Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(17): e2302152120, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37068249

RESUMEN

The primary antigenic and virulence determinant of the human malaria parasite Plasmodium falciparum is a variant surface protein called PfEMP1. Different forms of PfEMP1 are encoded by a multicopy gene family called var, and switching between active genes enables the parasites to evade the antibody response of their human hosts. var gene switching is key for the maintenance of chronic infections; however, what controls switching is unknown, although it has been suggested to occur at a constant frequency with little or no environmental influence. var gene transcription is controlled epigenetically through the activity of histone methyltransferases (HMTs). Studies in model systems have shown that metabolism and epigenetic control of gene expression are linked through the availability of intracellular S-adenosylmethionine (SAM), the principal methyl donor in biological methylation modifications, which can fluctuate based on nutrient availability. To determine whether environmental conditions and changes in metabolism can influence var gene expression, P. falciparum was cultured in media with altered concentrations of nutrients involved in SAM metabolism. We found that conditions that influence lipid metabolism induce var gene switching, indicating that parasites can respond to changes in their environment by altering var gene expression patterns. Genetic modifications that directly modified expression of the enzymes that control SAM levels similarly led to profound changes in var gene expression, confirming that changes in SAM availability modulate var gene switching. These observations directly challenge the paradigm that antigenic variation in P. falciparum follows an intrinsic, programed switching rate, which operates independently of any external stimuli.


Asunto(s)
Malaria Falciparum , Parásitos , Animales , Humanos , Plasmodium falciparum/metabolismo , Parásitos/metabolismo , Regulación de la Expresión Génica , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Malaria Falciparum/parasitología , Variación Antigénica/genética
2.
Mol Microbiol ; 115(4): 503-507, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33103309

RESUMEN

Malaria caused by the protozoan parasite Plasmodium falciparum continues to impose significant morbidity and mortality, despite substantial investment into drug and vaccine development and deployment. Underlying the resilience of this parasite is its remarkable ability to undergo genome modifications, thus, providing parasite populations with extensive genetic variability that accelerates selection of drug resistance and limits the efficacy of most vaccines. This genome plasticity is rooted in the mechanisms of DNA repair that parasites employ to maintain genome integrity, a process skewed toward homologous recombination through the evolutionary loss of classical nonhomologous end joining. Repair of DNA double-strand breaks have been shown to enable "shuffling" of antigen-encoding gene sequences to vastly increase antigen diversity and to enable copy number expansion of genes that contribute to drug resistance. The latter phenomenon has been proposed to be a major contributor to the rise of resistance to several classes of antimalarial drugs. In this issue of Molecular Microbiology, McDaniels and colleagues add yet another mechanism that malaria parasites use to reduce drug susceptibility by demonstrating that P. falciparum can maintain expanded arrays of drug resistance cassettes as stably replicating, circular, extrachromosomal DNAs, thus, expanding genome plasticity beyond the parasite's 14 nuclear chromosomes.


Asunto(s)
Resistencia a Medicamentos/genética , Genoma de Protozoos , Malaria Falciparum/prevención & control , Malaria Falciparum/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Animales , Variación Antigénica , Antimaláricos/farmacología , Reparación del ADN , Humanos , Vacunas
3.
Annu Rev Microbiol ; 71: 625-641, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28697665

RESUMEN

Malaria is a significant threat throughout the developing world. Among the most fascinating aspects of the protozoan parasites responsible for this disease are the methods they employ to avoid the immune system and perpetuate chronic infections. Key among these is antigenic variation: By systematically altering antigens that are displayed to the host's immune system, the parasite renders the adaptive immune response ineffective. For Plasmodium falciparum, the species responsible for the most severe form of human malaria, this process involves a complicated molecular mechanism that results in continuously changing patterns of variant-antigen-encoding gene expression. Although many features of this process remain obscure, significant progress has been made in recent years to decipher various molecular aspects of the regulatory cascade that causes chronic infection.


Asunto(s)
Variación Antigénica , Regulación de la Expresión Génica , Evasión Inmune , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Malaria/inmunología , Malaria/parasitología
4.
PLoS Biol ; 17(5): e3000271, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31083650

RESUMEN

Malaria parasites possess the remarkable ability to maintain chronic infections that fail to elicit a protective immune response, characteristics that have stymied vaccine development and cause people living in endemic regions to remain at risk of malaria despite previous exposure to the disease. These traits stem from the tremendous antigenic diversity displayed by parasites circulating in the field. For Plasmodium falciparum, the most virulent of the human malaria parasites, this diversity is exemplified by the variant gene family called var, which encodes the major surface antigen displayed on infected red blood cells (RBCs). This gene family exhibits virtually limitless diversity when var gene repertoires from different parasite isolates are compared. Previous studies indicated that this remarkable genome plasticity results from extensive ectopic recombination between var genes during mitotic replication; however, the molecular mechanisms that direct this process to antigen-encoding loci while the rest of the genome remains relatively stable were not determined. Using targeted DNA double-strand breaks (DSBs) and long-read whole-genome sequencing, we show that a single break within an antigen-encoding region of the genome can result in a cascade of recombination events leading to the generation of multiple chimeric var genes, a process that can greatly accelerate the generation of diversity within this family. We also found that recombinations did not occur randomly, but rather high-probability, specific recombination products were observed repeatedly. These results provide a molecular basis for previously described structured rearrangements that drive diversification of this highly polymorphic gene family.


Asunto(s)
Variación Antigénica/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Mitosis/genética , Parásitos/genética , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Recombinación Genética , Animales , Secuencia de Bases , Cromosomas/genética , Roturas del ADN de Doble Cadena , Humanos , Telómero/genética
5.
Artículo en Inglés | MEDLINE | ID: mdl-32179524

RESUMEN

Malaria parasites invade and replicate within red blood cells (RBCs), extensively modifying their structure and gaining access to the extracellular environment by placing the plasmodial surface anion channel (PSAC) into the RBC membrane. Expression of members of the cytoadherence linked antigen gene 3 (clag3) family is required for PSAC activity, a process that is regulated epigenetically. PSAC is a well-established route of uptake for large, hydrophilic antimalarial compounds, and parasites can acquire resistance by silencing clag3 gene expression, thereby reducing drug uptake. We found that exposure to sub-IC50 concentrations of the histone methyltransferase inhibitor chaetocin caused substantial changes in both clag3 gene expression and RBC permeability, and reversed acquired resistance to the antimalarial compound blasticidin S that is transported through PSACs. Chaetocin treatment also altered progression of parasites through their replicative cycle, presumably by changing their ability to modify chromatin appropriately to enable DNA replication. These results indicate that targeting histone modifiers could represent a novel tool for reversing epigenetically acquired drug resistance in P. falciparum.


Asunto(s)
Malaria Falciparum , Parásitos , Preparaciones Farmacéuticas , Animales , Resistencia a Medicamentos/genética , Eritrocitos/metabolismo , Histona Metiltransferasas , Histonas/genética , Parásitos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
6.
Proc Natl Acad Sci U S A ; 112(38): 11935-40, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26351679

RESUMEN

Identification and genetic validation of new targets from available genome sequences are critical steps toward the development of new potent and selective antimalarials. However, no methods are currently available for large-scale functional analysis of the Plasmodium falciparum genome. Here we present evidence for successful use of morpholino oligomers (MO) to mediate degradation of target mRNAs or to inhibit RNA splicing or translation of several genes of P. falciparum involved in chloroquine transport, apicoplast biogenesis, and phospholipid biosynthesis. Consistent with their role in the parasite life cycle, down-regulation of these essential genes resulted in inhibition of parasite development. We show that a MO conjugate that targets the chloroquine-resistant transporter PfCRT is effective against chloroquine-sensitive and -resistant parasites, causes enlarged digestive vacuoles, and renders chloroquine-resistant strains more sensitive to chloroquine. Similarly, we show that a MO conjugate that targets the PfDXR involved in apicoplast biogenesis inhibits parasite growth and that this defect can be rescued by addition of isopentenyl pyrophosphate. MO-based gene regulation is a viable alternative approach to functional analysis of the P. falciparum genome.


Asunto(s)
Morfolinos/farmacología , Plasmodium falciparum/genética , Biosíntesis de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Empalme del ARN/efectos de los fármacos , Animales , Antimaláricos/farmacología , Artemisininas/farmacología , Cloroquina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Citometría de Flujo , Genes Reporteros , Hemiterpenos/metabolismo , Luciferasas/metabolismo , Compuestos Organofosforados/metabolismo , Parásitos/efectos de los fármacos , Parásitos/genética , Parásitos/crecimiento & desarrollo , Péptidos/farmacología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN
7.
PLoS Genet ; 11(5): e1005234, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25993442

RESUMEN

Mutually exclusive gene expression, whereby only one member of a multi-gene family is selected for activation, is used by the malaria parasite Plasmodium falciparum to escape the human immune system and perpetuate long-term, chronic infections. A family of genes called var encodes the chief antigenic and virulence determinant of P. falciparum malaria. var genes are transcribed in a mutually exclusive manner, with switching between active genes resulting in antigenic variation. While recent work has shed considerable light on the epigenetic basis for var gene activation and silencing, how switching is controlled remains a mystery. In particular, switching seems not to be random, but instead appears to be coordinated to result in timely activation of individual genes leading to sequential waves of antigenically distinct parasite populations. The molecular basis for this apparent coordination is unknown. Here we show that var2csa, an unusual and highly conserved var gene, occupies a unique position within the var gene switching hierarchy. Induction of switching through the destabilization of var specific chromatin using both genetic and chemical methods repeatedly led to the rapid and exclusive activation of var2csa. Additional experiments demonstrated that these represent "true" switching events and not simply de-silencing of the var2csa promoter, and that activation is limited to the unique locus on chromosome 12. Combined with translational repression of var2csa transcripts, frequent "default" switching to this locus and detection of var2csa untranslated transcripts in non-pregnant individuals, these data suggest that var2csa could play a central role in coordinating switching, fulfilling a prediction made by mathematical models derived from population switching patterns. These studies provide the first insights into the mechanisms by which var gene switching is coordinated as well as an example of how a pharmacological agent can disrupt antigenic variation in Plasmodium falciparum.


Asunto(s)
Antígenos de Protozoos/metabolismo , Evasión Inmune , Plasmodium falciparum/inmunología , Proteínas Protozoarias/metabolismo , Variación Antigénica , Antígenos de Protozoos/genética , Azepinas/farmacología , Cloroquina/farmacología , Regulación de la Expresión Génica , Sitios Genéticos , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Concentración 50 Inhibidora , Malaria Falciparum/parasitología , Modelos Teóricos , Piperazinas/farmacología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Regiones Promotoras Genéticas , Proteínas Protozoarias/genética , Quinazolinas/farmacología , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Terpenos/farmacología , Activación Transcripcional , Transcriptoma
8.
PLoS Pathog ; 10(1): e1003854, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24391504

RESUMEN

Histone modifications are important regulators of gene expression in all eukaryotes. In Plasmodium falciparum, these epigenetic marks regulate expression of genes involved in several aspects of host-parasite interactions, including antigenic variation. While the identities and genomic positions of many histone modifications have now been cataloged, how they are targeted to defined genomic regions remains poorly understood. For example, how variant antigen encoding loci (var) are targeted for deposition of unique histone marks is a mystery that continues to perplex the field. Here we describe the recruitment of an ortholog of the histone modifier SET2 to var genes through direct interactions with the C-terminal domain (CTD) of RNA polymerase II. In higher eukaryotes, SET2 is a histone methyltransferase recruited by RNA pol II during mRNA transcription; however, the ortholog in P. falciparum (PfSET2) has an atypical architecture and its role in regulating transcription is unknown. Here we show that PfSET2 binds to the unphosphorylated form of the CTD, a property inconsistent with its recruitment during mRNA synthesis. Further, we show that H3K36me3, the epigenetic mark deposited by PfSET2, is enriched at both active and silent var gene loci, providing additional evidence that its recruitment is not associated with mRNA production. Over-expression of a dominant negative form of PfSET2 designed to disrupt binding to RNA pol II induced rapid var gene expression switching, confirming both the importance of PfSET2 in var gene regulation and a role for RNA pol II in its recruitment. RNA pol II is known to transcribe non-coding RNAs from both active and silent var genes, providing a possible mechanism by which it could recruit PfSET2 to var loci. This work unifies previous reports of histone modifications, the production of ncRNAs, and the promoter activity of var introns into a mechanism that contributes to antigenic variation by malaria parasites.


Asunto(s)
Variación Antigénica/fisiología , Antígenos de Protozoos/inmunología , N-Metiltransferasa de Histona-Lisina/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , ARN Polimerasa II/inmunología , Antígenos de Protozoos/genética , Epigénesis Genética/inmunología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , ARN Polimerasa II/genética , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Protozoario/genética , ARN Protozoario/inmunología
9.
Nucleic Acids Res ; 42(1): 370-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24089143

RESUMEN

Malaria parasites replicate asexually within their mammalian hosts as haploid cells and are subject to DNA damage from the immune response and chemotherapeutic agents that can significantly disrupt genomic integrity. Examination of the annotated genome of the parasite Plasmodium falciparum identified genes encoding core proteins required for the homologous recombination (HR) pathway for repairing DNA double-strand breaks (DSBs), but surprisingly none of the components of the canonical non-homologous end joining (C-NHEJ) pathway were identified. To better understand how malaria parasites repair DSBs and maintain genome integrity, we modified the yeast I-SceI endonuclease system to generate inducible, site-specific DSBs within the parasite's genome. Analysis of repaired genomic DNA showed that parasites possess both a typical HR pathway resulting in gene conversion events as well as an end joining (EJ) pathway for repair of DSBs when no homologous sequence is available. The products of EJ were limited in number and identical products were observed in multiple independent experiments. The repair junctions frequently contained short insertions also found in the surrounding sequences, suggesting the possibility of a templated repair process. We propose that an alternative end-joining pathway rather than C-NHEJ, serves as a primary method for repairing DSBs in malaria parasites.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Plasmodium falciparum/genética , Reparación del ADN por Recombinación , Secuencia de Bases , Roturas del ADN de Doble Cadena , Conversión Génica , Genoma de Protozoos , Datos de Secuencia Molecular
10.
Nucleic Acids Res ; 42(4): 2270-81, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24253306

RESUMEN

Many bacterial, viral and parasitic pathogens undergo antigenic variation to counter host immune defense mechanisms. In Plasmodium falciparum, the most lethal of human malaria parasites, switching of var gene expression results in alternating expression of the adhesion proteins of the Plasmodium falciparum-erythrocyte membrane protein 1 class on the infected erythrocyte surface. Recombination clearly generates var diversity, but the nature and control of the genetic exchanges involved remain unclear. By experimental and bioinformatic identification of recombination events and genome-wide recombination hotspots in var genes, we show that during the parasite's sexual stages, ectopic recombination between isogenous var paralogs occurs near low folding free energy DNA 50-mers and that these sequences are heavily concentrated at the boundaries of regions encoding individual Plasmodium falciparum-erythrocyte membrane protein 1 structural domains. The recombinogenic potential of these 50-mers is not parasite-specific because these sequences also induce recombination when transferred to the yeast Saccharomyces cerevisiae. Genetic cross data suggest that DNA secondary structures (DSS) act as inducers of recombination during DNA replication in P. falciparum sexual stages, and that these DSS-regulated genetic exchanges generate functional and diverse P. falciparum adhesion antigens. DSS-induced recombination may represent a common mechanism for optimizing the evolvability of virulence gene families in pathogens.


Asunto(s)
Variación Antigénica/genética , Antígenos de Protozoos/genética , ADN Protozoario/química , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Recombinación Genética , Antígenos de Protozoos/química , Genes Protozoarios , Familia de Multigenes , Conformación de Ácido Nucleico , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Saccharomyces cerevisiae/genética
11.
Mol Biol Evol ; 31(7): 1649-60, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24739308

RESUMEN

Biological robustness allows mutations to accumulate while maintaining functional phenotypes. Despite its crucial role in evolutionary processes, the mechanistic details of how robustness originates remain elusive. Using an evolutionary trajectory analysis approach, we demonstrate how robustness evolved in malaria parasites under selective pressure from an antimalarial drug inhibiting the folate synthesis pathway. A series of four nonsynonymous amino acid substitutions at the targeted enzyme, dihydrofolate reductase (DHFR), render the parasites highly resistant to the antifolate drug pyrimethamine. Nevertheless, the stepwise gain of these four dhfr mutations results in tradeoffs between pyrimethamine resistance and parasite fitness. Here, we report the epistatic interaction between dhfr mutations and amplification of the gene encoding the first upstream enzyme in the folate pathway, GTP cyclohydrolase I (GCH1). gch1 amplification confers low level pyrimethamine resistance and would thus be selected for by pyrimethamine treatment. Interestingly, the gch1 amplification can then be co-opted by the parasites because it reduces the cost of acquiring drug-resistant dhfr mutations downstream in the same metabolic pathway. The compensation of compromised fitness by extra GCH1 is an example of how robustness can evolve in a system and thus expand the accessibility of evolutionary trajectories leading toward highly resistant alleles. The evolution of robustness during the gain of drug-resistant mutations has broad implications for both the development of new drugs and molecular surveillance for resistance to existing drugs.


Asunto(s)
Evolución Biológica , Resistencia a Medicamentos , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Plasmodium falciparum/fisiología , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Sustitución de Aminoácidos , Antimaláricos/farmacología , Epistasis Genética , Genes Protozoarios , Aptitud Genética , Humanos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/genética , Pirimetamina/farmacología , Transducción de Señal/efectos de los fármacos
12.
Mol Microbiol ; 90(3): 472-88, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23980802

RESUMEN

Plasmodium falciparum malaria parasites export the protein PfEMP1 to the surface of infected erythrocytes, enabling them to adhere to receptors in the microvasculature and thereby avoid clearance by the spleen. The gene var2csa encodes the form of PfEMP1 that binds specifically within the placenta, causing pregnancy-associated malaria, and appears to not be expressed in the absence of a placenta. We previously described an upstream open reading frame (uORF) that is responsible for repression of translation of the downstream ORF (dORF) that encodes VAR2CSA, thus keeping the gene silent when parasites infect non-pregnant individuals. To elucidate the molecular mechanism by which this repression is overcome during pregnancy, we stably transformed parasites with reporter gene constructs designed to detect switches in the efficiency of dORF translation. We found that proper regulation of switching relies on two separate components, (i) active translation of the uORF and (ii) sequence-specific characteristics of the surrounding transcript, which together control the ability of the ribosome complex to reinitiate a second round of translation and thus express VAR2CSA. These results provide the first details of a molecular switch that allows parasites take advantage of the unique niche provided by the placenta.


Asunto(s)
Antígenos de Protozoos/genética , Malaria Falciparum/parasitología , Placenta/parasitología , Plasmodium falciparum/genética , Antígenos de Protozoos/metabolismo , ADN Protozoario/análisis , Femenino , Regulación de la Expresión Génica , Humanos , Malaria , Sistemas de Lectura Abierta , Plasmodium falciparum/crecimiento & desarrollo , Embarazo
13.
Mol Microbiol ; 88(4): 702-12, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23347134

RESUMEN

Resistance to antimalarials targeting the folate pathway is widespread. GTP-cyclohydrolase (gch1), the first enzyme in this pathway, exhibits extensive copy number variation (CN) in parasite isolates from areas with a history of longstanding antifolate use. Increased CN of gch1 is associated with a greater number of point mutations in enzymes targeted by the antifolates, pyrimethamine and sulphadoxine. While these observations suggest that increases in gch1 CN are an adaptation to drug pressure, changes in CN have not been experimentally demonstrated to directly alter drug susceptibility. To determine if changes in gch1 expression alone modify pyrimethamine sensitivity, we manipulated gch1 CN in several parasite lines to test the effect on drug sensitivity. We report that increases in gch1 CN alter pyrimethamine resistance in most parasites lines. However we find evidence of a detrimental effect of very high levels of gch1 overexpression in parasite lines with high endogenous levels of gch1 expression, revealing the importance of maintaining balance in the folate pathway and implicating changes in gch1 expression in preserving proper metabolic flux. This work expands our understanding of parasite adaptation to drug pressure and provides a possible mechanism for how specific mutations become fixed within parasite populations.


Asunto(s)
Adaptación Biológica , Antimaláricos/farmacología , Resistencia a Medicamentos , Antagonistas del Ácido Fólico/farmacología , Dosificación de Gen , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Genes Protozoarios , Pirimetamina/farmacología
14.
Blood ; 119(2): e1-8, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22106347

RESUMEN

Infection of erythrocytes with the human malaria parasite, Plasmodium falciparum, results in dramatic changes to the host cell structure and morphology. The predicted functional localization of the STEVOR proteins at the erythrocyte surface suggests that they may be involved in parasite-induced modifications of the erythrocyte membrane during parasite development. To address the biologic function of STEVOR proteins, we subjected a panel of stevor transgenic parasites and wild-type clonal lines exhibiting different expression levels for stevor genes to functional assays exploring parasite-induced modifications of the erythrocyte membrane. Using this approach, we show that stevor expression impacts deformability of the erythrocyte membrane. This process may facilitate parasite sequestration in deep tissue vasculature.


Asunto(s)
Antígenos de Protozoos/metabolismo , Membrana Eritrocítica/patología , Eritrocitos/patología , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Antígenos de Protozoos/genética , Células Cultivadas , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Plasmodium falciparum/aislamiento & purificación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
bioRxiv ; 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38496509

RESUMEN

Chronic, asymptomatic malaria infections contribute substantially to disease transmission and likely represent the most significant impediment preventing malaria elimination and eradication. Plasmodium falciparum parasites evade antibody recognition through transcriptional switching between members of the var gene family, which encodes the major virulence factor and surface antigen on infected red blood cells. This process can extend infections for up to a year; however, infections have been documented to last for over a decade, constituting an unseen reservoir of parasites that undermine eradication and control efforts. How parasites remain immunologically "invisible" for such lengthy periods is entirely unknown. Here we show that in addition to the accepted paradigm of mono-allelic var gene expression, individual parasites can simultaneously express multiple var genes or enter a state in which little or no var gene expression is detectable. This unappreciated flexibility provides parasites with greater adaptive capacity than previously understood and challenges the dogma of mutually exclusive var gene expression. It also provides an explanation for the antigenically "invisible" parasites observed in chronic asymptomatic infections.

16.
BMC Evol Biol ; 13: 37, 2013 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-23398820

RESUMEN

BACKGROUND: The acquisition of complex transcriptional regulatory abilities and epigenetic machinery facilitated the transition of the ancestor of apicomplexans from a free-living organism to an obligate parasite. The ability to control sophisticated gene expression patterns enabled these ancient organisms to evolve several differentiated forms, invade multiple hosts and evade host immunity. How these abilities were acquired remains an outstanding question in protistan biology. RESULTS: In this work, we study SET domain bearing genes that are implicated in mediating immune evasion, invasion and cytoadhesion pathways of modern apicomplexans, including malaria parasites. We provide the first conclusive evidence of a horizontal gene transfer of a Histone H4 Lysine 20 (H4K20) modifier, Set8, from an animal host to the ancestor of apicomplexans. Set8 is known to contribute to the coordinated expression of genes involved in immune evasion in modern apicomplexans. We also show the likely transfer of a H3K36 methyltransferase (Ashr3 from plants), possibly derived from algal endosymbionts. These transfers appear to date to the transition from free-living organisms to parasitism and coincide with the proposed horizontal acquisition of cytoadhesion domains, the O-glycosyltransferase that modifies these domains, and the primary family of transcription factors found in apicomplexan parasites. Notably, phylogenetic support for these conclusions is robust and the genes clearly are dissimilar to SET sequences found in the closely related parasite Perkinsus marinus, and in ciliates, the nearest free-living organisms with complete genome sequences available. CONCLUSIONS: Animal and plant sources of epigenetic machinery provide new insights into the evolution of parasitism in apicomplexans. Along with the horizontal transfer of cytoadhesive domains, O-linked glycosylation and key transcription factors, the acquisition of SET domain methyltransferases marks a key transitional event in the evolution to parasitism in this important protozoan lineage.


Asunto(s)
Apicomplexa/genética , Evolución Biológica , Transferencia de Gen Horizontal , Plasmodium falciparum/genética , Secuencia de Aminoácidos , Animales , Teorema de Bayes , ADN Protozoario/genética , Dictyostelium/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Funciones de Verosimilitud , Nematodos/genética , Parásitos/genética , Filogenia , Análisis de Secuencia de ADN
17.
PLoS Pathog ; 12(3): e1005329, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27031699
19.
Gene ; 877: 147516, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37286021

RESUMEN

Protozoan parasites of the genus Plasmodium cause malaria, a mosquito borne disease responsible for substantial health and economic costs throughout the developing world. During transition from human host to insect vector, the parasites undergo profound changes in morphology, host cell tropism and gene expression. Unique among eukaryotes, Plasmodium differentiation through each stage of development includes differential expression of singular, stage-specific ribosomal RNAs, permitting real-time adaptability to major environmental changes. In the mosquito vector, these Plasmodium parasites respond to changes in temperature by modulating transcriptional activities, allowing real-time responses to environmental cues. Here, we identify a novel form of long noncoding RNA: a temperature-regulated untranslated lncRNA (tru-lncRNA) that influences the Plasmodium parasite's ability to respond to changes in its local environment. Expression of this tru-lncRNA is specifically induced by shifts in temperature from 37 °C to ambient temperature that parallels the transition from mammalian host to insect vector. Interestingly, deletion of tru-lncRNA from the genome may prevent processing of S-type rRNA thereby affecting the protein synthesis machinery. Malaria prevention and mitigation strategies aimed at disrupting the Plasmodium life cycle will benefit from the characterization of ancillary biomolecules (including tru-lncRNAs) that are constitutively sensitive to micro- environmental parameters.


Asunto(s)
Malaria , Parásitos , Plasmodium , ARN Largo no Codificante , Animales , Humanos , Parásitos/genética , ARN Largo no Codificante/genética , Temperatura , Plasmodium/genética , Malaria/genética , ARN Ribosómico/genética , Mamíferos/genética
20.
PLoS One ; 18(11): e0294066, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38019839

RESUMEN

Leucocytozoon is a haemosporidian parasite known to cause leucocytozoonosis in domestic and wild birds in most parts of the world. It is an important pathogen, as some species can be pathogenic, especially in domestic birds. One of the factors affecting poultry health management worldwide is parasitism. However, the study of haemosporidian parasites in Ghana is still lacking. This study sought to assess the prevalence and diversity of Leucocytozoon parasites in domestic birds in Ghana. Blood samples were collected from domestic birds in Ghana's Bono and Eastern regions to screen for Leucocytozoon parasites. Thin blood smears were prepared for microscopy and DNA was extracted from whole blood kept in ethylenediaminetetraacetic acid (EDTA) tubes for PCR. Due to the large number of samples, real-time PCR was performed to amplify the conserved rDNA gene. Two different nested PCR protocols were performed on the positive samples obtained from real-time PCR results, to amplify a partial region of the mitochondrial cytochrome b gene and the amplicons were sequenced. Sequencing revealed six new lineages of Leucocytozoon sp. recovered in 976 individual domestic birds and these sequences were deposited in the National Center for Biotechnology Information (NCBI) GenBank. An overall Leucocytozoon prevalence of 11.6% was reported in all birds sampled. The most prevalent lineage LGHA146 (GenBank accession no. OM643346) (93.8%) was found infecting 3 bird species, Gallus gallus, Meleagris gallopavo, and Anas platyrhynchos. Phylogenetic analysis revealed that the new lineages (GenBank accession nos. OM643342, OM643343, OM643344, OM643345, OM643346, and OM643347), reported in this study were closely related to Leucocytozoon schoutedeni. We suggest that further studies be conducted to evaluate the effect of these parasite species on the general well-being of poultry in Ghana.


Asunto(s)
Enfermedades de las Aves , Haemosporida , Parásitos , Infecciones Protozoarias en Animales , Animales , Filogenia , Prevalencia , Ghana/epidemiología , Enfermedades de las Aves/epidemiología , Enfermedades de las Aves/parasitología , Haemosporida/genética , Aves , Parásitos/genética , Infecciones Protozoarias en Animales/epidemiología , Infecciones Protozoarias en Animales/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA