macdonald model with an intra-host continuous-time Moran model, thus explicitly representing the evolution of individual parasite genomes in a traditional epidemiological framework. Implemented as a stochastic simulation, we use the model to explore relationships between measures of parasite genetic diversity and parasite prevalence, a widely-used metric of transmission intensity. First, we explore how varying parasite prevalence influences genetic diversity at equilibrium. We find that multiple genetic diversity statistics are correlated with prevalence, but the strength of the relationships depends on whether variation in prevalence is driven by host- or vector-related factors. Next, we assess the responsiveness of a variety of statistics to malaria control interventions, finding that those related to mixed infections respond quickly (∼months) whereas other statistics, such as nucleotide diversity, may take decades to respond. These findings provide insights into the opportunities and challenges associated with using genetic data to monitor malaria epidemiology.


Asunto(s)
Variación Genética , Malaria Falciparum/epidemiología , Plasmodium falciparum/patogenicidad , Animales , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Modelos Teóricos , Plasmodium falciparum/genética , Prevalencia
14.
Mol Biol Evol ; 37(10): 2900-2917, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32449755

RESUMEN

The evolution of insecticide resistance mechanisms in natural populations of Anopheles malaria vectors is a major public health concern across Africa. Using genome sequence data, we study the evolution of resistance mutations in the resistance to dieldrin locus (Rdl), a GABA receptor targeted by several insecticides, but most notably by the long-discontinued cyclodiene, dieldrin. The two Rdl resistance mutations (296G and 296S) spread across West and Central African Anopheles via two independent hard selective sweeps that included likely compensatory nearby mutations, and were followed by a rare combination of introgression across species (from A. gambiae and A. arabiensis to A. coluzzii) and across nonconcordant karyotypes of the 2La chromosomal inversion. Rdl resistance evolved in the 1950s as the first known adaptation to a large-scale insecticide-based intervention, but the evolutionary lessons from this system highlight contemporary and future dangers for management strategies designed to combat development of resistance in malaria vectors.


Asunto(s)
Anopheles/genética , Dieldrín , Evolución Molecular , Introgresión Genética , Animales , Inversión Cromosómica , Proteínas de Drosophila , Haplotipos , Resistencia a los Insecticidas/genética , Mutación , Receptores de GABA-A , Selección Genética
15.
Mol Ecol ; 30(21): 5303-5317, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33590926

RESUMEN

Resistance to pyrethroid insecticides is a major concern for malaria vector control. Pyrethroids target the voltage-gated sodium channel (VGSC), an essential component of the mosquito nervous system. Substitutions in the amino acid sequence can induce a resistance phenotype. We use whole-genome sequence data from phase 2 of the Anopheles gambiae 1000 Genomes Project (Ag1000G) to provide a comprehensive account of genetic variation in the Vgsc gene across 13 African countries. In addition to known resistance alleles, we describe 20 other non-synonymous nucleotide substitutions at appreciable population frequency and map these variants onto a protein model to investigate the likelihood of pyrethroid resistance phenotypes. Thirteen of these novel alleles were found to occur almost exclusively on haplotypes carrying the known L995F kdr (knock-down resistance) allele and may enhance or compensate for the L995F resistance genotype. A novel mutation I1527T, adjacent to a predicted pyrethroid-binding site, was found in tight linkage with V402L substitutions, similar to allele combinations associated with resistance in other insect species. We also analysed genetic backgrounds carrying resistance alleles, to determine which alleles have experienced recent positive selection, and describe ten distinct haplotype groups carrying known kdr alleles. Five of these groups are observed in more than one country, in one case separated by over 3000 km, providing new information about the potential for the geographical spread of resistance. Our results demonstrate that the molecular basis of target-site pyrethroid resistance in malaria vectors is more complex than previously appreciated, and provide a foundation for the development of new genetic tools for insecticide resistance management.


Asunto(s)
Anopheles , Insecticidas , Malaria , Piretrinas , Animales , Anopheles/genética , Insecticidas/farmacología , Malaria/genética , Mosquitos Vectores/genética , Piretrinas/farmacología
16.
Malar J ; 20(1): 152, 2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731134

RESUMEN

BACKGROUND: Anti-malarial drug resistance remains a key concern for the global fight against malaria. In Ghana sulfadoxine-pyrimethamine (SP) is used for intermittent preventive treatment of malaria in pregnancy and combined with amodiaquine for Seasonal Malaria Chemoprevention (SMC) during the high malaria season. Thus, surveillance of molecular markers of SP resistance is important to guide decision-making for these interventions in Ghana. METHODS: A total of 4469 samples from uncomplicated malaria patients collected from 2009 to 2018 was submitted to the Wellcome Trust Sanger Institute, UK for DNA sequencing using MiSeq. Genotypes were successfully translated into haplotypes in 2694 and 846 mono infections respectively for pfdhfr and pfdhps genes and the combined pfhdfr/pfdhps genes across all years. RESULTS: At the pfdhfr locus, a consistently high (> 60%) prevalence of parasites carrying triple mutants (IRNI) were detected from 2009 to 2018. Two double mutant haplotypes (NRNI and ICNI) were found, with haplotype NRNI having a much higher prevalence (average 13.8%) than ICNI (average 3.2%) across all years. Six pfdhps haplotypes were detected. Of these, prevalence of five fluctuated in a downward trend over time from 2009 to 2018, except a pfdhps double mutant (AGKAA), which increased consistently from 2.5% in 2009 to 78.2% in 2018. Across both genes, pfdhfr/pfdhps combined triple (NRNI + AAKAA) mutants were only detected in 2009, 2014, 2015 and 2018, prevalence of which fluctuated between 3.5 and 5.5%. The combined quadruple (IRNI + AAKAA) genotype increased in prevalence from 19.3% in 2009 to 87.5% in 2011 before fluctuating downwards to 19.6% in 2018 with an average prevalence of 37.4% within the nine years. Prevalence of parasites carrying the quintuple (IRNI + AGKAA or SGEAA) mutant haplotypes, which are highly refractory to SP increased over time from 14.0% in 2009 to 89.0% in 2016 before decreasing to 78.9 and 76.6% in 2017 and 2018 respectively. Though quintuple mutants are rising in prevalence in both malaria seasons, together these combined genotypes vary significantly within season but not between seasons. CONCLUSIONS: Despite high prevalence of pfdhfr triple mutants and combined pfdhfr/pfdhps quadruple and quintuple mutants in this setting SP may still be efficacious. These findings are significant as they highlight the need to continuously monitor SP resistance, particularly using deep targeted sequencing to ascertain changing resistance patterns.


Asunto(s)
Antimaláricos/farmacología , Resistencia a Medicamentos/genética , Variación Genética , Genotipo , Malaria Falciparum/prevención & control , Plasmodium falciparum/genética , Pirimetamina/farmacología , Sulfadoxina/farmacología , Adolescente , Niño , Preescolar , Combinación de Medicamentos , Femenino , Variación Genética/efectos de los fármacos , Ghana , Humanos , Masculino , Plasmodium falciparum/efectos de los fármacos , Estaciones del Año , Adulto Joven
17.
Nature ; 526(7572): 253-7, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26416757

RESUMEN

The high prevalence of sickle haemoglobin in Africa shows that malaria has been a major force for human evolutionary selection, but surprisingly few other polymorphisms have been proven to confer resistance to malaria in large epidemiological studies. To address this problem, we conducted a multi-centre genome-wide association study (GWAS) of life-threatening Plasmodium falciparum infection (severe malaria) in over 11,000 African children, with replication data in a further 14,000 individuals. Here we report a novel malaria resistance locus close to a cluster of genes encoding glycophorins that are receptors for erythrocyte invasion by P. falciparum. We identify a haplotype at this locus that provides 33% protection against severe malaria (odds ratio = 0.67, 95% confidence interval = 0.60-0.76, P value = 9.5 × 10(-11)) and is linked to polymorphisms that have previously been shown to have features of ancient balancing selection, on the basis of haplotype sharing between humans and chimpanzees. Taken together with previous observations on the malaria-protective role of blood group O, these data reveal that two of the strongest GWAS signals for severe malaria lie in or close to genes encoding the glycosylated surface coat of the erythrocyte cell membrane, both within regions of the genome where it appears that evolution has maintained diversity for millions of years. These findings provide new insights into the host-parasite interactions that are critical in determining the outcome of malaria infection.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Malaria Falciparum/genética , Selección Genética/genética , Sistema del Grupo Sanguíneo ABO , África/epidemiología , Animales , Niño , Secuencia Conservada/genética , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Evolución Molecular , Proteínas de la Matriz Extracelular/genética , Femenino , Glicoforinas/genética , Haplotipos/genética , Interacciones Huésped-Parásitos/genética , Humanos , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Masculino , Pan troglodytes/genética , Plasmodium falciparum/fisiología , Polimorfismo de Nucleótido Simple/genética
18.
Nature ; 517(7534): 327-32, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25470054

RESUMEN

Given the importance of Africa to studies of human origins and disease susceptibility, detailed characterization of African genetic diversity is needed. The African Genome Variation Project provides a resource with which to design, implement and interpret genomic studies in sub-Saharan Africa and worldwide. The African Genome Variation Project represents dense genotypes from 1,481 individuals and whole-genome sequences from 320 individuals across sub-Saharan Africa. Using this resource, we find novel evidence of complex, regionally distinct hunter-gatherer and Eurasian admixture across sub-Saharan Africa. We identify new loci under selection, including loci related to malaria susceptibility and hypertension. We show that modern imputation panels (sets of reference genotypes from which unobserved or missing genotypes in study sets can be inferred) can identify association signals at highly differentiated loci across populations in sub-Saharan Africa. Using whole-genome sequencing, we demonstrate further improvements in imputation accuracy, strengthening the case for large-scale sequencing efforts of diverse African haplotypes. Finally, we present an efficient genotype array design capturing common genetic variation in Africa.


Asunto(s)
Variación Genética/genética , Genética Médica/tendencias , Genoma Humano/genética , Genómica/tendencias , África , África del Sur del Sahara , Asia/etnología , Europa (Continente)/etnología , Humanos , Factores de Riesgo , Selección Genética/genética
19.
BMC Med ; 18(1): 45, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32127002

RESUMEN

BACKGROUND: Spread of malaria and antimalarial resistance through human movement present major threats to current goals to eliminate the disease. Bordering the Greater Mekong Subregion, southeast Bangladesh is a potentially important route of spread to India and beyond, but information on travel patterns in this area are lacking. METHODS: Using a standardised short survey tool, 2090 patients with malaria were interviewed at 57 study sites in 2015-2016 about their demographics and travel patterns in the preceding 2 months. RESULTS: Most travel was in the south of the study region between Cox's Bazar district (coastal region) to forested areas in Bandarban (31% by days and 45% by nights), forming a source-sink route. Less than 1% of travel reported was between the north and south forested areas of the study area. Farmers (21%) and students (19%) were the top two occupations recorded, with 67 and 47% reporting travel to the forest respectively. Males aged 25-49 years accounted for 43% of cases visiting forests but only 24% of the study population. Children did not travel. Women, forest dwellers and farmers did not travel beyond union boundaries. Military personnel travelled the furthest especially to remote forested areas. CONCLUSIONS: The approach demonstrated here provides a framework for identifying key traveller groups and their origins and destinations of travel in combination with knowledge of local epidemiology to inform malaria control and elimination efforts. Working with the NMEP, the findings were used to derive a set of policy recommendations to guide targeting of interventions for elimination.


Asunto(s)
Malaria/epidemiología , Viaje/tendencias , Adolescente , Adulto , Bangladesh , Femenino , Humanos , India , Masculino , Estudios Prospectivos , Adulto Joven
20.
PLoS Pathog ; 14(11): e1007436, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30496294

RESUMEN

Invasion of human erythrocytes is essential for Plasmodium falciparum parasite survival and pathogenesis, and is also a complex phenotype. While some later steps in invasion appear to be invariant and essential, the earlier steps of recognition are controlled by a series of redundant, and only partially understood, receptor-ligand interactions. Reverse genetic analysis of laboratory adapted strains has identified multiple genes that when deleted can alter invasion, but how the relative contributions of each gene translate to the phenotypes of clinical isolates is far from clear. We used a forward genetic approach to identify genes responsible for variable erythrocyte invasion by phenotyping the parents and progeny of previously generated experimental genetic crosses. Linkage analysis using whole genome sequencing data revealed a single major locus was responsible for the majority of phenotypic variation in two invasion pathways. This locus contained the PfRh2a and PfRh2b genes, members of one of the major invasion ligand gene families, but not widely thought to play such a prominent role in specifying invasion phenotypes. Variation in invasion pathways was linked to significant differences in PfRh2a and PfRh2b expression between parasite lines, and their role in specifying alternative invasion was confirmed by CRISPR-Cas9-mediated genome editing. Expansion of the analysis to a large set of clinical P. falciparum isolates revealed common deletions, suggesting that variation at this locus is a major cause of invasion phenotypic variation in the endemic setting. This work has implications for blood-stage vaccine development and will help inform the design and location of future large-scale studies of invasion in clinical isolates.


Asunto(s)
Eritrocitos/parasitología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Animales , Anticuerpos Antiprotozoarios/inmunología , Proteínas Portadoras/metabolismo , Pruebas Genéticas/métodos , Humanos , Ligandos , Fenotipo , Proteínas Protozoarias/metabolismo , Reticulocitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA