Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 32(1): 203-213, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34764149

RESUMEN

Cryptosporidiosis is a leading cause of waterborne diarrheal disease globally and an important contributor to mortality in infants and the immunosuppressed. Despite its importance, the Cryptosporidium community has only had access to a good, but incomplete, Cryptosporidium parvum IOWA reference genome sequence. Incomplete reference sequences hamper annotation, experimental design, and interpretation. We have generated a new C. parvum IOWA genome assembly supported by Pacific Biosciences (PacBio) and Oxford Nanopore long-read technologies and a new comparative and consistent genome annotation for three closely related species: C. parvum, Cryptosporidium hominis, and Cryptosporidium tyzzeri We made 1926 C. parvum annotation updates based on experimental evidence. They include new transporters, ncRNAs, introns, and altered gene structures. The new assembly and annotation revealed a complete Dnmt2 methylase ortholog. Comparative annotation between C. parvum, C. hominis, and C. tyzzeri revealed that most "missing" orthologs are found, suggesting that the biological differences between the species must result from gene copy number variation, differences in gene regulation, and single-nucleotide variants (SNVs). Using the new assembly and annotation as reference, 190 genes are identified as evolving under positive selection, including many not detected previously. The new C. parvum IOWA reference genome assembly is larger, gap free, and lacks ambiguous bases. This chromosomal assembly recovers all 16 chromosome ends, 13 of which are contiguously assembled. The three remaining chromosome ends are provisionally placed. These ends represent duplication of entire chromosome ends including subtelomeric regions revealing a new level of genome plasticity that will both inform and impact future research.


Asunto(s)
Criptosporidiosis , Cryptosporidium , Criptosporidiosis/genética , Cryptosporidium/genética , Variaciones en el Número de Copia de ADN , Genoma , Humanos , Telómero/genética
2.
Antimicrob Agents Chemother ; 68(3): e0143223, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38289079

RESUMEN

We previously performed a genome-wide association study (GWAS) to identify the genetic basis of praziquantel (PZQ) response in schistosomes, identifying two quantitative trait loci situated on chromosomes 2 and 3. We reanalyzed this GWAS using the latest (version 10) genome assembly showing that a single locus on chromosome 3, rather than two independent loci, determines drug response. These results reveal that PZQ response is monogenic and demonstrates the importance of high-quality genomic information.


Asunto(s)
Antihelmínticos , Esquistosomiasis mansoni , Animales , Praziquantel/farmacología , Praziquantel/uso terapéutico , Schistosoma mansoni/genética , Estudio de Asociación del Genoma Completo , Resistencia a Medicamentos , Esquistosomiasis mansoni/tratamiento farmacológico , Antihelmínticos/farmacología , Antihelmínticos/uso terapéutico
3.
PLoS Pathog ; 18(8): e1010706, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35939508

RESUMEN

Hybridization between different species of parasites is increasingly being recognised as a major public and veterinary health concern at the interface of infectious diseases biology, evolution, epidemiology and ultimately control. Recent research has revealed that viable hybrids and introgressed lineages between Schistosoma spp. are prevalent across Africa and beyond, including those with zoonotic potential. However, it remains unclear whether these hybrid lineages represent recent hybridization events, suggesting hybridization is ongoing, and/or whether they represent introgressed lineages derived from ancient hybridization events. In human schistosomiasis, investigation is hampered by the inaccessibility of adult-stage worms due to their intravascular location, an issue which can be circumvented by post-mortem of livestock at abattoirs for Schistosoma spp. of known zoonotic potential. To characterise the composition of naturally-occurring schistosome hybrids, we performed whole-genome sequencing of 21 natural livestock infective schistosome isolates. To facilitate this, we also assembled a de novo chromosomal-scale draft assembly of Schistosoma curassoni. Genomic analyses identified isolates of S. bovis, S. curassoni and hybrids between the two species, all of which were early generation hybrids with multiple generations found within the same host. These results show that hybridization is an ongoing process within natural populations with the potential to further challenge elimination efforts against schistosomiasis.


Asunto(s)
Schistosoma , Esquistosomiasis , Animales , Genoma , Genómica , Humanos , Hibridación Genética , Ganado/parasitología , Schistosoma/genética , Esquistosomiasis/epidemiología , Esquistosomiasis/genética , Esquistosomiasis/veterinaria
4.
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
5.
Proc Natl Acad Sci U S A ; 117(40): 25159-25168, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958676

RESUMEN

The tropical Andes are an important natural laboratory to understand speciation in many taxa. Here we examined the evolutionary history of parasites of the Leishmania braziliensis species complex based on whole-genome sequencing of 67 isolates from 47 localities in Peru. We first show the origin of Andean Leishmania as a clade of near-clonal lineages that diverged from admixed Amazonian ancestors, accompanied by a significant reduction in genome diversity and large structural variations implicated in host-parasite interactions. Within the Andean species, patterns of population structure were strongly associated with biogeographical origin. Molecular clock and ecological niche modeling suggested that the history of diversification of the Andean lineages is limited to the Late Pleistocene and intimately associated with habitat contractions driven by climate change. These results suggest that changes in forestation over the past 150,000 y have influenced speciation and diversity of these Neotropical parasites. Second, genome-scale analyses provided evidence of meiotic-like recombination between Andean and Amazonian Leishmania species, resulting in full-genome hybrids. The mitochondrial genome of these hybrids consisted of homogeneous uniparental maxicircles, but minicircles originated from both parental species. We further show that mitochondrial minicircles-but not maxicircles-show a similar evolutionary pattern to the nuclear genome, suggesting that compatibility between nuclear-encoded mitochondrial genes and minicircle-encoded guide RNA genes is essential to maintain efficient respiration. By comparing full nuclear and mitochondrial genome ancestries, our data expand our appreciation on the genetic consequences of diversification and hybridization in parasitic protozoa.


Asunto(s)
Genoma Mitocondrial/genética , Interacciones Huésped-Parásitos/genética , Leishmania braziliensis/genética , Leishmaniasis Cutánea/genética , Ecosistema , Bosques , Especiación Genética , Humanos , Leishmania braziliensis/patogenicidad , Leishmaniasis Cutánea/epidemiología , Leishmaniasis Cutánea/parasitología , Perú/epidemiología , Filogeografía
6.
J Infect Dis ; 225(8): 1435-1446, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-32524140

RESUMEN

BACKGROUND: Schistosomiasis is a major global health problem caused by blood-dwelling parasitic worms, which is currently tackled primarily by mass administration of the drug praziquantel. Appropriate drug treatment strategies are informed by diagnostics that establish the prevalence and intensity of infection, which, in regions of low transmission, should be highly sensitive. METHODS: To identify sensitive new serological markers of Schistosoma mansoni infections, we have compiled a recombinant protein library of parasite cell-surface and secreted proteins expressed in mammalian cells. RESULTS: Together with a time series of sera samples from volunteers experimentally infected with a defined number of male parasites, we probed this protein library to identify several markers that can detect primary infections with as low as 10 parasites and as early as 5 weeks postinfection. CONCLUSIONS: These new markers could be further explored as valuable tools to detect ongoing and previous S mansoni infections, including in endemic regions where transmission is low.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Animales , Biomarcadores , Humanos , Masculino , Mamíferos , Ratones , Praziquantel/uso terapéutico , Proteínas Recombinantes , Schistosoma mansoni , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis mansoni/diagnóstico , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/parasitología
7.
BMC Genomics ; 23(1): 398, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35610562

RESUMEN

Genome architecture describes how genes and other features are arranged in genomes. These arrangements reflect the evolutionary pressures on genomes and underlie biological processes such as chromosomal segregation and the regulation of gene expression. We present a new tool called Genome Decomposition Analysis (GDA) that characterises genome architectures and acts as an accessible approach for discovering hidden features of a genome assembly. With the imminent deluge of high-quality genome assemblies from projects such as the Darwin Tree of Life and the Earth BioGenome Project, GDA has been designed to facilitate their exploration and the discovery of novel genome biology. We highlight the effectiveness of our approach in characterising the genome architectures of single-celled eukaryotic parasites from the phylum Apicomplexa and show that it scales well to large genomes.


Asunto(s)
Apicomplexa , Parásitos , Animales , Apicomplexa/genética , Evolución Biológica , Eucariontes/genética , Genoma , Parásitos/genética
8.
BMC Genomics ; 23(1): 780, 2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36451097

RESUMEN

BACKGROUND: Important regulation occurs at the level of transcription in Plasmodium falciparum and growing evidence suggests that these apicomplexan parasites have complex regulatory networks. Recent studies implicate long noncoding RNAs (lncRNAs) as transcriptional regulators in P. falciparum. However, due to limited research and the lack of necessary experimental tools, our understanding of their role in the malaria-causing parasite remains largely unelucidated. In this work, we address one of these limitations, the lack of an updated and improved lncRNA annotation in P. falciparum. RESULTS: We generated long-read RNA sequencing data and integrated information extracted and curated from multiple sources to manually annotate lncRNAs. We identified 1119 novel lncRNAs and validated and refined 1250 existing annotations. Utilising the collated datasets, we generated evidence-based ranking scores for each annotation and characterised the distinct genomic contexts and features of P. falciparum lncRNAs. Certain features indicated subsets with potential biological significance such as 25 lncRNAs containing multiple introns, 335 lncRNAs lacking mutations in piggyBac mutagenic studies and lncRNAs associated with specific biologic processes including two new types of lncRNAs found proximal to var genes. CONCLUSIONS: The insights and the annotation presented in this study will serve as valuable tools for researchers seeking to understand the role of lncRNAs in parasite biology through both bioinformatics and experimental approaches.


Asunto(s)
Malaria Falciparum , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Genómica , Malaria Falciparum/genética , Plasmodium falciparum/genética , Biología Computacional
9.
PLoS Pathog ; 16(8): e1008717, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32745123

RESUMEN

Hepatocystis is a genus of single-celled parasites infecting, amongst other hosts, monkeys, bats and squirrels. Although thought to have descended from malaria parasites (Plasmodium spp.), Hepatocystis spp. are thought not to undergo replication in the blood-the part of the Plasmodium life cycle which causes the symptoms of malaria. Furthermore, Hepatocystis is transmitted by biting midges, not mosquitoes. Comparative genomics of Hepatocystis and Plasmodium species therefore presents an opportunity to better understand some of the most important aspects of malaria parasite biology. We were able to generate a draft genome for Hepatocystis sp. using DNA sequencing reads from the blood of a naturally infected red colobus monkey. We provide robust phylogenetic support for Hepatocystis sp. as a sister group to Plasmodium parasites infecting rodents. We show transcriptomic support for a lack of replication in the blood and genomic support for a complete loss of a family of genes involved in red blood cell invasion. Our analyses highlight the rapid evolution of genes involved in parasite vector stages, revealing genes that may be critical for interactions between malaria parasites and mosquitoes.


Asunto(s)
Apicomplexa/genética , Sangre/parasitología , Colobus/parasitología , Malaria/veterinaria , Enfermedades de los Monos/parasitología , Plasmodium/genética , Infecciones Protozoarias en Animales/parasitología , Animales , Apicomplexa/clasificación , Apicomplexa/fisiología , Genoma de Protozoos , Malaria/sangre , Malaria/parasitología , Enfermedades de los Monos/sangre , Filogenia , Plasmodium/clasificación , Plasmodium/fisiología , Infecciones Protozoarias en Animales/sangre , Transcriptoma
10.
BMC Biol ; 19(1): 255, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34852797

RESUMEN

BACKGROUND: The consequences of the earth's daily rotation have led to 24-h biological rhythms in most organisms. Even some parasites are known to have daily rhythms, which, when in synchrony with host rhythms, can optimise their fitness. Understanding these rhythms may enable the development of control strategies that take advantage of rhythmic vulnerabilities. Recent work on protozoan parasites has revealed 24-h rhythms in gene expression, drug sensitivity and the presence of an intrinsic circadian clock; however, similar studies on metazoan parasites are lacking. To address this, we investigated if a metazoan parasite has daily molecular oscillations, whether they reveal how these longer-lived organisms can survive host daily cycles over a lifespan of many years and if animal circadian clock genes are present and rhythmic. We addressed these questions using the human blood fluke Schistosoma mansoni that lives in the vasculature for decades and causes the tropical disease schistosomiasis. RESULTS: Using round-the-clock transcriptomics of male and female adult worms collected from experimentally infected mice, we discovered that ~ 2% of its genes followed a daily pattern of expression. Rhythmic processes included a stress response during the host's active phase and a 'peak in metabolic activity' during the host's resting phase. Transcriptional profiles in the female reproductive system were mirrored by daily patterns in egg laying (eggs are the main drivers of the host pathology). Genes cycling with the highest amplitudes include predicted drug targets and a vaccine candidate. These 24-h rhythms may be driven by host rhythms and/or generated by a circadian clock; however, orthologs of core clock genes are missing and secondary clock genes show no 24-h rhythmicity. CONCLUSIONS: There are daily rhythms in the transcriptomes of adult S. mansoni, but they appear less pronounced than in other organisms. The rhythms reveal temporally compartmentalised internal processes and host interactions relevant to within-host survival and between-host transmission. Our findings suggest that if these daily rhythms are generated by an intrinsic circadian clock then the oscillatory mechanism must be distinct from that in other animals. We have shown which transcripts oscillate at this temporal scale and this will benefit the development and delivery of treatments against schistosomiasis.


Asunto(s)
Relojes Circadianos , Parásitos , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Femenino , Humanos , Masculino , Ratones , Parásitos/genética , Schistosoma mansoni/genética , Transcriptoma
11.
Genome Res ; 28(9): 1383-1394, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30006414

RESUMEN

African trypanosomes are vector-borne hemoparasites of humans and animals. In the mammal, parasites evade the immune response through antigenic variation. Periodic switching of the variant surface glycoprotein (VSG) coat covering their cell surface allows sequential expansion of serologically distinct parasite clones. Trypanosome genomes contain many hundreds of VSG genes, subject to rapid changes in nucleotide sequence, copy number, and chromosomal position. Thus, analyzing, or even quantifying, VSG diversity over space and time presents an enormous challenge to conventional techniques. Indeed, previous population genomic studies have overlooked this vital aspect of pathogen biology for lack of analytical tools. Here we present a method for analyzing population-scale VSG diversity in Trypanosoma congolense from deep sequencing data. Previously, we suggested that T. congolense VSGs segregate into defined "phylotypes" that do not recombine. In our data set comprising 41 T. congolense genome sequences from across Africa, these phylotypes are universal and exhaustive. Screening sequence contigs with diagnostic protein motifs accurately quantifies relative phylotype frequencies, providing a metric of VSG diversity, called the "variant antigen profile." We applied our metric to VSG expression in the tsetse fly, showing that certain, rare VSG phylotypes may be preferentially expressed in infective, metacyclic-stage parasites. Hence, variant antigen profiling accurately and rapidly determines the T. congolense VSG gene and transcript repertoire from sequence data, without need for manual curation or highly contiguous sequences. It offers a tractable approach to measuring VSG diversity across strains and during infections, which is imperative to understanding the host-parasite interaction at population and individual scales.


Asunto(s)
Polimorfismo Genético , Análisis de Secuencia de ADN/métodos , Trypanosoma congolense/genética , Glicoproteínas Variantes de Superficie de Trypanosoma/genética , Secuencias de Aminoácidos , Animales , Masculino , Trypanosoma congolense/inmunología , Trypanosoma congolense/patogenicidad , Moscas Tse-Tse/parasitología , Glicoproteínas Variantes de Superficie de Trypanosoma/química , Glicoproteínas Variantes de Superficie de Trypanosoma/inmunología
12.
Genome Res ; 28(4): 547-560, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29500236

RESUMEN

Avian malaria parasites are prevalent around the world and infect a wide diversity of bird species. Here, we report the sequencing and analysis of high-quality draft genome sequences for two avian malaria species, Plasmodium relictum and Plasmodium gallinaceum We identify 50 genes that are specific to avian malaria, located in an otherwise conserved core of the genome that shares gene synteny with all other sequenced malaria genomes. Phylogenetic analysis suggests that the avian malaria species form an outgroup to the mammalian Plasmodium species, and using amino acid divergence between species, we estimate the avian- and mammalian-infective lineages diverged in the order of 10 million years ago. Consistent with their phylogenetic position, we identify orthologs of genes that had previously appeared to be restricted to the clades of parasites containing Plasmodium falciparum and Plasmodium vivax, the species with the greatest impact on human health. From these orthologs, we explore differential diversifying selection across the genus and show that the avian lineage is remarkable in the extent to which invasion-related genes are evolving. The subtelomeres of the P. relictum and P. gallinaceum genomes contain several novel gene families, including an expanded surf multigene family. We also identify an expansion of reticulocyte binding protein homologs in P. relictum, and within these proteins, we detect distinct regions that are specific to nonhuman primate, humans, rodent, and avian hosts. For the first time in the Plasmodium lineage, we find evidence of transposable elements, including several hundred fragments of LTR-retrotransposons in both species and an apparently complete LTR-retrotransposon in the genome of P. gallinaceum.


Asunto(s)
Malaria Aviar/genética , Plasmodium falciparum/genética , Plasmodium vivax/genética , Plasmodium/genética , Animales , Aves/parasitología , Evolución Molecular , Humanos , Malaria Aviar/parasitología , Mamíferos/parasitología , Filogenia , Plasmodium/patogenicidad , Plasmodium falciparum/patogenicidad , Plasmodium vivax/patogenicidad
13.
PLoS Pathog ; 15(1): e1007265, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30640950

RESUMEN

The whipworm Trichuris trichiura is a soil-transmitted helminth that dwells in the epithelium of the caecum and proximal colon of their hosts causing the human disease, trichuriasis. Trichuriasis is characterized by colitis attributed to the inflammatory response elicited by the parasite while tunnelling through intestinal epithelial cells (IECs). The IL-10 family of receptors, comprising combinations of subunits IL-10Rα, IL-10Rß, IL-22Rα and IL-28Rα, modulates intestinal inflammatory responses. Here we carefully dissected the role of these subunits in the resistance of mice to infection with T. muris, a mouse model of the human whipworm T. trichiura. Our findings demonstrate that whilst IL-22Rα and IL-28Rα are dispensable in the host response to whipworms, IL-10 signalling through IL-10Rα and IL-10Rß is essential to control caecal pathology, worm expulsion and survival during T. muris infections. We show that deficiency of IL-10, IL-10Rα and IL-10Rß results in dysbiosis of the caecal microbiota characterised by expanded populations of opportunistic bacteria of the families Enterococcaceae and Enterobacteriaceae. Moreover, breakdown of the epithelial barrier after whipworm infection in IL-10, IL-10Rα and IL-10Rß-deficient mice, allows the translocation of these opportunistic pathogens or their excretory products to the liver causing organ failure and lethal disease. Importantly, bone marrow chimera experiments indicate that signalling through IL-10Rα and IL-10Rß in haematopoietic cells, but not IECs, is crucial to control worm expulsion and immunopathology. These findings are supported by worm expulsion upon infection of conditional mutant mice for the IL-10Rα on IECs. Our findings emphasize the pivotal and complex role of systemic IL-10Rα signalling on immune cells in promoting microbiota homeostasis and maintaining the intestinal epithelial barrier, thus preventing immunopathology during whipworm infections.


Asunto(s)
Interleucina-10/metabolismo , Receptores de Interleucina-10/metabolismo , Trichuris/inmunología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/inmunología , Homeostasis , Interleucinas/metabolismo , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Tricuriasis/inmunología , Trichuris/parasitología , Interleucina-22
14.
J Infect Dis ; 221(6): 948-955, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-30852586

RESUMEN

BACKGROUND: Plasmodium malariae is considered a minor malaria parasite, although its global disease burden is underappreciated. The aim of this study was to develop an induced blood-stage malaria (IBSM) model of P. malariae to study parasite biology, diagnostic assays, and treatment. METHODS: This clinical trial involved 2 healthy subjects who were intravenously inoculated with cryopreserved P. malariae-infected erythrocytes. Subjects were treated with artemether-lumefantrine after development of clinical symptoms. Prior to antimalarial therapy, mosquito-feeding assays were performed to investigate transmission, and blood samples were collected for rapid diagnostic testing and parasite transcription profiling. Serial blood samples were collected for biomarker analysis. RESULTS: Both subjects experienced symptoms and signs typical of early malaria. Parasitemia was detected 7 days after inoculation, and parasite concentrations increased until antimalarial treatment was initiated 25 and 21 days after inoculation for subjects 1 and 2 respectively (peak parasitemia levels, 174 182 and 50 291 parasites/mL, respectively). The parasite clearance half-life following artemether-lumefantrine treatment was 6.7 hours. Mosquito transmission was observed for 1 subject, while in vivo parasite transcription and biomarkers were successfully profiled. CONCLUSIONS: An IBSM model of P. malariae has been successfully developed and may be used to study the biology of, diagnostic testing for, and treatment of this neglected malaria species. CLINICAL TRIALS REGISTRATION: ACTRN12617000048381.


Asunto(s)
Antimaláricos/uso terapéutico , Combinación Arteméter y Lumefantrina/uso terapéutico , Malaria/sangre , Malaria/parasitología , Plasmodium malariae/genética , Adolescente , Animales , Anopheles/parasitología , Conducta Alimentaria , Humanos , Malaria/patología , Masculino , Parasitemia/sangre , Parasitemia/tratamiento farmacológico , Parasitemia/parasitología , Plasmodium malariae/fisiología , Transcriptoma , Adulto Joven
15.
BMC Genomics ; 21(1): 395, 2020 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-32513207

RESUMEN

BACKGROUND: Plasmodium parasites undergo several major developmental transitions during their complex lifecycle, which are enabled by precisely ordered gene expression programs. Transcriptomes from the 48-h blood stages of the major human malaria parasite Plasmodium falciparum have been described using cDNA microarrays and RNA-seq, but these assays have not always performed well within non-coding regions, where the AT-content is often 90-95%. RESULTS: We developed a directional, amplification-free RNA-seq protocol (DAFT-seq) to reduce bias against AT-rich cDNA, which we have applied to three strains of P. falciparum (3D7, HB3 and IT). While strain-specific differences were detected, overall there is strong conservation between the transcriptional profiles. For the 3D7 reference strain, transcription was detected from 89% of the genome, with over 78% of the genome transcribed into mRNAs. We also find that transcription from bidirectional promoters frequently results in non-coding, antisense transcripts. These datasets allowed us to refine the 5' and 3' untranslated regions (UTRs), which can be variable, long (> 1000 nt), and often overlap those of adjacent transcripts. CONCLUSIONS: The approaches applied in this study allow a refined description of the transcriptional landscape of P. falciparum and demonstrate that very little of the densely packed P. falciparum genome is inactive or redundant. By capturing the 5' and 3' ends of mRNAs, we reveal both constant and dynamic use of transcriptional start sites across the intraerythrocytic developmental cycle that will be useful in guiding the definition of regulatory regions for use in future experimental gene expression studies.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/genética , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Humanos , Estadios del Ciclo de Vida , Técnicas de Amplificación de Ácido Nucleico/métodos , Plasmodium falciparum/clasificación , Plasmodium falciparum/genética , ARN Mensajero/genética , Especificidad de la Especie
16.
PLoS Pathog ; 14(1): e1006718, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29346437

RESUMEN

Schistosomes are blood-dwelling trematodes with global impact on human and animal health. Because medical treatment is currently based on a single drug, praziquantel, there is urgent need for the development of alternative control strategies. The Schistosoma mansoni genome project provides a platform to study and connect the genetic repertoire of schistosomes to specific biological functions essential for successful parasitism. G protein-coupled receptors (GPCRs) form the largest superfamily of transmembrane receptors throughout the Eumetazoan phyla, including platyhelminths. Due to their involvement in diverse biological processes, their pharmacological importance, and proven druggability, GPCRs are promising targets for new anthelmintics. However, to identify candidate receptors, a more detailed understanding of the roles of GPCR signalling in schistosome biology is essential. An updated phylogenetic analysis of the S. mansoni GPCR genome (GPCRome) is presented, facilitated by updated genome data that allowed a more precise annotation of GPCRs. Additionally, we review the current knowledge on GPCR signalling in this parasite and provide new insights into the potential roles of GPCRs in schistosome reproduction based on the findings of a recent tissue-specific transcriptomic study in paired and unpaired S. mansoni. According to the current analysis, GPCRs contribute to gonad-specific functions but also to nongonad, pairing-dependent processes. The latter may regulate gonad-unrelated functions during the multifaceted male-female interaction. Finally, we compare the schistosome GPCRome to that of another parasitic trematode, Fasciola, and discuss the importance of GPCRs to basic and applied research. Phylogenetic analyses display GPCR diversity in free-living and parasitic platyhelminths and suggest diverse functions in schistosomes. Although their roles need to be substantiated by functional studies in the future, the data support the selection of GPCR candidates for basic and applied studies, invigorating the exploitation of this important receptor class for drug discovery against schistosomes but also other trematodes.


Asunto(s)
Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Proteínas del Helminto/metabolismo , Modelos Biológicos , Schistosoma mansoni/metabolismo , Transducción de Señal , Animales , Antiplatelmínticos/farmacología , Fasciola/efectos de los fármacos , Fasciola/genética , Fasciola/metabolismo , Fasciola/patogenicidad , Quinasas de Receptores Acoplados a Proteína-G/antagonistas & inhibidores , Quinasas de Receptores Acoplados a Proteína-G/química , Quinasas de Receptores Acoplados a Proteína-G/genética , Perfilación de la Expresión Génica , Genoma de los Helmintos , Genómica/métodos , Proteínas del Helminto/antagonistas & inhibidores , Proteínas del Helminto/química , Proteínas del Helminto/genética , Humanos , Especificidad de Órganos , Filogenia , Inhibidores de Proteínas Quinasas/farmacología , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/genética , Schistosoma mansoni/patogenicidad , Transducción de Señal/efectos de los fármacos
17.
Nature ; 507(7491): 253-257, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24572359

RESUMEN

Commitment to and completion of sexual development are essential for malaria parasites (protists of the genus Plasmodium) to be transmitted through mosquitoes. The molecular mechanism(s) responsible for commitment have been hitherto unknown. Here we show that PbAP2-G, a conserved member of the apicomplexan AP2 (ApiAP2) family of DNA-binding proteins, is essential for the commitment of asexually replicating forms to sexual development in Plasmodium berghei, a malaria parasite of rodents. PbAP2-G was identified from mutations in its encoding gene, PBANKA_143750, which account for the loss of sexual development frequently observed in parasites transmitted artificially by blood passage. Systematic gene deletion of conserved ApiAP2 genes in Plasmodium confirmed the role of PbAP2-G and revealed a second ApiAP2 member (PBANKA_103430, here termed PbAP2-G2) that significantly modulates but does not abolish gametocytogenesis, indicating that a cascade of ApiAP2 proteins are involved in commitment to the production and maturation of gametocytes. The data suggest a mechanism of commitment to gametocytogenesis in Plasmodium consistent with a positive feedback loop involving PbAP2-G that could be exploited to prevent the transmission of this pernicious parasite.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Germinativas/crecimiento & desarrollo , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Desarrollo Sexual/genética , Animales , Culicidae/parasitología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Mutación/genética , Plasmodium berghei/citología , Transporte de Proteínas , Proteínas Protozoarias/genética , Reproducción Asexuada , Transcripción Genética
18.
Nucleic Acids Res ; 46(D1): D869-D874, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29069413

RESUMEN

WormBase (http://www.wormbase.org) is an important knowledge resource for biomedical researchers worldwide. To accommodate the ever increasing amount and complexity of research data, WormBase continues to advance its practices on data acquisition, curation and retrieval to most effectively deliver comprehensive knowledge about Caenorhabditis elegans, and genomic information about other nematodes and parasitic flatworms. Recent notable enhancements include user-directed submission of data, such as micropublication; genomic data curation and presentation, including additional genomes and JBrowse, respectively; new query tools, such as SimpleMine, Gene Enrichment Analysis; new data displays, such as the Person Lineage browser and the Summary of Ontology-based Annotations. Anticipating more rapid data growth ahead, WormBase continues the process of migrating to a cutting-edge database technology to achieve better stability, scalability, reproducibility and a faster response time. To better serve the broader research community, WormBase, with five other Model Organism Databases and The Gene Ontology project, have begun to collaborate formally as the Alliance of Genome Resources.


Asunto(s)
Bases de Datos Genéticas , Genoma , Nematodos/genética , Animales , Caenorhabditis/genética , Caenorhabditis elegans/genética , Curaduría de Datos , Minería de Datos , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Predicción , Ontología de Genes , Humanos , Almacenamiento y Recuperación de la Información , Platelmintos/genética , Edición , Interferencia de ARN , Alineación de Secuencia , Interfaz Usuario-Computador , Navegador Web
19.
BMC Genomics ; 20(1): 218, 2019 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-30876405

RESUMEN

BACKGROUND: Infections with helminths cause an enormous disease burden in billions of animals and plants worldwide. Large scale use of anthelmintics has driven the evolution of resistance in a number of species that infect livestock and companion animals, and there are growing concerns regarding the reduced efficacy in some human-infective helminths. Understanding the mechanisms by which resistance evolves is the focus of increasing interest; robust genetic analysis of helminths is challenging, and although many candidate genes have been proposed, the genetic basis of resistance remains poorly resolved. RESULTS: Here, we present a genome-wide analysis of two genetic crosses between ivermectin resistant and sensitive isolates of the parasitic nematode Haemonchus contortus, an economically important gastrointestinal parasite of small ruminants and a model for anthelmintic research. Whole genome sequencing of parental populations, and key stages throughout the crosses, identified extensive genomic diversity that differentiates populations, but after backcrossing and selection, a single genomic quantitative trait locus (QTL) localised on chromosome V was revealed to be associated with ivermectin resistance. This QTL was common between the two geographically and genetically divergent resistant populations and did not include any leading candidate genes, suggestive of a previously uncharacterised mechanism and/or driver of resistance. Despite limited resolution due to low recombination in this region, population genetic analyses and novel evolutionary models supported strong selection at this QTL, driven by at least partial dominance of the resistant allele, and that large resistance-associated haplotype blocks were enriched in response to selection. CONCLUSIONS: We have described the genetic architecture and mode of ivermectin selection, revealing a major genomic locus associated with ivermectin resistance, the most conclusive evidence to date in any parasitic nematode. This study highlights a novel genome-wide approach to the analysis of a genetic cross in non-model organisms with extreme genetic diversity, and the importance of a high-quality reference genome in interpreting the signals of selection so identified.


Asunto(s)
Resistencia a Medicamentos , Evolución Molecular , Haemonchus/efectos de los fármacos , Haemonchus/genética , Ivermectina/farmacología , Metagenómica , Sitios de Carácter Cuantitativo , Animales , ADN de Helmintos , Variación Genética , Insecticidas/farmacología
20.
BMC Med ; 17(1): 60, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30862316

RESUMEN

BACKGROUND: There are over 200 million reported cases of malaria each year, and most children living in endemic areas will experience multiple episodes of clinical disease before puberty. We set out to understand how frequent clinical malaria, which elicits a strong inflammatory response, affects the immune system and whether these modifications are observable in the absence of detectable parasitaemia. METHODS: We used a multi-dimensional approach comprising whole blood transcriptomic, cellular and plasma cytokine analyses on a cohort of children living with endemic malaria, but uninfected at sampling, who had been under active surveillance for malaria for 8 years. Children were categorised into two groups depending on the cumulative number of episodes experienced: high (≥ 8) or low (< 5). RESULTS: We observe that multiple episodes of malaria are associated with modification of the immune system. Children who had experienced a large number of episodes demonstrated upregulation of interferon-inducible genes, a clear increase in circulating levels of the immunoregulatory cytokine IL-10 and enhanced activation of neutrophils, B cells and CD8+ T cells. CONCLUSION: Transcriptomic analysis together with cytokine and immune cell profiling of peripheral blood can robustly detect immune differences between children with different numbers of prior malaria episodes. Multiple episodes of malaria are associated with modification of the immune system in children. Such immune modifications may have implications for the initiation of subsequent immune responses and the induction of vaccine-mediated protection.


Asunto(s)
Enfermedades del Sistema Inmune/inmunología , Malaria/inmunología , Niño , Preescolar , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA