Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Microbiol Spectr ; 11(1): e0304922, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36515553

RESUMO

The survival of malaria parasites in the changing human blood environment largely depends on their ability to alter gene expression by epigenetic mechanisms. The active state of Plasmodium falciparum clonally variant genes (CVGs) is associated with euchromatin characterized by the histone mark H3K9ac, whereas the silenced state is characterized by H3K9me3-based heterochromatin. Expression switches are linked to euchromatin-heterochromatin transitions, but these transitions have not been characterized for the majority of CVGs. To define the heterochromatin distribution patterns associated with the alternative transcriptional states of CVGs, we compared H3K9me3 occupancy at a genome-wide level among several parasite subclones of the same genetic background that differed in the transcriptional state of many CVGs. We found that de novo heterochromatin formation or the complete disruption of a heterochromatin domain is a relatively rare event, and for the majority of CVGs, expression switches can be explained by the expansion or retraction of heterochromatin domains. We identified different modalities of heterochromatin changes linked to transcriptional differences, but despite this complexity, heterochromatin distribution patterns generally enable the prediction of the transcriptional state of specific CVGs. We also found that in some subclones, several var genes were simultaneously in an active state. Furthermore, the heterochromatin levels in the putative regulatory region of the gdv1 antisense noncoding RNA, a regulator of sexual commitment, varied between parasite lines with different sexual conversion rates. IMPORTANCE The malaria parasite P. falciparum is responsible for more than half a million deaths every year. P. falciparum clonally variant genes (CVGs) mediate fundamental host-parasite interactions and play a key role in parasite adaptation to fluctuations in the conditions of the human host. The expression of CVGs is regulated at the epigenetic level by changes in the distribution of a type of chromatin called heterochromatin. Here, we describe at a genome-wide level the changes in the heterochromatin distribution associated with the different transcriptional states of CVGs. Our results also reveal a likely role for heterochromatin at a particular locus in determining the parasite investment in transmission to mosquitoes. Additionally, this data set will enable the prediction of the transcriptional state of CVGs from epigenomic data, which is important for the study of parasite adaptation to the conditions of the host in natural malaria infections.


Assuntos
Malária Falciparum , Plasmodium falciparum , Animais , Humanos , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Eucromatina/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Malária Falciparum/parasitologia , Regulação da Expressão Gênica
2.
Artigo em Inglês | MEDLINE | ID: mdl-30782998

RESUMO

During the intraerythrocytic asexual cycle malaria parasites acquire nutrients and other solutes through a broad selectivity channel localized at the membrane of the infected erythrocyte termed the plasmodial surface anion channel (PSAC). The protein product of the Plasmodium falciparum clonally variant clag3.1 and clag3.2 genes determines PSAC activity. Switches in the expression of clag3 genes, which are regulated by epigenetic mechanisms, are associated with changes in PSAC-dependent permeability that can result in resistance to compounds toxic for the parasite, such as blasticidin S. Here, we investigated whether other antimalarial drugs require CLAG3 to reach their intracellular target and consequently are prone to parasite resistance by epigenetic mechanisms. We found that the bis-thiazolium salts T3 (also known as albitiazolium) and T16 require the product of clag3 genes to enter infected erythrocytes. P. falciparum populations can develop resistance to these compounds via the selection of parasites with dramatically reduced expression of both genes. However, other compounds previously demonstrated or predicted to enter infected erythrocytes through transport pathways absent from noninfected erythrocytes, such as fosmidomycin, doxycycline, azithromycin, lumefantrine, or pentamidine, do not require expression of clag3 genes for their antimalarial activity. This suggests that they use alternative CLAG3-independent routes to access parasites. Our results demonstrate that P. falciparum can develop resistance to diverse antimalarial compounds by epigenetic changes in the expression of clag3 genes. This is of concern for drug development efforts because drug resistance by epigenetic mechanisms can arise quickly, even during the course of a single infection.


Assuntos
Antimaláricos/uso terapêutico , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Epigênese Genética , Malária Falciparum/metabolismo , Plasmodium falciparum/genética , Proteínas de Protozoários/genética
3.
Nat Microbiol ; 4(1): 144-154, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30478286

RESUMO

Human to vector transmission of malaria requires that some blood-stage parasites abandon asexual growth and convert into non-replicating sexual forms called gametocytes. The initial steps of gametocytogenesis remain largely uncharacterized. Here, we study this part of the malaria life cycle in Plasmodium falciparum using PfAP2-G, the master regulator of sexual conversion, as a marker of commitment. We demonstrate the existence of PfAP2-G-positive sexually committed parasite stages that precede the previously known committed schizont stage. We also found that sexual conversion can occur by two different routes: the previously described route in which PfAP2-G-expressing parasites complete a replicative cycle as committed forms before converting into gametocytes upon re-invasion, or a direct route with conversion within the same cycle as initial PfAP2-G expression. The latter route is linked to early PfAP2-G expression in ring stages. Reanalysis of published single-cell RNA-sequencing (RNA-seq) data confirmed the presence of both routes. Consistent with these results, using plaque assays we observed that, in contrast to the prevailing model, many schizonts produced mixed plaques containing both asexual parasites and gametocytes. Altogether, our results reveal unexpected features of the initial steps of sexual development and extend the current view of this part of the malaria life cycle.


Assuntos
Estágios do Ciclo de Vida/fisiologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Desenvolvimento Sexual/fisiologia , Sequência de Bases , Eritrócitos/parasitologia , Humanos , Malária Falciparum/patologia , Esquizontes/metabolismo , Análise de Sequência de RNA
4.
J Infect Dis ; 215(6): 938-945, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28419281

RESUMO

Background: Many genes of the malaria parasite Plasmodium falciparum show clonally variant expression regulated at the epigenetic level. These genes participate in fundamental host-parasite interactions and contribute to adaptive processes. However, little is known about their expression patterns during human infections. A peculiar case of clonally variant genes are the 2 nearly identical clag3 genes, clag3.1 and clag3.2, which mediate nutrient uptake and are linked to resistance to some toxic compounds. Methods: We developed a procedure to characterize the expression of clag3 genes in naturally infected patients and in experimentally infected human volunteers. Results: We provide the first description of clag3 expression during human infections, which revealed mutually exclusive expression and identified the gene predominantly expressed. Adaptation to culture conditions or selection with a toxic compound resulted in isolate-dependent changes in clag3 expression. We also found that clag3 expression patterns were reset during transmission stages. Conclusions: Different environment conditions select for parasites with different clag3 expression patterns, implying functional differences between the proteins encoded. The epigenetic memory is likely erased before parasites start infection of a new human host. Altogether, our findings support the idea that clonally variant genes facilitate the adaptation of parasite populations to changing conditions through bet-hedging strategies.


Assuntos
Malária Falciparum/genética , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Adulto , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Criança , Estudos de Coortes , Resistência a Medicamentos , Epigênese Genética , Gâmbia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genes de Protozoários , Interações Hospedeiro-Parasita , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/transmissão , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/sangue
5.
PLoS One ; 11(10): e0165358, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27780272

RESUMO

The growth phenotype of asexual blood stage malaria parasites can influence their virulence and also their ability to survive and achieve transmission to the next host, but there are few methods available to characterise parasite growth parameters in detail. We developed a new assay to measure growth rates at different starting parasitaemias in a 96-well format and applied it to characterise the growth of Plasmodium falciparum lines 3D7-A and 3D7-B, previously shown to have different invasion rates and to use different invasion pathways. Using this simple and accurate assay we found that 3D7-B is more sensitive to high initial parasitaemia than 3D7-A. This result indicates that different parasite lines show variation in their levels of density-dependent growth inhibition. We also developed a new assay to compare the duration of the asexual blood cycle between different parasite lines. The assay is based on the tight synchronisation of cultures to a 1 h parasite age window and the subsequent monitoring of schizont bursting and formation of new rings by flow cytometry. Using this assay we observed differences in the duration of the asexual blood cycle between parasite lines 3D7 and HB3. These two new assays will be useful to characterise variation in growth-related parameters and to identify growth phenotypes associated with the targeted deletion of specific genes or with particular genomic, transcriptomic or proteomic patterns. Furthermore, the identification of density-dependent growth inhibition as an intrinsic parasite property that varies between parasite lines expands the repertoire of measurable growth-related phenotypic traits that have the potential to influence the outcome of a malarial blood infection.


Assuntos
Parasitemia/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Eritrócitos/parasitologia , Citometria de Fluxo , Genômica , Humanos , Estágios do Ciclo de Vida , Malária Falciparum/diagnóstico , Malária Falciparum/parasitologia , Merozoítos/fisiologia , Análise em Microsséries , Parasitemia/diagnóstico , Fenótipo , Plasmodium falciparum/fisiologia , Proteômica , Esquizontes/crescimento & desenvolvimento
6.
Nucleic Acids Res ; 43(17): 8243-57, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26202963

RESUMO

The product of the Plasmodium falciparum genes clag3.1 and clag3.2 plays a fundamental role in malaria parasite biology by determining solute transport into infected erythrocytes. Expression of the two clag3 genes is mutually exclusive, such that a single parasite expresses only one of the two genes at a time. Here we investigated the properties and mechanisms of clag3 mutual exclusion using transgenic parasite lines with extra copies of clag3 promoters located either in stable episomes or integrated in the parasite genome. We found that the additional clag3 promoters in these transgenic lines are silenced by default, but under strong selective pressure parasites with more than one clag3 promoter simultaneously active are observed, demonstrating that clag3 mutual exclusion is strongly favored but it is not strict. We show that silencing of clag3 genes is associated with the repressive histone mark H3K9me3 even in parasites with unusual clag3 expression patterns, and we provide direct evidence for heterochromatin spreading in P. falciparum. We also found that expression of a neighbor ncRNA correlates with clag3.1 expression. Altogether, our results reveal a scenario where fitness costs and non-deterministic molecular processes that favor mutual exclusion shape the expression patterns of this important gene family.


Assuntos
Regulação da Expressão Gênica , Genes de Protozoários , Plasmodium falciparum/genética , Inativação Gênica , Genes Reporter , Heterocromatina/metabolismo , Histonas/metabolismo , Família Multigênica , Plasmídeos , Plasmodium falciparum/metabolismo , Regiões Promotoras Genéticas , RNA não Traduzido/metabolismo , Ativação Transcricional
7.
Nature ; 507(7491): 248-52, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24572369

RESUMO

The life cycles of many parasites involve transitions between disparate host species, requiring these parasites to go through multiple developmental stages adapted to each of these specialized niches. Transmission of malaria parasites (Plasmodium spp.) from humans to the mosquito vector requires differentiation from asexual stages replicating within red blood cells into non-dividing male and female gametocytes. Although gametocytes were first described in 1880, our understanding of the molecular mechanisms involved in commitment to gametocyte formation is extremely limited, and disrupting this critical developmental transition remains a long-standing goal. Here we show that expression levels of the DNA-binding protein PfAP2-G correlate strongly with levels of gametocyte formation. Using independent forward and reverse genetics approaches, we demonstrate that PfAP2-G function is essential for parasite sexual differentiation. By combining genome-wide PfAP2-G cognate motif occurrence with global transcriptional changes resulting from PfAP2-G ablation, we identify early gametocyte genes as probable targets of PfAP2-G and show that their regulation by PfAP2-G is critical for their wild-type level expression. In the asexual blood-stage parasites pfap2-g appears to be among a set of epigenetically silenced loci prone to spontaneous activation. Stochastic activation presents a simple mechanism for a low baseline of gametocyte production. Overall, these findings identify PfAP2-G as a master regulator of sexual-stage development in malaria parasites and mark the first discovery of a transcriptional switch controlling a differentiation decision in protozoan parasites.


Assuntos
Regulação da Expressão Gênica/genética , Células Germinativas/crescimento & desenvolvimento , Malária/parasitologia , Parasitos/fisiologia , Plasmodium falciparum/genética , Desenvolvimento Sexual/genética , Transcrição Gênica/genética , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Inativação Gênica , Genes de Protozoários/genética , Genoma de Protozoário/genética , Células Germinativas/citologia , Células Germinativas/metabolismo , Masculino , Parasitos/citologia , Parasitos/genética , Plasmodium falciparum/citologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Reprodução Assexuada , Diferenciação Sexual/genética
8.
Cell Microbiol ; 15(11): 1913-23, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23819786

RESUMO

Malaria parasites induce changes in the permeability of the infected erythrocyte membrane to numerous solutes, including toxic compounds. In Plasmodium falciparum, this is mainly mediated by PSAC, a broad-selectivity channel that requires the product of parasite clag3 genes for its activity. The two paralogous clag3 genes, clag3.1 and clag3.2, can be silenced by epigenetic mechanisms and show mutually exclusive expression. Here we show that resistance to the antibiotic blasticidin S (BSD) is associated with switches in the expression of these genes that result in altered solute uptake. Low concentrations of the drug selected parasites that switched from clag3.2 to clag3.1 expression, implying that expression of one or the other clag3 gene confers different transport efficiency to PSAC for some solutes. Selection with higher BSD concentrations resulted in simultaneous silencing of both clag3 genes, which severely compromises PSAC formation as demonstrated by blocked uptake of other PSAC substrates. Changes in the expression of clag3 genes were not accompanied by large genetic rearrangements or mutations at the clag3 loci or elsewhere in the genome. These results demonstrate that malaria parasites can become resistant to toxic compounds such as drugs by epigenetic switches in the expression of genes necessary for the formation of solute channels.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos , Epigênese Genética , Regulação da Expressão Gênica , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/biossíntese , Nucleosídeos/farmacologia , Plasmodium falciparum/genética
9.
Genome Res ; 22(5): 925-38, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22415456

RESUMO

Malaria genetic variation has been extensively characterized, but the level of epigenetic plasticity remains largely unexplored. Here we provide a comprehensive characterization of transcriptional variation in the most lethal malaria parasite, Plasmodium falciparum, based on highly accurate transcriptional analysis of isogenic parasite lines grown under homogeneous conditions. This analysis revealed extensive transcriptional heterogeneity within genetically homogeneous clonal parasite populations. We show that clonally variant expression controlled at the epigenetic level is an intrinsic property of specific genes and gene families, the majority of which participate in host-parasite interactions. Intrinsic transcriptional variability is not restricted to genes involved in immune evasion, but also affects genes linked to lipid metabolism, protein folding, erythrocyte remodeling, or transcriptional regulation, among others, indicating that epigenetic variation results in both antigenic and functional variation. We observed a general association between heterochromatin marks and clonally variant expression, extending previous observations for specific genes to essentially all variantly expressed gene families. These results suggest that phenotypic variation of functionally unrelated P. falciparum gene families is mediated by a common mechanism based on reversible formation of H3K9me3-based heterochromatin. In changing environments, diversity confers fitness to a population. Our results support the idea that P. falciparum uses a bet-hedging strategy, as an alternative to directed transcriptional responses, to adapt to common fluctuations in its environment. Consistent with this idea, we found that transcriptionally different isogenic parasite lines markedly differed in their survival to heat-shock mimicking febrile episodes and adapted to periodic heat-shock with a pattern consistent with natural selection of pre-existing parasites.


Assuntos
Epigênese Genética , Genes de Protozoários , Plasmodium falciparum/genética , Transcriptoma , Adaptação Fisiológica/genética , Técnicas de Cultura , Perfilação da Expressão Gênica , Resposta ao Choque Térmico , Heterocromatina/metabolismo , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/fisiologia , Transcrição Gênica , Trofozoítos/crescimento & desenvolvimento , Trofozoítos/metabolismo , Trofozoítos/fisiologia
10.
Mol Microbiol ; 80(2): 391-406, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21306446

RESUMO

Clonally variant gene expression is a common survival strategy used by many pathogens, including the malaria parasite Plasmodium falciparum. Among the genes that show variant expression in this parasite are several members of small gene families linked to erythrocyte invasion, including the clag and eba families. The active or repressed state of these genes is clonally transmitted by epigenetic mechanisms. Here we characterized the promoters of clag3.1, clag3.2 and eba-140, and compared nuclease accessibility and post-translational histone modifications between their active and repressed states. Activity of these promoters in an episomal context is similar between parasite subclones characterized by different patterns of expression of the endogenous genes. Variant expression is controlled by the euchromatic or heterochromatic state of bistable chromatin domains. Repression is mediated by H3K9me3-based heterochromatin, whereas the active state is characterized by H3K9ac. These marks are maintained throughout the asexual blood cycle to transmit the epigenetic memory. Furthermore, eba-140 is organized in two distinct chromatin domains, probably separated by a barrier insulator located within its ORF. The 5' chromatin domain controls expression of the gene, whereas the 3' domain shares the chromatin conformation with the upstream region of the neighbouring phista family gene, which also shows variant expression.


Assuntos
Regulação para Baixo , Epigênese Genética , Eritrócitos/parasitologia , Regulação da Expressão Gênica , Heterocromatina/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Proteínas de Transporte/genética , Proteínas de Membrana , Regiões Promotoras Genéticas , Proteínas de Protozoários/genética
11.
FEMS Microbiol Lett ; 270(2): 250-4, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17343676

RESUMO

An episomal RNAi silencing construct containing the inducible cbhB promoter and a hairpin structure has been made to downregulate the alb1 gene in the human pathogen Aspergillus fumigatus. Transformation of fungal protoplasts resulted in a high number of transformants with an inducible silenced phenotype (white spores). Efficient downregulation of the alb1 gene using this system suggests that this approach may overcome the variable downregulation observed with integrative constructs.


Assuntos
Aspergillus fumigatus/genética , Regulação para Baixo/genética , Genes Fúngicos/genética , Plasmídeos/genética , Interferência de RNA , Aspergillus fumigatus/efeitos dos fármacos , Aspergillus fumigatus/crescimento & desenvolvimento , Carboximetilcelulose Sódica/farmacologia , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Modelos Genéticos , Mutação , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
FEMS Microbiol Lett ; 264(2): 246-54, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16984401

RESUMO

The utility of the Aspergillus fumigatus cellobiohydrolase cbhB promoter for controlled gene expression has been investigated. cbhB message was present at high levels in the presence of carboxymethylcellulose and undetected in the presence of glucose. A reporter construct using the cbhB promoter showed similar behaviour and gave lower message levels than the Aspergillus nidulans alcA promoter under repressing conditions. An RNAi construct driven by the cbhB promoter was used to down-regulate the alb1 gene; transformants showed low alb1 message levels and a loss-of-function phenotype with carboxymethylcellulose, while both wild-type message levels and phenotype were seen with glucose. The cbhB promoter is therefore tightly controlled and can be exploited for the study of A. fumigatus.


Assuntos
Aspergillus fumigatus/genética , Celulose 1,4-beta-Celobiosidase/genética , Regulação Fúngica da Expressão Gênica , Regiões Promotoras Genéticas , Interferência de RNA/fisiologia , Aspergillus fumigatus/metabolismo , Celulose 1,4-beta-Celobiosidase/metabolismo , Genes Fúngicos , Plasmídeos/genética , Plasmídeos/metabolismo
13.
FEMS Microbiol Lett ; 248(1): 23-30, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15936900

RESUMO

The ADAMs are a family of integral membrane proteases involved in shedding and fusion events in animal tissues. Here, we report the identification of two ADAMs, ADM-A and ADM-B, in the pathogenic fungus Aspergillus fumigatus. The domain structure of metazoan ADAMs was seen in ADM-A and -B, although with some differences. ADAMs were identified in other filamentous fungi and phylogenetic analysis indicated that the fungal ADAMs were monophyletic and most closely related to metazoan ADAM 10 and 17. Recombinant ADM-B protease specifically cleaved casein and albumin while recombinant propeptide+protease was inactive. A sheddase function is therefore proposed for fungal ADAMs.


Assuntos
Aspergillus fumigatus/genética , Metaloendopeptidases/genética , Aspergillus fumigatus/enzimologia , Genes Fúngicos , Genoma Fúngico , Metaloendopeptidases/isolamento & purificação , Metaloendopeptidases/metabolismo , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA