Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 31(9): 1539-1551.e6, 2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37708854

RESUMO

Malaria remains one of the most devastating infectious diseases. Reverse genetic screens offer a powerful approach to identify genes and molecular processes governing malaria parasite biology. However, the complex regulation of gene expression and genotype-phenotype associations in the mosquito vector, along with sexual reproduction, have hindered the development of screens in this critical part of the parasite life cycle. To address this, we developed a genetic approach in the rodent parasite Plasmodium berghei that, in combination with barcode sequencing, circumvents the fertilization roadblock and enables screening for gametocyte-expressed genes required for parasite infection of the mosquito Anopheles coluzzii. Our results confirm previous findings, validating our approach for scaling up, and identify genes necessary for mosquito midgut infection, oocyst development, and salivary gland infection. These findings can aid efforts to study malaria transmission biology and to develop interventions for controlling disease transmission.


Assuntos
Anopheles , Esporozoítos , Animais , Esporozoítos/genética , Mosquitos Vetores/genética , Plasmodium berghei/genética , Anopheles/genética
2.
Microbiol Spectr ; : e0382022, 2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37698406

RESUMO

Our overall understanding of the developmental biology of malaria parasites has been greatly enhanced by recent advances in transcriptomic analysis. However, most of these investigations rely on laboratory strains (LS) that were adapted into in vitro culture many years ago, and the transcriptomes of clinical isolates (CI) circulating in human populations have not been assessed. In this study, RNA-seq was used to compare the global transcriptome of mid-stage gametocytes derived from three short-term cultured CI, with gametocytes derived from the NF54 reference laboratory strain. The core transcriptome appeared to be consistent between CI- and LS-derived gametocyte preparations, but some important differences were also observed. A majority of gametocyte-specific genes (43/53) appear to have relatively higher expression in CI-derived gametocytes than in LS-derived gametocytes, but a K-means clustering analysis showed that genes involved in flagellum- and microtubule-based processes (movement/motility) were more abundant in both groups, albeit with some differences between them. In addition, gametocytes from one CI described as CI group II gametocytes (CI:GGII) showed gene expression variation in the form of reduced gametocyte-specific gene expression compared to the other two CI-derived gametocytes (CI gametocyte group I, CI:GGI), although the mixed developmental stages used in our study is a potential confounder, only partially mitigated by the inclusion of multiple replicates for each CI. Overall, our study suggests that there may be subtle differences in the gene expression profiles of mid-stage gametocytes from CI relative to the NF54 reference strain of Plasmodium falciparum. Thus, it is necessary to deploy gametocyte-producing clinical parasite isolates to fully understand the diversity of gene expression strategies that may occur during the sequestered development of parasite sexual stages. IMPORTANCE Maturing gametocytes of Plasmodium falciparum are known to sequester away from peripheral circulation into the bone marrow until they are mature. Blocking gametocyte sequestration can prevent malaria transmission from humans to mosquitoes, but most studies aim to understand gametocyte development utilizing long-term adapted laboratory lines instead of clinical isolates. This is a particular issue for our understanding of the sexual stages, which are known to decrease rapidly during adaptation to long-term culture, meaning that many LS are unable to produce transmissible gametocytes. Using RNA-seq, we investigated the global transcriptome of mid-stage gametocytes derived from three clinical isolates and a reference strain (NF54). This identified important differences in gene expression profiles between immature gametocytes of CI and the NF54 reference strain of P. falciparum, suggesting increased investment in gametocytogenesis in clinical isolates. Our transcriptomic data highlight the use of clinical isolates in studying the morphological, cellular features and molecular biology of gametocytes.

3.
Trends Parasitol ; 39(5): 317-318, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36964075

RESUMO

Genome-wide screening in apicomplexan species has transformed our understanding of these parasitic protozoa. Kimmel et al. report a 'knock sideways' system and provide a powerful use case for its feasibility in a gene-by-gene screening in Plasmodium falciparum. Carefully deployed, a novel toolkit helps to dissect the biological uniqueness of an important parasite.


Assuntos
Malária , Parasitos , Animais , Humanos , Plasmodium falciparum/genética , Malária/parasitologia
4.
Commun Biol ; 6(1): 205, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36810637

RESUMO

Eukaryotes have canonical pathways for responding to amino acid (AA) availability. Under AA-limiting conditions, the TOR complex is repressed, whereas the sensor kinase GCN2 is activated. While these pathways have been highly conserved throughout evolution, malaria parasites are a rare exception. Despite auxotrophic for most AA, Plasmodium does not have either a TOR complex nor the GCN2-downstream transcription factors. While Ile starvation has been shown to trigger eIF2α phosphorylation and a hibernation-like response, the overall mechanisms mediating detection and response to AA fluctuation in the absence of such pathways has remained elusive. Here we show that Plasmodium parasites rely on an efficient sensing pathway to respond to AA fluctuations. A phenotypic screen of kinase knockout mutant parasites identified nek4, eIK1 and eIK2-the last two clustering with the eukaryotic eIF2α kinases-as critical for Plasmodium to sense and respond to distinct AA-limiting conditions. Such AA-sensing pathway is temporally regulated at distinct life cycle stages, allowing parasites to actively fine-tune replication and development in response to AA availability. Collectively, our data disclose a set of heterogeneous responses to AA depletion in malaria parasites, mediated by a complex mechanism that is critical for modulating parasite growth and survival.


Assuntos
Aminoácidos , Plasmodium , Aminoácidos/deficiência , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo , Fosforilação , Fosfotransferases/metabolismo , Plasmodium/enzimologia , Plasmodium/genética
5.
Cell Host Microbe ; 31(2): 305-319.e10, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36634679

RESUMO

Malaria transmission to mosquitoes requires a developmental switch in asexually dividing blood-stage parasites to sexual reproduction. In Plasmodium berghei, the transcription factor AP2-G is required and sufficient for this switch, but how a particular sex is determined in a haploid parasite remains unknown. Using a global screen of barcoded mutants, we here identify genes essential for the formation of either male or female sexual forms and validate their importance for transmission. High-resolution single-cell transcriptomics of ten mutant parasites portrays the developmental bifurcation and reveals a regulatory cascade of putative gene functions in the determination and subsequent differentiation of each sex. A male-determining gene with a LOTUS/OST-HTH domain as well as the protein interactors of a female-determining zinc-finger protein indicate that germ-granule-like ribonucleoprotein complexes complement transcriptional processes in the regulation of both male and female development of a malaria parasite.


Assuntos
Culicidae , Malária , Parasitos , Animais , Feminino , Masculino , Parasitos/metabolismo , Malária/parasitologia , Plasmodium berghei/genética , Desenvolvimento Sexual/genética , Culicidae/parasitologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
6.
Curr Opin Microbiol ; 63: 259-266, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34461385

RESUMO

Genome scale metabolic models (GEMs) offer a powerful means of integrating genome and biochemical information on an organism to make testable predictions of metabolic functions at different conditions and to systematically predict essential genes that may be targeted by drugs. This review describes how Plasmodium GEMs have become increasingly more accurate through the integration of omics and experimental genetic data. We also discuss how GEMs contribute to our increasing understanding of how Plasmodium metabolism is reprogrammed between life cycle stages.


Assuntos
Modelos Biológicos , Plasmodium , Animais , Genoma/genética , Estágios do Ciclo de Vida/genética , Fígado , Redes e Vias Metabólicas , Plasmodium/genética
7.
mSphere ; 6(3)2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34011691

RESUMO

Malaria is a mosquito-borne disease caused by apicomplexan parasites of the genus Plasmodium. Completion of the parasite's life cycle depends on the transmission of sexual stages, the gametocytes, from an infected human host to the mosquito vector. Sexual commitment occurs in only a small fraction of asexual blood-stage parasites and is initiated by external cues. The gametocyte development protein 1 (GDV1) has been described as a key facilitator to trigger sexual commitment. GDV1 interacts with the silencing factor heterochromatin protein 1 (HP1), leading to its dissociation from heterochromatic DNA at the genomic locus encoding AP2-G, the master transcription factor of gametocytogenesis. How this process is regulated is not known. In this study, we have addressed the role of protein kinases implicated in gametocyte development. From a pool of available protein kinase knockout (KO) lines, we identified two kinase knockout lines which fail to produce gametocytes. However, independent genetic verification revealed that both kinases are not required for gametocytogenesis but that both lines harbor the same mutation that leads to a truncation in the extreme C terminus of GDV1. Introduction of the identified nonsense mutation into the genome of wild-type parasite lines replicates the observed phenotype. Using a GDV1 overexpression line, we show that the truncation in the GDV1 C terminus does not interfere with the nuclear import of GDV1 or its interaction with HP1 in vitro but appears to be important to sustain GDV1 protein levels and thereby sexual commitment.IMPORTANCE Transmission of malaria-causing Plasmodium species by mosquitos requires the parasite to change from a continuously growing asexual parasite form growing in the blood to a sexually differentiated form, the gametocyte. Only a small subset of asexual parasites differentiates into gametocytes that are taken up by the mosquito. Transmission represents a bottleneck in the life cycle of the parasite, so a molecular understanding of the events that lead to stage conversion may identify novel intervention points. Here, we screened a subset of kinases we hypothesized to play a role in this process. While we did not identify kinases required for sexual conversion, we identified a mutation in the C terminus of the gametocyte development 1 protein (GDV1), which abrogates sexual development. The mutation destabilizes the protein but not its interaction with its cognate binding partner HP1. This suggests an important role for the GDV1 C terminus beyond trafficking and protein stability.


Assuntos
Aminoácidos/genética , Gametogênese/genética , Estágios do Ciclo de Vida/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Regulação da Expressão Gênica , Humanos , Malária Falciparum , Plasmodium falciparum/química , Proteínas de Protozoários/química , Análise de Sequência de RNA , Diferenciação Sexual/genética
8.
Sci Rep ; 11(1): 1888, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479319

RESUMO

New antimalarial therapeutics are needed to ensure that malaria cases continue to be driven down, as both emerging parasite resistance to frontline chemotherapies and mosquito resistance to current insecticides threaten control programmes. Plasmodium, the apicomplexan parasite responsible for malaria, causes disease pathology through repeated cycles of invasion and replication within host erythrocytes (the asexual cycle). Antimalarial drugs primarily target this cycle, seeking to reduce parasite burden within the host as fast as possible and to supress recrudescence for as long as possible. Intense phenotypic drug screening efforts have identified a number of promising new antimalarial molecules. Particularly important is the identification of compounds with new modes of action within the parasite to combat existing drug resistance and suitable for formulation of efficacious combination therapies. Here we detail the antimalarial properties of DDD01034957-a novel antimalarial molecule which is fast-acting and potent against drug resistant strains in vitro, shows activity in vivo, and possesses a resistance mechanism linked to the membrane transporter PfABCI3. These data support further medicinal chemistry lead-optimization of DDD01034957 as a novel antimalarial chemical class and provide new insights to further reduce in vivo metabolic clearance.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Malária/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Animais , Antimaláricos/química , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Malária/parasitologia , Camundongos , Estrutura Molecular , Plasmodium/efeitos dos fármacos , Plasmodium/parasitologia , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/parasitologia , Plasmodium falciparum/fisiologia , Especificidade da Espécie
9.
Mol Cell Proteomics ; 20: 100038, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33515807

RESUMO

Sporozoites are a motile form of malaria-causing Plasmodium falciparum parasites that migrate from the site of transmission in the dermis through the bloodstream to invade hepatocytes. Sporozoites interact with many cells within the host, but the molecular identity of these interactions and their role in the pathology of malaria is poorly understood. Parasite proteins that are secreted and embedded within membranes are known to be important for these interactions, but our understanding of how they interact with each other to form functional complexes is largely unknown. Here, we compile a library of recombinant proteins representing the repertoire of cell surface and secreted proteins from the P. falciparum sporozoite and use an assay designed to detect extracellular interactions to systematically identify complexes. We identify three protein complexes including an interaction between two components of the p24 complex that is involved in the trafficking of glycosylphosphatidylinositol-anchored proteins through the secretory pathway. Plasmodium parasites lacking either gene are strongly inhibited in the establishment of liver-stage infections. These findings reveal an important role for the p24 complex in malaria pathogenesis and show that the library of recombinant proteins represents a valuable resource to investigate P. falciparum sporozoite biology.


Assuntos
Interações Hospedeiro-Parasita , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Esporozoítos/metabolismo , Animais , Feminino , Malária/parasitologia , Camundongos Endogâmicos BALB C , Organismos Geneticamente Modificados , Fenótipo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/fisiologia , Mapas de Interação de Proteínas , Proteínas de Protozoários/genética , Proteínas Recombinantes/metabolismo , Esporozoítos/fisiologia
10.
Science ; 369(6507): 1128-1132, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32855340

RESUMO

Hemocytes limit the capacity of mosquitoes to transmit human pathogens. Here we profile the transcriptomes of 8506 hemocytes of Anopheles gambiae and Aedes aegypti mosquito vectors. Our data reveal the functional diversity of hemocytes, with different subtypes of granulocytes expressing distinct and evolutionarily conserved subsets of effector genes. A previously unidentified cell type in An. gambiae, which we term "megacyte," is defined by a specific transmembrane protein marker (TM7318) and high expression of lipopolysaccharide-induced tumor necrosis factor-α transcription factor 3 (LL3). Knockdown experiments indicate that LL3 mediates hemocyte differentiation during immune priming. We identify and validate two main hemocyte lineages and find evidence of proliferating granulocyte populations. This atlas of medically relevant invertebrate immune cells at single-cell resolution identifies cellular events that underpin mosquito immunity to malaria infection.


Assuntos
Aedes/imunologia , Anopheles/imunologia , Hemócitos/imunologia , Imunidade Celular , Malária/transmissão , Mosquitos Vetores/imunologia , Aedes/genética , Animais , Anopheles/genética , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Granulócitos/imunologia , Hemócitos/metabolismo , Malária/imunologia , Malária/parasitologia , Camundongos , Mosquitos Vetores/genética , RNA-Seq , Análise de Célula Única
11.
Sci Rep ; 10(1): 13630, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32788672

RESUMO

Cerebral malaria (CM) is the deadliest form of severe Plasmodium infections. Currently, we have limited understanding of the mechanisms by which Plasmodium parasites induce CM. The mouse model of CM, experimental CM (ECM), induced by infection with the rodent parasite, Plasmodium berghei ANKA (PbANKA) has been extensively used to study the pathophysiology of CM. Recent genomic analyses revealed that the coding regions of PbANKA and the closely related Plasmodium berghei NK65 (PbNK65), that does not cause ECM, differ in only 21 single nucleotide polymorphysims (SNPs). Thus, the SNP-containing genes might contribute to the pathogenesis of ECM. Although the majority of these SNPs are located in genes of unknown function, one SNP is located in the DNA binding site of a member of the Plasmodium ApiAP2 transcription factor family, that we recently showed functions as a virulence factor alternating the host's immune response to the parasite. Here, we investigated the impact of this SNP on the development of ECM. Our results using CRISPR-Cas9 engineered parasites indicate that despite its immune modulatory function, the SNP is neither necessary nor sufficient to induce ECM and thus cannot account for parasite strain-specific differences in ECM phenotypes.


Assuntos
Sistemas CRISPR-Cas/genética , Matriz Extracelular/parasitologia , Malária Cerebral/parasitologia , Plasmodium berghei/genética , Polimorfismo de Nucleotídeo Único , Proteínas de Protozoários/genética , Fatores de Virulência/genética , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/fisiologia , Proteínas de Protozoários/antagonistas & inibidores , Fatores de Virulência/antagonistas & inibidores
12.
Nat Commun ; 11(1): 4015, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32782246

RESUMO

Intracellular pathogens mobilize host signaling pathways of their host cell to promote their own survival. Evidence is emerging that signal transduction elements are activated in a-nucleated erythrocytes in response to infection with malaria parasites, but the extent of this phenomenon remains unknown. Here, we fill this knowledge gap through a comprehensive and dynamic assessment of host erythrocyte signaling during infection with Plasmodium falciparum. We used arrays of 878 antibodies directed against human signaling proteins to interrogate the activation status of host erythrocyte phospho-signaling pathways at three blood stages of parasite asexual development. This analysis reveals a dynamic modulation of many host signalling proteins across parasite development. Here we focus on the hepatocyte growth factor receptor (c-MET) and the MAP kinase pathway component B-Raf, providing a proof of concept that human signaling kinases identified as activated by malaria infection represent attractive targets for antimalarial intervention.


Assuntos
Antimaláricos/farmacologia , Eritrócitos/metabolismo , Plasmodium falciparum/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Humanos , Concentração Inibidora 50 , Estágios do Ciclo de Vida/efeitos dos fármacos , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Fosforilação/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Análise Serial de Proteínas , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Wellcome Open Res ; 5: 71, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32500098

RESUMO

The rodent parasite Plasmodium chabaudi is an important in vivo model of malaria. The ability to produce chronic infections makes it particularly useful for investigating the development of anti- Plasmodium immunity, as well as features associated with parasite virulence during both the acute and chronic phases of infection. P. chabaudi also undergoes asexual maturation (schizogony) and erythrocyte invasion in culture, so offers an experimentally-amenable in vivo to in vitro model for studying gene function and drug activity during parasite replication. To extend the usefulness of this model, we have further optimised transfection protocols and plasmids for P. chabaudi and generated stable, fluorescent lines that are free from drug-selectable marker genes. These mother-lines show the same infection dynamics as wild-type parasites throughout the lifecycle in mice and mosquitoes; furthermore, their virulence can be increased by serial blood passage and reset by mosquito transmission. We have also adapted the large-insert, linear PlasmoGEM vectors that have revolutionised the scale of experimental genetics in another rodent malaria parasite and used these to generate barcoded P. chabaudi gene-deletion and -tagging vectors for transfection in our fluorescent P. chabaudi mother-lines. This produces a tool-kit of P. chabaudi lines, vectors and transfection approaches that will be of broad utility to the research community.

14.
Stem Cell Reports ; 14(6): 1123-1134, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32442532

RESUMO

Asymptomatic and obligatory liver stage (LS) infection of Plasmodium parasites presents an attractive target for antimalarial vaccine and drug development. Lack of robust cellular models to study LS infection has hindered the discovery and validation of host genes essential for intrahepatic parasite development. Here, we present a chemically differentiated mouse embryonic stem cell (ESC)-based LS model, which supports complete development of Plasmodium berghei exoerythrocytic forms (EEFs) and can be used to define new host-parasite interactions. Using our model, we established that host Pnpla2, coding for adipose triglyceride lipase, is dispensable for P. berghei EEF development. In addition, we also evaluated in-vitro-differentiated human hepatocyte-like cells (iHLCs) to study LS of P. berghei and found it to be a sub-optimal infection model. Overall, our results present a new mouse ESC-based P. berghei LS infection model that can be utilized to study the impact of host genetic variation on parasite development.


Assuntos
Diferenciação Celular , Hepatócitos/parasitologia , Interações Hospedeiro-Parasita , Malária/parasitologia , Células-Tronco Embrionárias Murinas/citologia , Plasmodium berghei/patogenicidade , Animais , Linhagem Celular , Células Cultivadas , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Lipase/genética , Lipase/metabolismo , Malária/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo
15.
Sci Adv ; 6(6): eaaw6957, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32076635

RESUMO

The acquisition of malaria immunity is both remarkably slow and unpredictable. At present, we know little about the malaria parasite genes that influence the host's ability to mount a protective immune response. Here, we show that a single-nucleotide polymorphism (SNP) resulting in a single amino acid change (S to F) in an ApiAP2 transcription factor in the rodent malaria parasite Plasmodium berghei (Pb) NK65 allowed infected mice to mount a T helper cell 1 (TH1)-type immune response that controlled subsequent infections. As compared to PbNK65S, PbNK65F parasites differentially expressed 46 genes, most of which are predicted to play roles in immune evasion. PbNK65F infections resulted in an early interferon-γ response and a later expansion of germinal centers, resulting in high levels of infected red blood cell-specific TH1-type immunoglobulin G2b (IgG2b) and IgG2c antibodies. Thus, the Pb ApiAP2 transcription factor functions as a critical parasite virulence factor in malaria infections.


Assuntos
Culicidae/parasitologia , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Imunidade , Malária/parasitologia , Plasmodium berghei/genética , Polimorfismo de Nucleotídeo Único , Fator de Transcrição AP-2/genética , Imunidade Adaptativa , Animais , Proteínas de Ligação a DNA , Plasmodium berghei/metabolismo , Domínios e Motivos de Interação entre Proteínas , Células Th1/imunologia , Células Th1/metabolismo , Fator de Transcrição AP-2/química , Fator de Transcrição AP-2/metabolismo
16.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31730853

RESUMO

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Assuntos
Genoma de Protozoário , Estágios do Ciclo de Vida/genética , Fígado/metabolismo , Fígado/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/genética , Alelos , Amino Açúcares/biossíntese , Animais , Culicidae/parasitologia , Eritrócitos/parasitologia , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/metabolismo , Técnicas de Inativação de Genes , Genótipo , Modelos Biológicos , Mutação/genética , Parasitos/genética , Parasitos/crescimento & desenvolvimento , Fenótipo , Plasmodium berghei/metabolismo , Ploidias , Reprodução
17.
Cell Rep ; 28(6): 1635-1647.e5, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31390575

RESUMO

Malaria represents a major global health issue, and the identification of new intervention targets remains an urgent priority. This search is hampered by more than one-third of the genes of malaria-causing Plasmodium parasites being uncharacterized. We report a large-scale protein interaction network in Plasmodium schizonts, generated by combining blue native-polyacrylamide electrophoresis with quantitative mass spectrometry and machine learning. This integrative approach, spanning 3 species, identifies >20,000 putative protein interactions, organized into 600 protein clusters. We validate selected interactions, assigning functions in chromatin regulation to previously unannotated proteins and suggesting a role for an EELM2 domain-containing protein and a putative microrchidia protein as mechanistic links between AP2-domain transcription factors and epigenetic regulation. Our interactome represents a high-confidence map of the native organization of core cellular processes in Plasmodium parasites. The network reveals putative functions for uncharacterized proteins, provides mechanistic and structural insight, and uncovers potential alternative therapeutic targets.


Assuntos
Plasmodium/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Eletroforese em Gel de Poliacrilamida , Feminino , Camundongos , Plasmodium/genética , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Mapas de Interação de Proteínas , Ratos , Especificidade da Espécie , Espectrometria de Massas em Tandem
18.
Science ; 365(6455)2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31439762

RESUMO

Malaria parasites adopt a remarkable variety of morphological life stages as they transition through multiple mammalian host and mosquito vector environments. We profiled the single-cell transcriptomes of thousands of individual parasites, deriving the first high-resolution transcriptional atlas of the entire Plasmodium berghei life cycle. We then used our atlas to precisely define developmental stages of single cells from three different human malaria parasite species, including parasites isolated directly from infected individuals. The Malaria Cell Atlas provides both a comprehensive view of gene usage in a eukaryotic parasite and an open-access reference dataset for the study of malaria parasites.


Assuntos
Atlas como Assunto , Genes de Protozoários/fisiologia , Estágios do Ciclo de Vida/genética , Malária/parasitologia , Plasmodium berghei/genética , Plasmodium berghei/fisiologia , Transcriptoma , Animais , Anopheles/parasitologia , Células HeLa , Humanos , Plasmodium berghei/isolamento & purificação , Análise de Célula Única
19.
Cell Host Microbe ; 24(6): 754-755, 2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30543773

RESUMO

The importance of guanylyl-cyclases (GCs) in apicomplexa has remained elusive due to the large size of the genes. Two recent studies, including Brown and Sibley, 2018 in this issue of Cell Host & Microbe, make elegant use of genome editing with CRISPR-Cas9 to characterize roles of GCs in Toxoplasma and Plasmodium.


Assuntos
Plasmodium/genética , Toxoplasma/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes
20.
Nat Commun ; 9(1): 4248, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30315162

RESUMO

In malaria parasites, evolution of parasitism has been linked to functional optimisation. Despite this optimisation, most members of a calcium-dependent protein kinase (CDPK) family show genetic redundancy during erythrocytic proliferation. To identify relationships between phospho-signalling pathways, we here screen 294 genetic interactions among protein kinases in Plasmodium berghei. This reveals a synthetic negative interaction between a hypomorphic allele of the protein kinase G (PKG) and CDPK4 to control erythrocyte invasion which is conserved in P. falciparum. CDPK4 becomes critical when PKG-dependent calcium signals are attenuated to phosphorylate proteins important for the stability of the inner membrane complex, which serves as an anchor for the acto-myosin motor required for motility and invasion. Finally, we show that multiple kinases functionally complement CDPK4 during erythrocytic proliferation and transmission to the mosquito. This study reveals how CDPKs are wired within a stage-transcending signalling network to control motility and host cell invasion in malaria parasites.


Assuntos
Epistasia Genética/genética , Plasmodium berghei/metabolismo , Plasmodium berghei/patogenicidade , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas Quinases/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Cálcio/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Feminino , Malária Falciparum/parasitologia , Masculino , Camundongos , Proteínas Quinases/genética , Proteínas de Protozoários/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA