Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Microbiol Spectr ; 10(3): e0070222, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35652638

RESUMEN

Human toxoplasmosis is a life-threatening disease caused by the apicomplexan parasite Toxoplasma gondii. Rapid replication of the tachyzoite is associated with symptomatic disease, while suppressed division of the bradyzoite is responsible for chronic disease. Here, we identified the T. gondii cell cycle mechanism, the G1 restriction checkpoint (R-point), that operates the switch between parasite growth and differentiation. Apicomplexans lack conventional R-point regulators, suggesting adaptation of alternative factors. We showed that Cdk-related G1 kinase TgCrk2 forms alternative complexes with atypical cyclins (TgCycP1, TgCycP2, and TgCyc5) in the rapidly dividing developmentally incompetent RH and slower dividing developmentally competent ME49 tachyzoites and bradyzoites. Examination of cyclins verified the correlation of cyclin expression with growth dependence and development capacity of RH and ME49 strains. We demonstrated that rapidly dividing RH tachyzoites were dependent on TgCycP1 expression, which interfered with bradyzoite differentiation. Using the conditional knockdown model, we established that TgCycP2 regulated G1 duration in the developmentally competent ME49 tachyzoites but not in the developmentally incompetent RH tachyzoites. We tested the functions of TgCycP2 and TgCyc5 in alkaline induced and spontaneous bradyzoite differentiation (rat embryonic brain cells) models. Based on functional and global gene expression analyses, we determined that TgCycP2 also regulated bradyzoite replication, while signal-induced TgCyc5 was critical for efficient tissue cyst maturation. In conclusion, we identified the central machinery of the T. gondii restriction checkpoint comprised of TgCrk2 kinase and three atypical T. gondii cyclins and demonstrated the independent roles of TgCycP1, TgCycP2, and TgCyc5 in parasite growth and development. IMPORTANCE Toxoplasma gondii is a virulent and abundant human pathogen that puts millions of silently infected people at risk of reactivation of the chronic disease. Encysted bradyzoites formed during the chronic stage are resistant to current therapies. Therefore, insights into the mechanism of tissue cyst formation and reactivation are major areas of investigation. The fact that rapidly dividing parasites differentiate poorly strongly suggests that there is a threshold of replication rate that must be crossed to be considered for differentiation. We discovered a cell cycle mechanism that controls the T. gondii growth-rest switch involved in the conversion of dividing tachyzoites into largely quiescent bradyzoites. This switch operates the T. gondii restriction checkpoint using a set of atypical and parasite-specific regulators. Importantly, the novel T. gondii R-point network was not present in the parasite's human and animal hosts, offering a wealth of new and parasite-specific drug targets to explore in the future.


Asunto(s)
Toxoplasma , Toxoplasmosis , Animales , Ciclo Celular , Diferenciación Celular , Ciclinas/metabolismo , Humanos , Ratas , Toxoplasma/genética
2.
Artículo en Inglés | MEDLINE | ID: mdl-32714878

RESUMEN

Apicomplexa are obligate intracellular parasites which cause various animal and human diseases including malaria, toxoplasmosis, and cryptosporidiosis. They proliferate by a unique mechanism that combines physically separated semi-closed mitosis of the nucleus and assembly of daughter cells by internal budding. Mitosis occurs in the presence of a nuclear envelope and with little appreciable chromatin condensation. A long standing question in the field has been how parasites keep track of their uncondensed chromatin chromosomes throughout their development, and hence secure proper chromosome segregation during division. Past work demonstrated that the centromeres, the region of kinetochore assembly at chromosomes, of Toxoplasma gondii remain clustered at a defined region of the nuclear periphery proximal to the main microtubule organizing center of the cell, the centrosome. We have proposed that this mechanism is likely involved in the process. Here we set out to identify underlying molecular players involved in centromere clustering. Through pharmacological treatment and structural analysis we show that centromere clustering is not mediated by persistent microtubules of the mitotic spindle. We identify the chromatin binding factor a homolog of structural maintenance of chromosomes 1 (SMC1). Additionally, we show that both TgSMC1, and a centromeric histone, interact with TgExportin1, a predicted soluble component of the nuclear pore complex. Our results suggest that the nuclear envelope, and in particular the nuclear pore complex may play a role in positioning centromeres in T. gondii.


Asunto(s)
Toxoplasma , Animales , Centrómero , Segregación Cromosómica , Cromosomas Humanos Par 1 , Humanos , Poro Nuclear , Toxoplasma/genética
3.
JCI Insight ; 5(12)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32554925

RESUMEN

BACKGROUNDPrediction of adverse outcomes in cerebral malaria (CM) is difficult. We hypothesized that cell-free DNA (cfDNA) levels would facilitate identification of severe and potentially fatal CM cases.METHODSIn this retrospective study, plasma from Malawian children with CM (n = 134), uncomplicated malaria (UM, n = 77), and healthy controls (HC, n = 60) was assayed for cfDNA using a fluorescence assay. Host and parasite cfDNA was measured by quantitative PCR. Immune markers were determined by ELISA, Luminex, or cytometric bead array.RESULTSTotal cfDNA increased with malaria severity (HC versus UM, P < 0.001; HC versus CM, P < 0.0001; UM versus CM, P < 0.0001), was elevated in retinopathy-positive (Ret+) CM relative to Ret- CM (7.66 versus 5.47 ng/µL, P = 0.027), and differentiated Ret+ fatal cases from survivors (AUC 0.779; P < 0.001). cfDNA levels in patients with non-malarial febrile illness (NMF, P = 0.25) and non-malarial coma (NMC, P = 0.99) were comparable with UM. Host DNA, rather than parasite DNA, was the major cfDNA contributor (UM, 268 versus 67 pg/µL; CM, 2824 versus 463 pg/µL). Host and parasite cfDNA distinguished CM by retinopathy (host, AUC 0.715, P = 0.0001; parasite, AUC 0.745, P = 0.0001), but only host cfDNA distinguished fatal cases (AUC 0.715, P = 0.0001). Total cfDNA correlated with neutrophil markers IL-8 (rs = 0.433, P < 0.0001) and myeloperoxidase (rs = 0.683, P < 0.0001).CONCLUSIONQuantifying plasma cfDNA is a simple assay useful in identifying children at risk for fatal outcome and has promise as a point-of-care assay. Elevated cfDNA suggests a link with host inflammatory pathways in fatal CM.FUNDINGNIH NCATS (AK), Burroughs-Wellcome (AK), and National Health and Medical Research Council of Australia (SJR).


Asunto(s)
Biomarcadores/sangre , Ácidos Nucleicos Libres de Células/sangre , Malaria Cerebral/diagnóstico , Malaria Falciparum/sangre , Plasma/metabolismo , Adolescente , Niño , Preescolar , Femenino , Humanos , Malaria Cerebral/sangre , Malaria Cerebral/parasitología , Malaria Falciparum/diagnóstico , Masculino , Neutrófilos/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-30838177

RESUMEN

Twenty-two compounds belonging to several classes of polyamine analogs have been examined for their ability to inhibit the growth of the human malaria parasite Plasmodium falciparum in vitro and in vivo. Four lead compounds from the thiourea sub-series and one compound from the urea-based analogs were found to be potent inhibitors of both chloroquine-resistant (Dd2) and chloroquine-sensitive (3D7) strains of Plasmodium with IC50 values ranging from 150 to 460 nM. In addition, the compound RHW, N1,N7-bis (3-(cyclohexylmethylamino) propyl) heptane-1,7-diamine tetrabromide was found to inhibit Dd2 with an IC50 of 200 nM. When RHW was administered to P. yoelii-infected mice at 35 mg/kg for 4 days, it significantly reduced parasitemia. RHW was also assayed in combination with the ornithine decarboxylase inhibitor difluoromethylornithine, and the two drugs were found not to have synergistic antimalarial activity. Furthermore, these inhibitors led to decreased cellular spermidine and spermine levels in P. falciparum, suggesting that they exert their antimalarial activities by inhibition of spermidine synthase.


Asunto(s)
Antimaláricos/farmacología , Plasmodium falciparum/efectos de los fármacos , Poliaminas/farmacología , Espermidina/análisis , Espermina/análisis , Animales , Antimaláricos/administración & dosificación , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Concentración 50 Inhibidora , Malaria/tratamiento farmacológico , Ratones , Carga de Parásitos , Parasitemia , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/química , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium yoelii/efectos de los fármacos , Poliaminas/administración & dosificación
5.
mBio ; 8(4)2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28830940

RESUMEN

Our knowledge of cell cycle regulatory mechanisms in apicomplexan parasites is very limited. In this study, we describe a novel Toxoplasma gondii factor that has a vital role in chromosome replication and the regulation of cytoplasmic and nuclear mitotic structures, and we named this factor ECR1 for essential for chromosome replication 1. ECR1 was discovered by complementation of a temperature-sensitive (ts) mutant that suffers lethal, uncontrolled chromosome replication at 40°C similar to a ts mutant carrying a defect in topoisomerase. ECR1 is a 52-kDa protein containing divergent RING and TRAF-Sina-like zinc binding domains that are dynamically expressed in the tachyzoite cell cycle. ECR1 first appears in the unique spindle compartment of the Apicomplexa (centrocone) of the nuclear envelope in early S phase and then in the nucleus in late S phase where it reaches maximum expression. Following nuclear division, but before daughter parasites separate from the mother parasite, ECR1 is downregulated and is absent in new daughter parasites. The proteomics of ECR1 identified interactions with the ubiquitin-mediated protein degradation machinery and the minichromosome maintenance complex, and the loss of ECR1 led to increased stability of a key member of this complex, MCM2. ECR1 also forms a stable complex with the cyclin-dependent kinase (CDK)-related kinase, Tgondii Crk5 (TgCrk5), which displays a similar cell cycle expression and localization during tachyzoite replication. Importantly, the localization of ECR1/TgCrk5 in the centrocone indicates that this Apicomplexa-specific spindle compartment houses important regulatory factors that control the parasite cell cycle.IMPORTANCE Parasites of the apicomplexan family are important causes of human disease, including malaria, toxoplasmosis, and cryptosporidiosis. Parasite growth is the underlying cause of pathogenesis, yet despite this importance, the molecular basis for parasite replication is poorly understood. Filling this knowledge gap cannot be accomplished by mining recent whole-genome sequencing data because apicomplexan cell cycles differ substantially and lack many of the key regulatory factors of well-studied yeast and mammalian cell division models. We have utilized forward genetics to discover essential factors that regulate cell division in these parasites using the Toxoplasma gondii model. An example of this approach is described here with the discovery of a putative E3 ligase/protein kinase mechanism involved in regulating chromosome replication and mitotic processes of asexual stage parasites.


Asunto(s)
Ciclo Celular/genética , Regulación de la Expresión Génica , Proteínas Protozoarias/metabolismo , Huso Acromático/metabolismo , Toxoplasma/genética , Toxoplasma/fisiología , Puntos de Control del Ciclo Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromosomas/genética , Cromosomas/fisiología , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Replicación del ADN , ADN-Topoisomerasas/genética , ADN-Topoisomerasas/metabolismo , Mitosis , Membrana Nuclear/genética , Proteínas Protozoarias/genética , Toxoplasmosis/parasitología , Ubiquitina-Proteína Ligasas/metabolismo
6.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28630062

RESUMEN

Repeated stimulation of T cells that occurs in the context of chronic infection results in progressively reduced responsiveness of T cells to pathogen-derived antigens. This phenotype, known as T cell exhaustion, occurs during chronic infections caused by a variety of pathogens, from persistent viruses to parasites. Unlike the memory cells that typically form after successful pathogen clearance following an acute infection, exhausted T cells secrete lower levels of effector cytokines, proliferate less in response to cognate antigen, and upregulate cell surface inhibitory molecules such as PD-1 and LAG-3. The molecular events that lead to the induction of this phenotype have, however, not been fully characterized. In T cells, members of the NFAT family of transcription factors not only are responsible for the expression of many activation-induced genes but also are crucial for the induction of transcriptional programs that inhibit T cell activation and maintain tolerance. Here we show that NFAT1-deficient CD4+ T cells maintain higher proliferative capacity and expression of effector cytokines following Plasmodium yoelii infection and are therefore more resistant to P. yoelii-induced exhaustion than their wild-type counterparts. Consequently, gene expression microarray analysis of CD4+ T cells following P. yoelii-induced exhaustion shows upregulation of effector T cell-associated genes in the absence of NFAT1 compared with wild-type exhausted T cells. Furthermore, adoptive transfer of NFAT1-deficient CD4+ T cells into mice infected with P. yoelii results in increased production of antibodies to cognate antigen. Our results support the idea that NFAT1 is necessary to fully suppress effector responses during Plasmodium-induced CD4+ T cell exhaustion.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interacciones Huésped-Patógeno , Tolerancia Inmunológica , Malaria/patología , Factores de Transcripción NFATC/metabolismo , Plasmodium yoelii/patogenicidad , Animales , Proliferación Celular , Citocinas/metabolismo , Activación de Linfocitos , Malaria/inmunología , Ratones Endogámicos C57BL
7.
Antimicrob Agents Chemother ; 59(9): 5555-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26124159

RESUMEN

ELQ-300 is a preclinical candidate that targets the liver and blood stages of Plasmodium falciparum, as well as the forms that are crucial to transmission of disease: gametocytes, zygotes, and ookinetes. A significant obstacle to the clinical development of ELQ-300 is related to its physicochemical properties. Its relatively poor aqueous solubility and high crystallinity limit absorption to the degree that only low blood concentrations can be achieved following oral dosing. While these low blood concentrations are sufficient for therapy, the levels are too low to establish an acceptable safety margin required by regulatory agencies for clinical development. One way to address the challenging physicochemical properties of ELQ-300 is through the development of prodrugs. Here, we profile ELQ-337, a bioreversible O-linked carbonate ester prodrug of the parent molecule. At the molar equivalent dose of 3 mg/kg of body weight, the delivery of ELQ-300 from ELQ-337 is enhanced by 3- to 4-fold, reaching a maximum concentration of drug in serum (C max) of 5.9 µM by 6 h after oral administration, and unlike ELQ-300 at any dose, ELQ-337 provides single-dose cures of patent malaria infections in mice at low-single-digit milligram per kilogram doses. Our findings show that the prodrug strategy represents a viable approach to overcome the physicochemical limitations of ELQ-300 to deliver the active drug to the bloodstream at concentrations sufficient for safety and toxicology studies, as well as achieving single-dose cures.


Asunto(s)
Antimaláricos/química , Antimaláricos/uso terapéutico , Malaria/tratamiento farmacológico , Profármacos/uso terapéutico , Quinolonas/uso terapéutico , Animales , Cristalografía por Rayos X , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Femenino , Ratones , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Profármacos/química , Quinolonas/química
8.
Am J Trop Med Hyg ; 92(6): 1195-201, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25918204

RESUMEN

Single-dose therapies for malaria have been proposed as a way to reduce the cost and increase the effectiveness of antimalarial treatment. However, no compound to date has shown single-dose activity against both the blood-stage Plasmodium parasites that cause disease and the liver-stage parasites that initiate malaria infection. Here, we describe a subset of cytochrome bc1 (cyt bc1) inhibitors, including the novel 4(1H)-quinolone ELQ-400, with single-dose activity against liver, blood, and transmission-stage parasites in mouse models of malaria. Although cyt bc1 inhibitors are generally classified as slow-onset antimalarials, we found that a single dose of ELQ-400 rapidly induced stasis in blood-stage parasites, which was associated with a rapid reduction in parasitemia in vivo. ELQ-400 also exhibited a low propensity for drug resistance and was active against atovaquone-resistant P. falciparum strains with point mutations in cyt bc1. Ultimately, ELQ-400 shows that cyt bc1 inhibitors can function as single-dose, blood-stage antimalarials and is the first compound to provide combined treatment, prophylaxis, and transmission blocking activity for malaria after a single oral administration. This remarkable multi-stage efficacy suggests that metabolic therapies, including cyt bc1 inhibitors, may be valuable additions to the collection of single-dose antimalarials in current development.


Asunto(s)
Antimaláricos/uso terapéutico , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Malaria Falciparum/tratamiento farmacológico , Éteres Fenílicos/uso terapéutico , Quinolonas/uso terapéutico , Animales , Antimaláricos/administración & dosificación , Resistencia a Medicamentos , Complejo III de Transporte de Electrones/metabolismo , Femenino , Ratones , Parasitemia/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Plasmodium yoelii/efectos de los fármacos
9.
Methods Mol Biol ; 1201: 177-91, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25388114

RESUMEN

Epigenetics is the study of heritable changes in gene expression that occur independent of the DNA sequence. Due to their intimacy with DNA, histones have a central role in chromatin structure and epigenetic regulation. Their tails are subject to posttranslational modifications (PTMs) that together with chromatin-remodeling proteins control the access of different proteins to DNA and allow a precise response to different environmental conditions. The first part of this chapter is dedicated to histone enrichment methods that allow the study of histones using techniques such as immunoblot or mass spectrometry for the mapping of the histone PTM network. Next we describe chromatin immunoprecipitation-based techniques (ChIP) for study of the epigenome. ChIP followed by microarray or next-generation sequencing enables the precise genomic localization of protein-DNA interactions. These techniques for genome-wide profiling of chromatin provide powerful and efficient tools to study the epigenome.


Asunto(s)
Epigénesis Genética , Epigenómica/métodos , Histonas/química , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Parásitos/genética , Animales , Cromatina , Inmunoprecipitación de Cromatina , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas/metabolismo
10.
PLoS Pathog ; 10(1): e1003830, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24391497

RESUMEN

Histone acetylation has been linked to developmental changes in gene expression and is a validated drug target of apicomplexan parasites, but little is known about the roles of individual histone modifying enzymes and how they are recruited to target genes. The protozoan parasite Toxoplasma gondii (phylum Apicomplexa) is unusual among invertebrates in possessing two GCN5-family lysine acetyltransferases (KATs). While GCN5a is required for gene expression in response to alkaline stress, this KAT is dispensable for parasite proliferation in normal culture conditions. In contrast, GCN5b cannot be disrupted, suggesting it is essential for Toxoplasma viability. To further explore the function of GCN5b, we generated clonal parasites expressing an inducible HA-tagged dominant-negative form of GCN5b containing a point mutation that ablates enzymatic activity (E703G). Stabilization of this dominant-negative GCN5b was mediated through ligand-binding to a destabilization domain (dd) fused to the protein. Induced accumulation of the ddHAGCN5b(E703G) protein led to a rapid arrest in parasite replication. Growth arrest was accompanied by a decrease in histone H3 acetylation at specific lysine residues as well as reduced expression of GCN5b target genes in GCN5b(E703G) parasites, which were identified using chromatin immunoprecipitation coupled with microarray hybridization (ChIP-chip). Proteomics studies revealed that GCN5b interacts with AP2-domain proteins, apicomplexan plant-like transcription factors, as well as a "core complex" that includes the co-activator ADA2-A, TFIID subunits, LEO1 polymerase-associated factor (Paf1) subunit, and RRM proteins. The dominant-negative phenotype of ddHAGCN5b(E703G) parasites, considered with the proteomics and ChIP-chip data, indicate that GCN5b plays a central role in transcriptional and chromatin remodeling complexes. We conclude that GCN5b has a non-redundant and indispensable role in regulating gene expression required during the Toxoplasma lytic cycle.


Asunto(s)
Proliferación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Histona Acetiltransferasas/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Sustitución de Aminoácidos , Estabilidad de Enzimas/fisiología , Histona Acetiltransferasas/genética , Histonas/genética , Histonas/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Mutación Missense , Proteómica/métodos , Proteínas Protozoarias/genética , Toxoplasma/genética , Factores de Transcripción/genética , Transcripción Genética/fisiología
11.
PLoS One ; 9(1): e84384, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416224

RESUMEN

Plasmodium parasites rely upon purine salvage for survival. Plasmodium purine nucleoside phosphorylase is part of the streamlined Plasmodium purine salvage pathway that leads to the phosphorylysis of both purines and 5'-methylthiopurines, byproducts of polyamine synthesis. We have explored structural features in Plasmodium falciparum purine nucleoside phosphorylase (PfPNP) that affect efficiency of catalysis as well as those that make it suitable for dual specificity. We used site directed mutagenesis to identify residues critical for PfPNP catalytic activity as well as critical residues within a hydrophobic pocket required for accommodation of the 5'-methylthio group. Kinetic analysis data shows that several mutants had disrupted binding of the 5'-methylthio group while retaining activity for inosine. A triple PfPNP mutant that mimics Toxoplasma gondii PNP had significant loss of 5'-methylthio activity with retention of inosine activity. Crystallographic investigation of the triple mutant PfPNP with Tyr160Phe, Val66Ile, andVal73Ile in complex with the transition state inhibitor immucillin H reveals fewer hydrogen bond interactions for the inhibitor in the hydrophobic pocket.


Asunto(s)
Metiltioinosina/análogos & derivados , Plasmodium falciparum/enzimología , Purina-Nucleósido Fosforilasa/química , Purina-Nucleósido Fosforilasa/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Cinética , Metiltioinosina/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/genética , Pirimidinonas/metabolismo , Pirimidinonas/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato
12.
Mol Microbiol ; 90(2): 338-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23964771

RESUMEN

Apicomplexa division involves several distinct phases shared with other eukaryote cell cycles including a gap period (G1) prior to chromosome synthesis, although how progression through the parasite cell cycle is controlled is not understood. Here we describe a cell cycle mutant that reversibly arrests in the G1 phase. The defect in this mutant was mapped by genetic complementation to a gene encoding a novel AAA-ATPase/CDC48 family member called TgNoAP1. TgNoAP1 is tightly regulated and expressed in the nucleolus during the G1/S phases. A tyrosine to a cysteine change upstream of the second AAA+ domain in the temperature sensitive TgNoAP1 allele leads to conditional protein instability, which is responsible for rapid cell cycle arrest and a primary defect in 28S rRNA processing as confirmed by knock-in of the mutation back into the parent genome. The interaction of TgNoAP1 with factors of the snoRNP and R2TP complexes indicates this protein has a role in pre-rRNA processing. This is a novel role for a cdc48-related chaperone protein and indicates that TgNoAP1 may be part of a dynamic mechanism that senses the health of the parasite protein machinery at the initial steps of ribosome biogenesis and conveys that information to the parasite cell cycle checkpoint controls.


Asunto(s)
Adenosina Trifosfatasas/genética , División Celular , Nucléolo Celular/enzimología , Puntos de Control de la Fase G1 del Ciclo Celular , Toxoplasma/citología , Toxoplasma/enzimología , Adenosina Trifosfatasas/metabolismo , Sustitución de Aminoácidos , Proteínas de Ciclo Celular/genética , Nucléolo Celular/ultraestructura , Cisteína/genética , Evolución Molecular , Regulación de la Expresión Génica , Prueba de Complementación Genética , Calor , Datos de Secuencia Molecular , Mutagénesis , Filogenia , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , ARN Ribosómico 28S/genética , Ribosomas/metabolismo , Toxoplasma/genética , Tirosina/genética , Proteína que Contiene Valosina
13.
PLoS Genet ; 9(2): e1003305, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23437009

RESUMEN

In the G1 phase of the cell division cycle, eukaryotic cells prepare many of the resources necessary for a new round of growth including renewal of the transcriptional and protein synthetic capacities and building the machinery for chromosome replication. The function of G1 has an early evolutionary origin and is preserved in single and multicellular organisms, although the regulatory mechanisms conducting G1 specific functions are only understood in a few model eukaryotes. Here we describe a new G1 mutant from an ancient family of apicomplexan protozoans. Toxoplasma gondii temperature-sensitive mutant 12-109C6 conditionally arrests in the G1 phase due to a single point mutation in a novel protein containing a single RNA-recognition-motif (TgRRM1). The resulting tyrosine to asparagine amino acid change in TgRRM1 causes severe temperature instability that generates an effective null phenotype for this protein when the mutant is shifted to the restrictive temperature. Orthologs of TgRRM1 are widely conserved in diverse eukaryote lineages, and the human counterpart (RBM42) can functionally replace the missing Toxoplasma factor. Transcriptome studies demonstrate that gene expression is downregulated in the mutant at the restrictive temperature due to a severe defect in splicing that affects both cell cycle and constitutively expressed mRNAs. The interaction of TgRRM1 with factors of the tri-SNP complex (U4/U6 & U5 snRNPs) indicate this factor may be required to assemble an active spliceosome. Thus, the TgRRM1 family of proteins is an unrecognized and evolutionarily conserved class of splicing regulators. This study demonstrates investigations into diverse unicellular eukaryotes, like the Apicomplexa, have the potential to yield new insights into important mechanisms conserved across modern eukaryotic kingdoms.


Asunto(s)
Empalme Alternativo/genética , Ciclo Celular/genética , ARN Mensajero , Proteínas de Unión al ARN , Toxoplasma , Secuencia Conservada/genética , Fase G1/genética , Regulación de la Expresión Génica , Humanos , Mutación , Motivos de Nucleótidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Homología de Secuencia de Aminoácido , Temperatura , Toxoplasma/genética , Toxoplasma/metabolismo
14.
Cancer Biother Radiopharm ; 27(9): 570-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23113595

RESUMEN

There is a need for radioprotectors that protect normal tissues from ionizing radiation in patients receiving high doses of radiation and during nuclear emergencies. We investigated the possibility of creating an efficient oral radioprotector based on the natural pigment melanin that would act as an internal shield and protect the tissues via Compton scattering followed by free radical scavenging. CD-1 mice were fed melanin-containing black edible mushrooms Auricularia auricila-judae before 9 Gy total body irradiation. The location of the mushrooms in the body before irradiation was determined by in vivo fluorescent imaging. Black mushrooms protected 80% of mice from the lethal dose, while control mice or those given melanin-devoid mushrooms died from gastrointestinal syndrome. The crypts of mice given black mushrooms showed less apoptosis and more cell division than those in control mice, and their white blood cell and platelet counts were restored at 45 days to preradiation levels. The role of melanin in radioprotection was proven by the fact that mice given white mushrooms supplemented with melanin survived at the same rate as mice given black mushrooms. The ability of melanin-containing mushrooms to provide remarkable protection against radiation suggests that they could be developed into oral radioprotectors.


Asunto(s)
Agaricales/química , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de la radiación , Melaninas/química , Melaninas/farmacología , Protectores contra Radiación/química , Protectores contra Radiación/farmacología , Animales , Antioxidantes/análisis , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Tracto Gastrointestinal/citología , Melaninas/análisis , Ratones , Irradiación Corporal Total
15.
Nat Methods ; 8(9): 771-7, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804536

RESUMEN

We report a photoswitchable monomeric Orange (PSmOrange) protein that is initially orange (excitation, 548 nm; emission, 565 nm) but becomes far-red (excitation, 636 nm; emission, 662 nm) after irradiation with blue-green light. Compared to its parental orange proteins, PSmOrange has greater brightness, faster maturation, higher photoconversion contrast and better photostability. The red-shifted spectra of both forms of PSmOrange enable its simultaneous use with cyan-to-green photoswitchable proteins to study four intracellular populations. Photoconverted PSmOrange has, to our knowledge, the most far-red excitation peak of all GFP-like fluorescent proteins, provides diffraction-limited and super-resolution imaging in the far-red light range, is optimally excited with common red lasers, and can be photoconverted subcutaneously in a mouse. PSmOrange photoswitching occurs via a two-step photo-oxidation process, which causes cleavage of the polypeptide backbone. The far-red fluorescence of photoconverted PSmOrange results from a new chromophore containing N-acylimine with a co-planar carbon-oxygen double bond.


Asunto(s)
Proteínas Luminiscentes/química , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Color , Femenino , Fluorescencia , Colorantes Fluorescentes/química , Colorantes Fluorescentes/efectos de la radiación , Células HEK293 , Células HeLa , Humanos , Luz , Proteínas Luminiscentes/efectos de la radiación , Ratones , Datos de Secuencia Molecular
16.
Nat Biotechnol ; 29(8): 757-61, 2011 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-21765402

RESUMEN

Imaging biological processes in mammalian tissues will be facilitated by fluorescent probes with excitation and emission bands within the near-infrared optical window of high transparency. Here we report a phytochrome-based near-infrared fluorescent protein (iRFP) with excitation and emission maxima at 690 nm and 713 nm, respectively. iRFP does not require an exogenous supply of the chromophore biliverdin and has higher effective brightness, intracellular stability and photostability than earlier phytochrome-derived fluorescent probes. Compared with far-red GFP-like proteins, iRFP has a substantially higher signal-to-background ratio in a mouse model due to its infrared-shifted spectra.


Asunto(s)
Diagnóstico por Imagen/métodos , Colorantes Fluorescentes/química , Proteínas Luminiscentes/química , Fitocromo/química , Adenoviridae/genética , Animales , Femenino , Citometría de Flujo , Colorantes Fluorescentes/metabolismo , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Fantasmas de Imagen , Estabilidad Proteica , Espectrometría de Fluorescencia
17.
J Infect Dis ; 200(9): 1470-8, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19811100

RESUMEN

BACKGROUND: The morbidity and mortality associated with malaria are heightened because of the spread of drug-resistant parasites and the lack of an effective vaccine. Plasmodium liver stages are the targets of new chemotherapeutics and vaccines, but there are limited tools available to study this stage in vivo. METHODS: To overcome this obstacle, we developed a method with which to study Plasmodium liver stages by means of bioluminescent imaging (BLI) of the rodent malaria parasite Plasmodium yoelii. We created a P. yoelii YM strain (PyLuc) that stably expresses firefly luciferase driven by a constitutive promoter. RESULTS: Using BLI, we performed imaging of the Plasmodium liver stages of mice infected with PyLuc sporozoites and monitored parasite dissemination during blood-stage infection. Because PyLuc luciferase activity is proportional to the number of parasites, BLI can be used to quantify the effect of drugs on liver-stage development. Moreover, using BLI, we demonstrated that immunization with blood-stage parasites confers partial protective immunity against the development of liver stages. CONCLUSIONS: BLI is a noninvasive technique that is useful for screening potential drugs and candidate vaccines with which to combat malaria. The prospect of cross-stage protective immunity increases the number of avenues to be explored in the development of an effective vaccine against malaria.


Asunto(s)
Hígado/parasitología , Malaria/parasitología , Plasmodium yoelii/aislamiento & purificación , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes , Luciferasas , Merozoítos , Ratones , Parasitemia , Plasmodium yoelii/crecimiento & desarrollo
18.
Biochemistry ; 48(40): 9618-26, 2009 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-19728741

RESUMEN

Plasmodium falciparum is a purine auxotroph requiring hypoxanthine as a key metabolic precursor. Erythrocyte adenine nucleotides are the source of the purine precursors, making adenosine deaminase (ADA) a key enzyme in the pathway of hypoxanthine formation. Methylthioadenosine (MTA) is a substrate for most malarial ADAs, but not for human ADA. The catalytic site specificity of malarial ADAs permits methylthiocoformycin (MT-coformycin) to act as a Plasmodium-specific transition state analogue with low affinity for human ADA [Tyler, P. C., Taylor, E. A., Frohlich, R. G. G., and Schramm, V. L. (2007) J. Am. Chem. Soc. 129, 6872-6879]. The structural basis for MTA and MT-coformycin specificity in malarial ADAs is the subject of speculation [Larson, E. T., et al. (2008) J. Mol. Biol. 381, 975-988]. Here, the crystal structure of ADA from Plasmodium vivax (PvADA) in a complex with MT-coformycin reveals an unprecedented binding geometry for 5'-methylthioribosyl groups in the malarial ADAs. Compared to malarial ADA complexes with adenosine or deoxycoformycin, 5'-methylthioribosyl groups are rotated 130 degrees . A hydrogen bonding network between Asp172 and the 3'-hydroxyl of MT-coformycin is essential for recognition of the 5'-methylthioribosyl group. Water occupies the 5'-hydroxyl binding site when MT-coformycin is bound. Mutagenesis of Asp172 destroys the substrate specificity for MTA and MT-coformycin. Kinetic, mutagenic, and structural analyses of PvADA and kinetic analysis of five other Plasmodium ADAs establish the unique structural basis for its specificity for MTA and MT-coformycin. Plasmodium gallinaceum ADA does not use MTA as a substrate, is not inhibited by MT-coformycin, and is missing Asp172. Treatment of P. falciparum cultures with coformycin or MT-coformycin in the presence of MTA is effective in inhibiting parasite growth.


Asunto(s)
Adenosina Desaminasa/metabolismo , Coformicina/análogos & derivados , Coformicina/química , Coformicina/metabolismo , Malaria Falciparum/enzimología , Plasmodium falciparum/enzimología , Adenosina Desaminasa/química , Animales , Antimaláricos/química , Antimaláricos/metabolismo , Antimaláricos/farmacología , Coformicina/farmacología , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/prevención & control , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Relación Estructura-Actividad , Especificidad por Sustrato
19.
J Biol Chem ; 283(51): 35899-907, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18957439

RESUMEN

Human malaria infections resulting from Plasmodium falciparum have become increasingly difficult to treat due to the emergence of drug-resistant parasites. The P. falciparum purine salvage enzyme purine nucleoside phosphorylase (PfPNP) is a potential drug target. Previous studies, in which PfPNP was targeted by transition state analogue inhibitors, found that those inhibiting human PNP and PfPNPs killed P. falciparum in vitro. However, many drugs have off-target interactions, and genetic evidence is required to demonstrate single target action for this class of potential drugs. We used targeted gene disruption in P. falciparum strain 3D7 to ablate PNP expression, yielding transgenic 3D7 parasites (Deltapfpnp). Lysates of the Deltapfpnp parasites showed no PNP activity, but activity of another purine salvage enzyme, adenosine deaminase (PfADA), was normal. When compared with wild-type 3D7, the Deltapfpnp parasites showed a greater requirement for exogenous purines and a severe growth defect at physiological concentrations of hypoxanthine. Drug assays using immucillins, specific transition state inhibitors of PNP, were performed on wild-type and Deltapfpnp parasites. The Deltapfpnp parasites were more sensitive to PNP inhibitors that bound hPNP tighter and less sensitive to MT-ImmH, an inhibitor with 100-fold preference for PfPNP over hPNP. The results demonstrate the importance of purine salvage in P. falciparum and validate PfPNP as the target of immucillins.


Asunto(s)
Malaria Falciparum/enzimología , Plasmodium falciparum/enzimología , Proteínas Protozoarias/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Animales , Animales Modificados Genéticamente , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Técnicas de Silenciamiento del Gen , Humanos , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/genética , Purinas/metabolismo , Pirimidinonas/farmacología , Pirimidinonas/uso terapéutico , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico
20.
Nat Med ; 14(9): 954-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18758447

RESUMEN

Malaria continues to devastate sub-Saharan Africa owing to the emergence of drug resistance to established antimalarials and to the lack of an efficacious vaccine. Plasmodium species have a unique streamlined purine pathway in which the dual specificity enzyme purine nucleoside phosphorylase (PNP) functions in both purine recycling and purine salvage. To evaluate the importance of PNP in an in vivo model of malaria, we disrupted PyPNP, the gene encoding PNP in the lethal Plasmodium yoelii YM strain. P. yoelii parasites lacking PNP were attenuated and cleared in mice. Although able to form gametocytes, PNP-deficient parasites did not form oocysts in mosquito midguts and were not transmitted from mosquitoes to mice. Mice given PNP-deficient parasites were immune to subsequent challenge to a lethal inoculum of P. yoelii YM and to challenge from P. yoelii 17XNL, another strain. These in vivo studies with PNP-deficient parasites support purine salvage as a target for antimalarials. They also suggest a strategy for the development of attenuated nontransmissible metabolic mutants as blood-stage malaria vaccine strains.


Asunto(s)
Vacunas contra la Malaria/genética , Malaria/tratamiento farmacológico , Plasmodium/enzimología , Plasmodium/genética , Purina-Nucleósido Fosforilasa/metabolismo , Animales , Clonación Molecular , Cartilla de ADN/genética , Femenino , Estadios del Ciclo de Vida/inmunología , Malaria/parasitología , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Fenotipo , Purina-Nucleósido Fosforilasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...