Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
PLoS Pathog ; 13(10): e1006659, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28985225

RESUMEN

Transmission of malaria parasites relies on the formation of a specialized blood form called the gametocyte. Gametocytes of the human pathogen, Plasmodium falciparum, adopt a crescent shape. Their dramatic morphogenesis is driven by the assembly of a network of microtubules and an underpinning inner membrane complex (IMC). Using super-resolution optical and electron microscopies we define the ultrastructure of the IMC at different stages of gametocyte development. We characterize two new proteins of the gametocyte IMC, called PhIL1 and PIP1. Genetic disruption of PhIL1 or PIP1 ablates elongation and prevents formation of transmission-ready mature gametocytes. The maturation defect is accompanied by failure to form an enveloping IMC and a marked swelling of the digestive vacuole, suggesting PhIL1 and PIP1 are required for correct membrane trafficking. Using immunoprecipitation and mass spectrometry we reveal that PhIL1 interacts with known and new components of the gametocyte IMC.


Asunto(s)
Microtúbulos/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Desarrollo Sexual/fisiología , Animales , Microscopía Electrónica/métodos , Microtúbulos/ultraestructura , Plasmodium falciparum/ultraestructura , Transporte de Proteínas
2.
Cell Microbiol ; 17(2): 207-25, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25224798

RESUMEN

In preparation for transmission to its mosquito vector, Plasmodium falciparum, the most virulent of the human malaria parasites, adopts an unusual elongated shape. Here we describe a previously unrecognized actin-based cytoskeleton that is assembled in maturing P. falciparum gametocytes. Differential extraction reveals the presence of a highly stabilized population of F-actin at all stages of development. Super-resolution microscopy reveals an F-actin cytoskeleton that is concentrated at the ends of the elongating gametocyte but extends inward along the microtubule cytoskeleton. Formin-1 is also concentrated at the gametocyte ends suggesting a role in actin stabilization. Immunoelectron microscopy confirms that the actin cytoskeleton is located under the inner membrane complex rather than in the sub-alveolar space. In stage V gametocytes, the actin and microtubule cytoskeletons are reorganized in a coordinated fashion. The actin-depolymerizing agent, cytochalasin D, depletes actin from the end of the gametocytes, whereas the actin-stabilizing compound, jasplakinolide, induces formation of large bundles and prevents late-stage disassembly of the actin cytoskeleton. Long-term treatment with these compounds is associated with disruption of the normal mitochondrial organization and decreased gametocyte viability.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Plasmodium falciparum/química , Microscopía , Multimerización de Proteína
3.
FASEB J ; 25(4): 1218-29, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21169382

RESUMEN

Glucose is considered essential for erythrocytic stages of the malaria parasite, Plasmodium falciparum. Importance of sugar and its permease for hepatic and sexual stages of Plasmodium, however, remains elusive. Moreover, increasing global resistance to current antimalarials necessitates the search for novel drugs. Here, we reveal that hexose transporter 1 (HT1) of Plasmodium berghei can transport glucose (K(m)~87 µM), mannose (K(i)~93 µM), fructose (K(i)~0.54 mM), and galactose (K(i)~5 mM) in Leishmania mexicana mutant and Xenopus laevis; and, therefore, is functionally equivalent to HT1 of P. falciparum (Glc, K(m)~175 µM; Man, K(i)~276 µM; Fru, K(i)~1.25 mM; Gal, K(i)~5.86 mM). Notably, a glucose analog, C3361, attenuated hepatic (IC(50)~15 µM) and ookinete development of P. berghei. The PbHT1 could be ablated during intraerythrocytic stages only by concurrent complementation with PbHT1-HA or PfHT1. Together; these results signify that PbHT1 and glucose are required for the entire life cycle of P. berghei. Accordingly, PbHT1 is expressed in the plasma membrane during all parasite stages. To permit a high-throughput screening of PfHT1 inhibitors and their subsequent in vivo assessment, we have generated Saccharomyces cerevisiae mutant expressing codon-optimized PfHT1, and a PfHT1-dependent Δpbht1 parasite strain. This work provides a platform to facilitate the development of drugs against malaria, and it suggests a disease-control aspect by reducing parasite transmission.


Asunto(s)
Proteínas de Transporte de Monosacáridos/metabolismo , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Animales , Antimaláricos/farmacología , Secuencia de Bases , Fructosa/metabolismo , Galactosa/metabolismo , Glucosa/metabolismo , Humanos , Leishmania mexicana , Estadios del Ciclo de Vida , Manosa/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/metabolismo , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Toxoplasma/efectos de los fármacos , Xenopus laevis
4.
Cell Microbiol ; 13(12): 1996-2006, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21899701

RESUMEN

Functional analysis of Plasmodium genes by classical reverse genetics is currently limited to mutants that are viable during erythrocytic schizogony, the pathogenic phase of the malaria parasite where transfection is performed. Here, we describe a conceptually simple experimental approach to study the function of genes essential to the asexual blood stages in a subsequent life cycle stage by a promoter-swap approach. As a proof of concept we targeted the unconventional class XIV myosin MyoA, which is known to be required for Toxoplasma gondii tachyzoite locomotion and host cell invasion. By placing the corresponding Plasmodium berghei gene, PbMyoA, under the control of the apical membrane antigen 1 (AMA1) promoter, expression in blood stages is maintained but switched off during transmission to the insect vector, i.e. ookinetes. In those mutant ookinetes gliding motility is entirely abolished resulting in a complete block of life cycle progression in Anopheles mosquitoes. Similar approaches should permit the analysis of gene function in the mosquito forms that are shared with the erythrocytic stages of the malaria parasite.


Asunto(s)
Antígenos de Protozoos/metabolismo , Locomoción , Proteínas de la Membrana/metabolismo , Miosinas/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/patogenicidad , Animales , Anopheles/parasitología , Antígenos de Protozoos/genética , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Protozoarios , Prueba de Complementación Genética , Proteínas de la Membrana/genética , Ratones , Microinyecciones , Miosinas/genética , Oocistos/metabolismo , Plasmodium berghei/genética , Regiones Promotoras Genéticas , Proteínas Protozoarias/genética , Esporozoítos/metabolismo , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasmosis/parasitología , Transfección
5.
J Biol Chem ; 285(15): 11572-83, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20083609

RESUMEN

Cyclase-associated proteins (CAPs) are evolutionary conserved G-actin-binding proteins that regulate microfilament turnover. CAPs have a modular structure consisting of an N-terminal adenylate cyclase binding domain, a central proline-rich segment, and a C-terminal actin binding domain. Protozoan parasites of the phylum Apicomplexa, such as Cryptosporidium and the malaria parasite Plasmodium, express small CAP orthologs with homology to the C-terminal actin binding domain (C-CAP). Here, we demonstrate by reverse genetics that C-CAP is dispensable for the pathogenic Plasmodium blood stages. However, c-cap(-) parasites display a complete defect in oocyst development in the insect vector. By trans-species complementation we show that the Cryptosporidium parvum ortholog complements the Plasmodium gene functions. Purified recombinant C. parvum C-CAP protein binds actin monomers and prevents actin polymerization. The crystal structure of C. parvum C-CAP shows two monomers with a right-handed beta-helical fold intercalated at their C termini to form the putative physiological dimer. Our results reveal a specific vital role for an apicomplexan G-actin-binding protein during sporogony, the parasite replication phase that precedes formation of malaria transmission stages. This study also exemplifies how Plasmodium reverse genetics combined with biochemical and structural analyses of orthologous proteins can offer a fast track toward systematic gene characterization in apicomplexan parasites.


Asunto(s)
Actinas/química , Malaria/metabolismo , Malaria/transmisión , Proteínas de Microfilamentos/química , Oocistos/metabolismo , Secuencia de Aminoácidos , Animales , Cryptosporidium parvum/metabolismo , Culicidae , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Fenotipo , Plasmodium/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
6.
Mol Biol Cell ; 27(14): 2234-44, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226484

RESUMEN

Plasmodium relies on actin-based motility to migrate from the site of infection and invade target cells. Using a substrate-dependent gliding locomotion, sporozoites are able to move at fast speed (1-3 µm/s). This motility relies on a minimal set of actin regulatory proteins and occurs in the absence of detectable filamentous actin (F-actin). Here we report an overexpression strategy to investigate whether perturbations of F-actin steady-state levels affect gliding locomotion and host invasion. We selected two vital Plasmodium berghei G-actin-binding proteins, C-CAP and profilin, in combination with three stage-specific promoters and mapped the phenotypes afforded by overexpression in all three extracellular motile stages. We show that in merozoites and ookinetes, additional expression does not impair life cycle progression. In marked contrast, overexpression of C-CAP and profilin in sporozoites impairs circular gliding motility and salivary gland invasion. The propensity for productive motility correlates with actin accumulation at the parasite tip, as revealed by combinations of an actin-stabilizing drug and transgenic parasites. Strong expression of profilin, but not C-CAP, resulted in complete life cycle arrest. Comparative overexpression is an alternative experimental genetic strategy to study essential genes and reveals effects of regulatory imbalances that are not uncovered from deletion-mutant phenotyping.


Asunto(s)
Plasmodium/genética , Plasmodium/metabolismo , Profilinas/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Anopheles/parasitología , Movimiento Celular/genética , Movimiento Celular/fisiología , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo , Esporozoítos/fisiología
7.
PLoS One ; 8(8): e72771, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24013507

RESUMEN

Live cell imaging of recombinant malarial parasites encoding fluorescent probes provides critical insights into parasite-host interactions and life cycle progression. In this study, we generated a red fluorescent line of the murine malarial parasite Plasmodium berghei. To allow constitutive and abundant expression of the mCherry protein we profiled expression of all members of the P. berghei heat shock protein 70 (HSP70) family. We identified PbHSP70/1, an invariant ortholog of Plasmodium falciparum HSP70-1, as the protein with the highest expression levels during Plasmodium blood, mosquito, and liver infection. Stable allelic insertion of a mCherry expression cassette into the PbHsp70/1 locus created constitutive red fluorescent P. berghei lines, termed Pbred. We show that these parasites can be used for live imaging of infected host cells and organs, including hepatocytes, erythrocytes, and whole Anopheles mosquitoes. Quantification of the fluorescence intensity of several Pbred parasite stages revealed significantly enhanced signal intensities in comparison to GFP expressed under the control of the constitutive EF1alpha promoter. We propose that systematic transcript profiling permits generation of reporter parasites, such as the Pbred lines described herein.


Asunto(s)
Proteínas del Choque Térmico HSP72/metabolismo , Proteínas Luminiscentes/biosíntesis , Malaria , Organismos Modificados Genéticamente/metabolismo , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Anopheles/parasitología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Eritrocitos/patología , Femenino , Proteínas del Choque Térmico HSP72/genética , Hepatocitos/metabolismo , Hepatocitos/parasitología , Hepatocitos/patología , Proteínas Luminiscentes/genética , Malaria/genética , Malaria/metabolismo , Malaria/parasitología , Malaria/patología , Ratones , Organismos Modificados Genéticamente/genética , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Proteína Fluorescente Roja
8.
Eur J Cell Biol ; 90(11): 966-71, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21256619

RESUMEN

Many intracellular pathogens hijack host cell actin or its regulators for cell-to-cell spreading. In marked contrast, apicomplexan parasites, obligate intracellular, single cell eukaryotes that are phylogenetically older than the last common ancestor of animals and plants, employ their own actin cytoskeleton for active motility through tissues and invasion of host cells. A hallmark of actin-based motility of the malaria parasite is a minimal set of proteins that potentially regulate microfilament dynamics. An intriguing feature of the Plasmodium motor machinery is the virtual absence of elongated filamentous actin in vivo. Despite this unusual actin regulation sporozoites, the transmission stages that are injected into the mammalian host by Anopheles mosquitoes, display fast (1-3 µm/s) extracellular motility. Experimental genetics and analysis of recombinant proteins have recently contributed to clarify some of the cellular roles of apicomplexan actin monomer- and filament-binding proteins in parasite life cycle progression. These studies established that the malaria parasite employs multiple proteins that bind actin to form pools of readily polymerizable monomers, a prerequisite for fast formation of actin polymers. The motile extracellular stages of Plasmodium parasites are an excellent in vivo model system for functional characterization of actin regulation in single cell eukaryotes.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Malaria/parasitología , Plasmodium/metabolismo , Plasmodium/patogenicidad , Proteínas Protozoarias/metabolismo , Animales , Interacciones Huésped-Patógeno , Humanos , Malaria/metabolismo , Malaria/transmisión , Plasmodium/crecimiento & desarrollo , Unión Proteica
9.
Biochemistry ; 45(44): 13361-8, 2006 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17073457

RESUMEN

Clostridium botulinum produces a binary toxin, which is composed of two separate proteins. The enzyme component, C2I, is an ADP-ribosyltransferase which modifies G-actin of eukaryotic cells. The proteolytically activated binding/translocation component, C2IIa, forms ring-shaped heptamers, which bind to cell receptors and mediate the transport of C2I into the cytosol of target cells. According to the current model, receptor-bound C2IIa serves as a docking platform for C2I on the cell surface. Following assembly of C2I, the toxin complex is taken up via receptor-mediated endocytosis, and finally, C2IIa facilitates translocation of C2I from acidic endosomes into the cytosol. Our data support an alternative scenario for the early steps of interaction of the C2 toxin and eukaryotic cells, due to the fact that C2IIa and C2I can interact prior to binding of the toxin to the cell surface. The C2IIa-C2I complex, which was formed in a cell-free system, was detected by native gel electrophoresis and subsequent immunoblot analysis or radiolabeling methods. The preformed C2 toxin complex ADP-ribosylated actin in vitro and induced cell rounding. The interaction of C2I with C2IIa did not enhance the binding of C2IIa to the cellular receptor. Intoxication of Vero cells and of human colon carcinoma cells (CaCo-2) was significantly enhanced when the preformed toxin complex was added to cultured cells as compared to addition of the single components.


Asunto(s)
Toxinas Botulínicas/química , Clostridium botulinum/química , Actinas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Western Blotting , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/farmacología , Toxinas Botulínicas Tipo A , Membrana Celular/efectos de los fármacos , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Unión Proteica , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA