Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 21(10): e3002341, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37883333

RESUMEN

There is a growing appreciation that the direct interaction between bacteriophages and the mammalian host can facilitate diverse and unexplored symbioses. Yet the impact these bacteriophages may have on mammalian cellular and immunological processes is poorly understood. Here, we applied highly purified phage T4, free from bacterial by-products and endotoxins to mammalian cells and analyzed the cellular responses using luciferase reporter and antibody microarray assays. Phage preparations were applied in vitro to either A549 lung epithelial cells, MDCK-I kidney cells, or primary mouse bone marrow derived macrophages with the phage-free supernatant serving as a comparative control. Highly purified T4 phages were rapidly internalized by mammalian cells and accumulated within macropinosomes but did not activate the inflammatory DNA response TLR9 or cGAS-STING pathways. Following 8 hours of incubation with T4 phage, whole cell lysates were analyzed via antibody microarray that detected expression and phosphorylation levels of human signaling proteins. T4 phage application led to the activation of AKT-dependent pathways, resulting in an increase in cell metabolism, survival, and actin reorganization, the last being critical for macropinocytosis and potentially regulating a positive feedback loop to drive further phage internalization. T4 phages additionally down-regulated CDK1 and its downstream effectors, leading to an inhibition of cell cycle progression and an increase in cellular growth through a prolonged G1 phase. These interactions demonstrate that highly purified T4 phages do not activate DNA-mediated inflammatory pathways but do trigger protein phosphorylation cascades that promote cellular growth and survival. We conclude that mammalian cells are internalizing bacteriophages as a resource to promote cellular growth and metabolism.


Asunto(s)
Anticuerpos , Bacteriófago T4 , Animales , Ratones , Humanos , Bacteriófago T4/genética , Ciclo Celular , ADN , Mamíferos/genética
2.
EMBO Rep ; 23(2): e54022, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34866326

RESUMEN

PfCDPK7 is an atypical member of the calcium-dependent protein kinase (CDPK) family and is crucial for the development of Plasmodium falciparum. However, the mechanisms whereby PfCDPK7 regulates parasite development remain unknown. Here, we perform quantitative phosphoproteomics and phospholipid analysis and find that PfCDPK7 promotes phosphatidylcholine (PC) synthesis by regulating two key enzymes involved in PC synthesis, phosphoethanolamine-N-methyltransferase (PMT) and ethanolamine kinase (EK). In the absence of PfCDPK7, both enzymes are hypophosphorylated and PMT is degraded. We further find that PfCDPK7 interacts with 4'-phosphorylated phosphoinositides (PIPs) generated by PI4-kinase. Inhibition of PI4K activity disrupts the vesicular localization PfCDPK7. P. falciparum PI4-kinase, PfPI4K is a prominent drug target and one of its inhibitors, MMV39048, has reached Phase I clinical trials. Using this inhibitor, we demonstrate that PfPI4K controls phospholipid biosynthesis and may act in part by regulating PfCDPK7 localization and activity. These studies not only unravel a signaling pathway involving PfPI4K/4'-PIPs and PfCDPK7 but also provide novel insights into the mechanism of action of a promising series of candidate anti-malarial drugs.


Asunto(s)
Malaria Falciparum , Plasmodium falciparum , Humanos , Fosfolípidos/metabolismo , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Transducción de Señal
3.
BMC Genomics ; 23(1): 237, 2022 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-35346035

RESUMEN

BACKGROUND: Novel antimalarials should be effective across all species of malaria parasites that infect humans, especially the two species that bear the most impact, Plasmodium falciparum and Plasmodium vivax. Protein kinases encoded by pathogens, as well as host kinases required for survival of intracellular pathogens, carry considerable potential as targets for antimalarial intervention (Adderley et al. Trends Parasitol 37:508-524, 2021;  Wei et al. Cell Rep Med 2:100423, 2021). To date, no comprehensive P. vivax kinome assembly has been conducted; and the P. falciparum kinome, first assembled in 2004, requires an update. The present study, aimed to fill these gaps, utilises a recently published structurally-validated multiple sequence alignment (MSA) of the human kinome (Modi et al. Sci Rep 9:19790, 2019). This MSA is used as a scaffold to assist the alignment of all protein kinase sequences from P. falciparum and P. vivax, and (where possible) their assignment to specific kinase groups/families. RESULTS: We were able to assign six P. falciparum previously classified as OPK or 'orphans' (i.e. with no clear phylogenetic relation to any of the established ePK groups) to one of the aforementioned ePK groups. Direct phylogenetic comparison established that despite an overall high level of similarity between the P. falciparum and P. vivax kinomes, which will help in selecting targets for intervention, there are differences that may underlie the biological specificities of these species. Furthermore, we highlight a number of Plasmodium kinases that have a surprisingly high level of similarity with their human counterparts and therefore not well suited as targets for drug discovery. CONCLUSIONS: Direct comparison of the kinomes of Homo sapiens, P. falciparum and P. vivax sheds additional light on the previously documented divergence of many P. falciparum and P. vivax kinases from those of their human host. We provide the first direct kinome comparison between the phylogenetically distinct species of P. falciparum and P. vivax, illustrating the key similarities and differences which must be considered in the context of kinase-directed antimalarial drug discovery, and discuss the divergences and similarities between the human and Plasmodium kinomes to inform future searches for selective antimalarial intervention.


Asunto(s)
Malaria Falciparum , Malaria Vivax , Humanos , Malaria Falciparum/parasitología , Malaria Vivax/parasitología , Filogenia , Plasmodium falciparum/genética , Plasmodium vivax/genética
4.
PLoS Biol ; 17(6): e3000308, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31181082

RESUMEN

Plasmodium falciparum is the main cause of disease and death from malaria. P. falciparum virulence resides in the ability of infected erythrocytes (IEs) to sequester in various tissues through the interaction between members of the polymorphic P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesin family to various host receptors. Here, we investigated the effect of phosphorylation of variant surface antigen 2-CSA (VAR2CSA), a member of the PfEMP1 family associated to placental sequestration, on its capacity to adhere to chondroitin sulfate A (CSA) present on the placental syncytium. We showed that phosphatase treatment of IEs impairs cytoadhesion to CSA. MS analysis of recombinant VAR2CSA phosphosites prior to and after phosphatase treatment, as well as of native VAR2CSA expressed on IEs, identified critical phosphoresidues associated with CSA binding. Site-directed mutagenesis on recombinant VAR2CSA of 3 phosphoresidues localised within the CSA-binding region confirmed in vitro their functional importance. Furthermore, using clustered regularly interspaced short palindromic repeats/CRISPR-associated protein-9 nuclease (CRISPR/Cas9), we generated a parasite line in which the phosphoresidue T934 is changed to alanine and showed that this mutation strongly impairs IEs cytoadhesion to CSA. Taken together, these results demonstrate that phosphorylation of the extracellular region of VAR2CSA plays a major role in IEs cytoadhesion to CSA and provide new molecular insights for strategies aiming to reduce the morbidity and mortality of PM.


Asunto(s)
Antígenos de Protozoos/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Animales , Variación Antigénica , Antígenos de Protozoos/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Eritrocitos/parasitología , Femenino , Humanos , Malaria , Malaria Falciparum/genética , Malaria Falciparum/parasitología , Parásitos , Fosforilación , Placenta , Plasmodium falciparum/genética , Embarazo , Unión Proteica
5.
IUBMB Life ; 72(6): 1243-1249, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32356940

RESUMEN

A pool of Plasmodium falciparum casein kinase 1 (PfCK1) has been shown to localize to the host red blood cell (RBC) membrane and be secreted to the extracellular medium during trophozoite stage of development. We attempted to identify mechanisms for secretion of PfCK1 and its appearance on the RBC membrane. We found that two host proteins with established functions in membrane trafficking in higher eukaryotes, GTPase-activating protein and Vps9 domain-containing protein 1 (GAPVD1), and Sorting nexin 22, consistently co-purify with PfCK1, suggesting that the parasite utilizes trafficking pathways previously thought to be inactive in RBCs. Furthermore, reciprocal immunoprecipitation experiments with GAPVD1 identified parasite proteins suggestive of a protein recycling pathway hitherto only described in higher eukaryotes. Thus, we have identified components of a trafficking pathway involving parasite proteins that act in concert with host proteins, and which we hypothesize mediates trafficking of PfCK1 to the RBC during infection.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Interacciones Huésped-Patógeno/fisiología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Quinasa de la Caseína I/genética , Membrana Celular/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Espectrometría de Masas , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Transporte de Proteínas , Proteínas Protozoarias/genética , Nexinas de Clasificación/metabolismo
6.
Chem Rev ; 118(11): 5330-5358, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29676564

RESUMEN

New technologies to diagnose malaria at high sensitivity and specificity are urgently needed in the developing world where the disease continues to pose a huge burden on society. Infrared and Raman spectroscopy-based diagnostic methods have a number of advantages compared with other diagnostic tests currently on the market. These include high sensitivity and specificity for detecting low levels of parasitemia along with ease of use and portability. Here, we review the application of vibrational spectroscopic techniques for monitoring and detecting malaria infection. We discuss the role of vibrational (infrared and Raman) spectroscopy in understanding the processes of parasite biology and its application to the study of interactions with antimalarial drugs. The distinct molecular phenotype that characterizes malaria infection and the high sensitivity enabling detection of low parasite densities provides a genuine opportunity for vibrational spectroscopy to become a front-line tool in the elimination of this deadly disease and provide molecular insights into the chemistry of this unique organism.


Asunto(s)
Malaria/diagnóstico , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Espectrometría Raman/métodos , Animales , Eritrocitos/microbiología , Eritrocitos/patología , Hemo/análisis , Hemoproteínas/análisis , Humanos , Plasmodium/crecimiento & desarrollo , Espectroscopía Infrarroja por Transformada de Fourier/instrumentación , Espectrometría Raman/instrumentación , Vibración
7.
Nucleic Acids Res ; 46(D1): D1091-D1106, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29149325

RESUMEN

The IUPHAR/BPS Guide to PHARMACOLOGY (GtoPdb, www.guidetopharmacology.org) and its precursor IUPHAR-DB, have captured expert-curated interactions between targets and ligands from selected papers in pharmacology and drug discovery since 2003. This resource continues to be developed in conjunction with the International Union of Basic and Clinical Pharmacology (IUPHAR) and the British Pharmacological Society (BPS). As previously described, our unique model of content selection and quality control is based on 96 target-class subcommittees comprising 512 scientists collaborating with in-house curators. This update describes content expansion, new features and interoperability improvements introduced in the 10 releases since August 2015. Our relationship matrix now describes ∼9000 ligands, ∼15 000 binding constants, ∼6000 papers and ∼1700 human proteins. As an important addition, we also introduce our newly funded project for the Guide to IMMUNOPHARMACOLOGY (GtoImmuPdb, www.guidetoimmunopharmacology.org). This has been 'forked' from the well-established GtoPdb data model and expanded into new types of data related to the immune system and inflammatory processes. This includes new ligands, targets, pathways, cell types and diseases for which we are recruiting new IUPHAR expert committees. Designed as an immunopharmacological gateway, it also has an emphasis on potential therapeutic interventions.


Asunto(s)
Bases de Datos Farmacéuticas , Fenómenos del Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Animales , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Ligandos , Farmacología , Proteínas/efectos de los fármacos
8.
Malar J ; 18(1): 348, 2019 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-31619246

RESUMEN

BACKGROUND: Widespread elimination of malaria requires an ultra-sensitive detection method that can detect low parasitaemia levels seen in asymptomatic carriers who act as reservoirs for further transmission of the disease, but is inexpensive and easy to deploy in the field in low income settings. It was hypothesized that a new method of malaria detection based on infrared spectroscopy, shown in the laboratory to have similar sensitivity to PCR based detection, could prove effective in detecting malaria in a field setting using cheap portable units with data management systems allowing them to be used by users inexpert in spectroscopy. This study was designed to determine whether the methodology developed in the laboratory could be translated to the field to diagnose the presence of Plasmodium in the blood of patients presenting at hospital with symptoms of malaria, as a precursor to trials testing the sensitivity of to detect asymptomatic carriers. METHODS: The field study tested 318 patients presenting with suspected malaria at four regional clinics in Thailand. Two portable infrared spectrometers were employed, operated from a laptop computer or a mobile telephone with in-built software that guided the user through the simple measurement steps. Diagnostic modelling and validation testing using linear and machine learning approaches was performed against the gold standard qPCR. Sample spectra from 318 patients were used for building calibration models (112 positive and 110 negative samples according to PCR testing) and independent validation testing (39 positive and 57 negatives samples by PCR). RESULTS: The machine learning classification (support vector machines; SVM) performed with 92% sensitivity (3 false negatives) and 97% specificity (2 false positives). The Area Under the Receiver Operation Curve (AUROC) for the SVM classification was 0.98. These results may be better than as stated as one of the spectroscopy false positives was infected by a Plasmodium species other than Plasmodium falciparum or Plasmodium vivax, not detected by the PCR primers employed. CONCLUSIONS: In conclusion, it was demonstrated that ATR-FTIR spectroscopy could be used as an efficient and reliable malaria diagnostic tool and has the potential to be developed for use at point of care under tropical field conditions with spectra able to be analysed via a Cloud-based system, and the diagnostic results returned to the user's mobile telephone or computer. The combination of accessibility to mass screening, high sensitivity and selectivity, low logistics requirements and portability, makes this new approach a potentially outstanding tool in the context of malaria elimination programmes. The next step in the experimental programme now underway is to reduce the sample requirements to fingerprick volumes.


Asunto(s)
Nube Computacional , Pruebas Diagnósticas de Rutina/métodos , Manejo de la Enfermedad , Malaria/diagnóstico , Espectrofotometría Infrarroja/métodos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Tailandia , Adulto Joven
9.
Malar J ; 18(1): 89, 2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30898128

RESUMEN

BACKGROUND: Malaria is one of the most prevalent tropical infectious diseases. Since recently cases of artemisinin resistance were reported, novel anti-malarial drugs are required which differ from artemisinins in structure and biological target. The plasmodial glycogen synthase kinase-3 (PfGSK-3) was suggested as a new anti-malarial drug target. 4-Phenylthieno[2,3-b]pyridines were previously identified as selective PfGSK-3 inhibitors with antiplasmodial activity. The present study aims at identifying a molecular position on this scaffold for the attachment of side chains in order to improve solubility and antiplasmodial activity. Furthermore, the role of axial chirality in the compound class for antiplasmodial activity and PfGSK-3 inhibition was investigated. METHODS: 4-Phenylthieno[2,3-b]pyridines with substituents in 4-position of the phenyl ring were docked into the ATP binding site of PfGSK-3. The compounds were synthesized employing a Thorpe reaction as final step. The enantiomers of one congener were separated by chiral HPLC. All derivatives were tested for inhibition of asexual erythrocytic stages of transgenic NF54-luc Plasmodium falciparum. Selected compounds with promising antiplasmodial activity were further evaluated for inhibition of HEK293 cells as well as inhibition of isolated PfGSK-3 and HsGSK-3. The kinetic aqueous solubility was assessed by laser nephelometry. RESULTS: The para position at the 4-phenyl ring of the title compounds was identified as a suitable point for the attachment of side chains. While alkoxy substituents in this position led to decreased antiplasmodial activity, alkylamino groups retained antiparasitic potency. The most promising of these congeners (4h) was investigated in detail. This compound is a selective PfGSK-3 inhibitor (versus the human GSK-3 orthologue), and exhibits improved antiplasmodial activity in vitro as well as better solubility in aqueous media than its unsubstituted parent structure. The derivative 4b was separated into the atropisomers, and it was shown that the (+)-enantiomer acts as eutomer. CONCLUSIONS: The attachment of alkylamino side chains leads to the improvement of antiplasmodial activity and aqueous solubility of selective PfGSK-inhibitors belonging to the class of 4-phenylthieno[2,3-b]pyridines. These molecules show axial chirality, a feature of high impact for biological activity. The findings can be exploited for the development of improved selective PfGSK-3 inhibitors.


Asunto(s)
Antimaláricos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Malaria Falciparum/prevención & control , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/antagonistas & inhibidores , Piridinas/farmacología , Células HEK293 , Humanos , Relación Estructura-Actividad
10.
J Proteome Res ; 17(6): 2112-2123, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29678115

RESUMEN

PfPK7 is an "orphan" kinase displaying regions of homology to multiple protein kinase families. PfPK7 functions in regulating parasite proliferation/development as evident from the phenotype analysis of knockout parasites. Despite this regulatory role, the functions of PfPK7 in signaling pathways are not known. To better understand PfPK7-regulated phosphorylation events, we performed isobaric tag-based quantitative comparative phosphoproteomics of the schizont and segmenter stages from wild-type and pfpk7 - parasite lines. This analysis identified 3,875 phosphorylation sites on 1,047 proteins. Among these phosphorylation events, 146 proteins with 239 phosphorylation sites displayed reduction in phosphorylation in the absence of PfPK7. Further analysis of the phosphopeptides revealed three motifs whose phosphorylation was down regulated in the pfpk7 - cell line in both schizonts and segmenters. Decreased phosphorylation following loss of PfPK7 indicates that these proteins may function as direct substrates of PfPK7. We demonstrated that PfPK7 is active toward three of these potential novel substrates; however, PfPK7 did not phosphorylate many of the other proteins, suggesting that decreased phosphorylation in these proteins is an indirect effect. Our phosphoproteomics analysis is the first study to identify direct substrates of PfPK7 and reveals potential downstream or compensatory signaling pathways.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/deficiencia , Fosfoproteínas/metabolismo , Fosforilación , Plasmodium falciparum/química , Plasmodium falciparum/enzimología , Proteínas Quinasas , Proteínas Protozoarias/fisiología , Esquizontes/química , Esquizontes/metabolismo , Transducción de Señal , Especificidad por Sustrato
11.
Anal Chem ; 90(5): 3140-3148, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29327915

RESUMEN

Atomic force microscopy-infrared (AFM-IR) spectroscopy is a powerful new technique that can be applied to study molecular composition of cells and tissues at the nanoscale. AFM-IR maps are acquired using a single wavenumber value: they show either the absorbance plotted against a single wavenumber value or a ratio of two absorbance values. Here, we implement multivariate image analysis to generate multivariate AFM-IR maps and use this approach to resolve subcellular structural information in red blood cells infected with Plasmodium falciparum at different stages of development. This was achieved by converting the discrete spectral points into a multispectral line spectrum prior to multivariate image reconstruction. The approach was used to generate compositional maps of subcellular structures in the parasites, including the food vacuole, lipid inclusions, and the nucleus, on the basis of the intensity of hemozoin, hemoglobin, lipid, and DNA IR marker bands, respectively. Confocal Raman spectroscopy was used to validate the presence of hemozoin in the regions identified by the AFM-IR technique. The high spatial resolution of AFM-IR combined with hyperspectral modeling enables the direct detection of subcellular components, without the need for cell sectioning or immunological/biochemical staining. Multispectral-AFM-IR thus has the capacity to probe the phenotype of the malaria parasite during its intraerythrocytic development. This enables novel approaches to studying the mode of action of antimalarial drugs and the phenotypes of drug-resistant parasites, thus contributing to the development of diagnostic and control measures.


Asunto(s)
Eritrocitos/metabolismo , Microscopía de Fuerza Atómica/métodos , Plasmodium falciparum/metabolismo , Espectrofotometría Infrarroja/métodos , Eritrocitos/parasitología , Hemoproteínas/análisis , Microscopía Confocal/métodos , Plasmodium falciparum/química , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/ultraestructura , Espectrometría Raman/métodos
12.
Genome Res ; 24(10): 1676-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25015382

RESUMEN

Global production of chickens has trebled in the past two decades and they are now the most important source of dietary animal protein worldwide. Chickens are subject to many infectious diseases that reduce their performance and productivity. Coccidiosis, caused by apicomplexan protozoa of the genus Eimeria, is one of the most important poultry diseases. Understanding the biology of Eimeria parasites underpins development of new drugs and vaccines needed to improve global food security. We have produced annotated genome sequences of all seven species of Eimeria that infect domestic chickens, which reveal the full extent of previously described repeat-rich and repeat-poor regions and show that these parasites possess the most repeat-rich proteomes ever described. Furthermore, while no other apicomplexan has been found to possess retrotransposons, Eimeria is home to a family of chromoviruses. Analysis of Eimeria genes involved in basic biology and host-parasite interaction highlights adaptations to a relatively simple developmental life cycle and a complex array of co-expressed surface proteins involved in host cell binding.


Asunto(s)
Eimeria/genética , Genoma de Protozoos , Proteínas Protozoarias/genética , Animales , Línea Celular , Pollos , Mapeo Cromosómico , Coccidiosis/parasitología , Coccidiosis/veterinaria , Eimeria/clasificación , Perfilación de la Expresión Génica , Filogenia , Enfermedades de las Aves de Corral/parasitología , Proteoma , Sintenía
13.
PLoS Pathog ; 11(12): e1005343, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26694741

RESUMEN

The most severe form of malaria in humans is caused by the protozoan parasite Plasmodium falciparum. The invasive form of malaria parasites is termed a merozoite and it employs an array of parasite proteins that bind to the host cell to mediate invasion. In Plasmodium falciparum, the erythrocyte binding-like (EBL) and reticulocyte binding-like (Rh) protein families are responsible for binding to specific erythrocyte receptors for invasion and mediating signalling events that initiate active entry of the malaria parasite. Here we have addressed the role of the cytoplasmic tails of these proteins in activating merozoite invasion after receptor engagement. We show that the cytoplasmic domains of these type 1 membrane proteins are phosphorylated in vitro. Depletion of PfCK2, a kinase implicated to phosphorylate these cytoplasmic tails, blocks P. falciparum invasion of red blood cells. We identify the crucial residues within the PfRh4 cytoplasmic domain that are required for successful parasite invasion. Live cell imaging of merozoites from these transgenic mutants show they attach but do not penetrate erythrocytes implying the PfRh4 cytoplasmic tail conveys signals important for the successful completion of the invasion process.


Asunto(s)
Eritrocitos/microbiología , Malaria Falciparum/metabolismo , Fosfotransferasas/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Humanos , Merozoítos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Plasmodium falciparum/patogenicidad
14.
Cell Microbiol ; 18(8): 1106-1120, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26833682

RESUMEN

Aurora kinases are eukaryotic serine/threonine protein kinases that regulate key events associated with chromatin condensation, centrosome and spindle function and cytokinesis. Elucidating the roles of Aurora kinases in apicomplexan parasites is crucial to understand the cell cycle control during Plasmodium schizogony or Toxoplasma endodyogeny. Here, we report on the localization of two previously uncharacterized Toxoplasma Aurora-related kinases (Ark2 and Ark3) in tachyzoites and of the uncharacterized Ark3 orthologue in Plasmodium falciparum erythrocytic stages. In Toxoplasma gondii, we show that TgArk2 and TgArk3 concentrate at specific sub-cellular structures linked to parasite division: the mitotic spindle and intranuclear mitotic structures (TgArk2), and the outer core of the centrosome and the budding daughter cells cytoskeleton (TgArk3). By tagging the endogenous PfArk3 gene with the green fluorescent protein in live parasites, we show that PfArk3 protein expression peaks late in schizogony and localizes at the periphery of budding schizonts. Disruption of the TgArk2 gene reveals no essential function for tachyzoite propagation in vitro, which is surprising giving that the P. falciparum and P. berghei orthologues are essential for erythrocyte schizogony. In contrast, knock-down of TgArk3 protein results in pronounced defects in parasite division and a major growth deficiency. TgArk3-depleted parasites display several defects, such as reduced parasite growth rate, delayed egress and parasite duplication, defect in rosette formation, reduced parasite size and invasion efficiency and lack of virulence in mice. Our study provides new insights into cell cycle control in Toxoplasma and malaria parasites and highlights Aurora kinase 3 as potential drug target.


Asunto(s)
Aurora Quinasas/fisiología , Proteínas Protozoarias/fisiología , Toxoplasma/enzimología , Toxoplasmosis/parasitología , Animales , Femenino , Interacciones Huésped-Parásitos , Ratones , Transporte de Proteínas , Toxoplasma/fisiología , Toxoplasma/ultraestructura , Virulencia
15.
Biochim Biophys Acta ; 1854(10 Pt B): 1637-43, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25770683

RESUMEN

With respect to parasite-induced infectious diseases of worldwide importance, members of the genera Plasmodium and Schistosoma are top pathogens. Nearly half a billion people suffer from malaria caused by Plasmodium spp. and schistosomiasis (bilharzia) induced by Schistosoma spp. Resistance against essentially all drugs used for malaria treatment has been reported. For schistosomiasis justified fear of upcoming resistance is discussed against the background of only one widely used drug for treatment. Research of the recent decade has demonstrated that essential steps of the biology of these and other parasites are controlled by kinases, which represent attractive targets for new-generation antiparasitic compounds. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Asunto(s)
Malaria/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Esquistosomiasis/tratamiento farmacológico , Animales , Antiparasitarios/química , Antiparasitarios/uso terapéutico , Humanos , Malaria/enzimología , Malaria/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Plasmodium falciparum/patogenicidad , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/química , Schistosoma/efectos de los fármacos , Schistosoma/enzimología , Schistosoma/patogenicidad , Esquistosomiasis/enzimología , Esquistosomiasis/parasitología
16.
Biochim Biophys Acta ; 1854(10 Pt B): 1657-62, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25896387

RESUMEN

Hepatitis C virus (HCV) infection relies on virus-host interactions with human hepatocytes, a context in which host cell kinases play critical roles in every step of the HCV life cycle. During viral entry, cellular kinases, including EGFR, EphA2 and PKA, regulate the localization of host HCV entry factors and induce receptor complex assembly. Following virion internalization, viral genomes replicate on endoplasmic reticulum-derived membranous webs. The formation of membranous webs depends on interactions between the HCV NS5a protein and PI4KIIIα. The phosphorylation status of NS5a, regulated by PI4KIIIα, CKI and other kinases, also acts as a molecular switch to virion assembly, which takes place on lipid droplets. The formation of lipid droplets is enhanced by HCV activation of IKKα. In view of the multiple crucial steps in the viral life cycle that are mediated by host cell kinases, these enzymes also represent complementary targets for antiviral therapy. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Asunto(s)
Receptores ErbB/metabolismo , Hepacivirus/genética , Hepatitis C/genética , Interacciones Huésped-Patógeno/genética , Animales , Receptores ErbB/genética , Hepacivirus/patogenicidad , Hepatitis C/virología , Hepatocitos/enzimología , Hepatocitos/virología , Humanos , Estadios del Ciclo de Vida/genética , Antígenos de Histocompatibilidad Menor , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptor EphA2/genética , Proteínas no Estructurales Virales/genética , Ensamble de Virus/genética , Internalización del Virus
17.
Mol Microbiol ; 98(1): 17-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26094711

RESUMEN

Plasmodium falciparum Origin Recognition Complex subunit 1 (PfORC1) has been implicated in DNA replication and var gene regulation. While the C-terminus is involved in DNA replication, the specific role of N-terminus has been suggested in var gene regulation in a Sir2-dependent manner. PfORC1 is localized at the nuclear periphery, where the clustering of chromosomal ends at the early stage of parasite development may be crucial for the regulation of subtelomeric var gene expression. Upon disassembly of telomeric clusters at later stages of parasite development, ORC1 is distributed in the nucleus and parasite cytoplasm where it may be required for its other cellular functions including DNA replication. The level of ORC1 decreases dramatically at the late schizont stage. The mechanisms that mediate regulation of PfORC1 function are largely unknown. Here we show, by the use of recombinant proteins and of transgenic parasites expressing wild type or mutant forms of ORC1, that phosphorylation of the PfORC1-N terminal domain by the cyclin-dependent kinase (CDK) PfPK5 abolishes DNA-binding activity and leads to changes in subcellular localization and proteasome-mediated degradation of the protein in schizonts. These results reveal that PfORC1 phosphorylation by a CDK is central to the regulation of important biological functions like DNA replication and var gene silencing.


Asunto(s)
Complejo de Reconocimiento del Origen/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Replicación del ADN , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Complejo de Reconocimiento del Origen/genética , Fosforilación , Fosfotransferasas , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Esquizontes/fisiología
18.
Malar J ; 15(1): 535, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27821169

RESUMEN

BACKGROUND: Examining essential biochemical pathways in Plasmodium falciparum presents serious challenges, as standard molecular techniques such as siRNA cannot be employed in this organism, and generating gene knock-outs of essential proteins requires specialized conditional approaches. In the study of protein kinases, pharmacological inhibition presents a feasible alternative option. However, as in mammalian systems, inhibitors often lack the desired selectivity. Described here is a chemical genetic approach to selectively inhibit Pfnek-2 in P. falciparum, a member of the NIMA-related kinase family that is essential for completion of the sexual development of the parasite. RESULTS: Introduction of a valine to cysteine mutation at position 24 in the glycine rich loop of Pfnek-2 does not affect kinase activity but confers sensitivity to the protein kinase inhibitor 4-(6-ethynyl-9H-purin-2-ylamino) benzene sulfonamide (NCL-00016066). Using a combination of in vitro kinase assays and mass spectrometry, (including phosphoproteomics) the study shows that this compound acts as an irreversible inhibitor to the mutant Pfnek2 likely through a covalent link with the introduced cysteine residue. In particular, this was shown by analysis of total protein mass using mass spectrometry which showed a shift in molecular weight of the mutant kinase in the presence of the inhibitor to be precisely equivalent to the molecular weight of NCL-00016066. A similar molecular weight shift was not observed in the wild type kinase. Importantly, this inhibitor has little activity towards the wild type Pfnek-2 and, therefore, has all the properties of an effective chemical genetic tool that could be employed to determine the cellular targets for Pfnek-2. CONCLUSIONS: Allelic replacement of wild-type Pfnek-2 with the mutated kinase will allow for targeted inhibition of Pfnek-2 with NCL-00016066 and hence pave the way for comparative studies aimed at understanding the biological role and transmission-blocking potential of Pfnek-2.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Proteínas Mutantes/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Plasmodium falciparum/enzimología , Purinas/metabolismo , Sulfonamidas/metabolismo , Espectrometría de Masas , Proteínas Mutantes/genética , Quinasas Relacionadas con NIMA/genética
19.
J Biol Chem ; 289(29): 20386-95, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24895132

RESUMEN

Second messengers such as phosphoinositides and calcium are known to control diverse processes involved in the development of malaria parasites. However, the underlying molecular mechanisms and pathways need to be unraveled, which may be achieved by understanding the regulation of effectors of these second messengers. Calcium-dependent protein kinase (CDPK) family members regulate diverse parasitic processes. Because CDPKs are absent from the host, these kinases are considered as potential drug targets. We have dissected the function of an atypical CDPK from Plasmodium falciparum, PfCDPK7. The domain architecture of PfCDPK7 is very different from that of other CDPKs; it has a pleckstrin homology domain adjacent to the kinase domain and two calcium-binding EF-hands at its N terminus. We demonstrate that PfCDPK7 interacts with PI(4,5)P2 via its pleckstrin homology domain, which may guide its subcellular localization. Disruption of PfCDPK7 caused a marked reduction in the growth of the blood stage parasites, as maturation of rings to trophozoites was markedly stalled. In addition, parasite proliferation was significantly attenuated. These findings shed light on an important role for PfCDPK7 in the erythrocytic asexual cycle of malaria parasites.


Asunto(s)
Plasmodium falciparum/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Quinasas/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Animales Modificados Genéticamente , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Técnicas de Inactivación de Genes , Genes Protozoarios , Fosfatidilinositol 4,5-Difosfato/metabolismo , Plasmodium falciparum/genética , Dominios y Motivos de Interacción de Proteínas , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sistemas de Mensajero Secundario
20.
Proc Natl Acad Sci U S A ; 109(47): E3278-87, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23112171

RESUMEN

The human malaria parasite Plasmodium falciparum is auxotrophic for most amino acids. Its amino acid needs are met largely through the degradation of host erythrocyte hemoglobin; however the parasite must acquire isoleucine exogenously, because this amino acid is not present in adult human hemoglobin. We report that when isoleucine is withdrawn from the culture medium of intraerythrocytic P. falciparum, the parasite slows its metabolism and progresses through its developmental cycle at a reduced rate. Isoleucine-starved parasites remain viable for 72 h and resume rapid growth upon resupplementation. Protein degradation during starvation is important for maintenance of this hibernatory state. Microarray analysis of starved parasites revealed a 60% decrease in the rate of progression through the normal transcriptional program but no other apparent stress response. Plasmodium parasites do not possess a TOR nutrient-sensing pathway and have only a rudimentary amino acid starvation-sensing eukaryotic initiation factor 2α (eIF2α) stress response. Isoleucine deprivation results in GCN2-mediated phosphorylation of eIF2α, but kinase-knockout clones still are able to hibernate and recover, indicating that this pathway does not directly promote survival during isoleucine starvation. We conclude that P. falciparum, in the absence of canonical eukaryotic nutrient stress-response pathways, can cope with an inconsistent bloodstream amino acid supply by hibernating and waiting for more nutrient to be provided.


Asunto(s)
Hibernación , Isoleucina/deficiencia , Plasmodium falciparum/metabolismo , Animales , Artemisininas/farmacología , Carbono/metabolismo , Factor 2B Eucariótico de Iniciación/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Genes Protozoarios/genética , Hibernación/efectos de los fármacos , Humanos , Metaboloma/efectos de los fármacos , Parásitos/efectos de los fármacos , Parásitos/genética , Parásitos/crecimiento & desarrollo , Péptido Hidrolasas/metabolismo , Fenotipo , Fosforilación/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Proteolisis/efectos de los fármacos , Proteínas Protozoarias/metabolismo , Inanición
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA