Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 319(2): C432-C440, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32608991

RESUMEN

microRNAs (miRNAs) are important regulators of cellular homeostasis and exert their effect by directly controlling protein expression. We have previously reported an age-dependent negative association between microRNA-99b (miR-99b-5p) expression and muscle protein synthesis in human muscle in vivo. Here we investigated the role of miR-99b-5p as a potential negative regulator of protein synthesis via inhibition of mammalian target for rapamycin (MTOR) signaling in human primary myocytes. Overexpressing miR-99b-5p in human primary myotubes from young and old subjects significantly decreased protein synthesis with no effect of donor age. A binding interaction between miR-99b-5p and its putative binding site within the MTOR 3'-untranslated region (UTR) was confirmed in C2C12 myoblasts. The observed decline in protein synthesis was, however, not associated with a suppression of the MTOR protein but of its regulatory associated protein of mTOR complex 1 (RPTOR). These results demonstrate that modulating the expression levels of a miRNA can regulate protein synthesis in human muscle cells and provide a potential mechanism for muscle wasting in vivo.


Asunto(s)
MicroARNs/genética , Fibras Musculares Esqueléticas/metabolismo , Biosíntesis de Proteínas/genética , Serina-Treonina Quinasas TOR/genética , Regiones no Traducidas 3'/genética , Animales , Proliferación Celular/genética , Regulación de la Expresión Génica/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Mioblastos/metabolismo , Transducción de Señal/genética
2.
mBio ; 10(3)2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164473

RESUMEN

Plasmodium parasites must export proteins into their erythrocytic host to survive. Exported proteins must cross the parasite plasma membrane (PPM) and the parasitophorous vacuolar membrane (PVM) encasing the parasite to access the host cell. Crossing the PVM requires protein unfolding and passage through a translocon, the Plasmodium translocon of exported proteins (PTEX). In this study, we provide the first direct evidence that heat shock protein 101 (HSP101), a core component of PTEX, unfolds proteins for translocation across the PVM by creating transgenic Plasmodium parasites in which the unfoldase and translocation functions of HSP101 have become uncoupled. Strikingly, while these parasites could export native proteins, they were unable to translocate soluble, tightly folded reporter proteins bearing the Plasmodium export element (PEXEL) across the PVM into host erythrocytes under the same conditions. In contrast, an identical PEXEL reporter protein but harboring a transmembrane domain could be exported, suggesting that a prior unfolding step occurs at the PPM. Together, these results demonstrate that the export of parasite proteins is dependent on how these proteins are presented to the secretory pathway before they reach PTEX as well as their folded status. Accordingly, only tightly folded soluble proteins secreted into the vacuolar space and not proteins containing transmembrane domains or the majority of erythrocyte-stage exported proteins have an absolute requirement for the full unfoldase activity of HSP101 to be exported.IMPORTANCE The Plasmodium parasites that cause malaria export hundreds of proteins into their host red blood cell (RBC). These exported proteins drastically alter the structural and functional properties of the RBC and play critical roles in parasite virulence and survival. To access the RBC cytoplasm, parasite proteins must pass through the Plasmodium translocon of exported proteins (PTEX) located at the membrane interfacing the parasite and host cell. Our data provide evidence that HSP101, a component of PTEX, serves to unfold protein cargo requiring translocation. We also reveal that addition of a transmembrane domain to soluble cargo influences its ability to be translocated by parasites in which the HSP101 motor and unfolding activities have become uncoupled. Therefore, we propose that proteins with transmembrane domains use an alternative unfolding pathway prior to PTEX to facilitate export.


Asunto(s)
Eritrocitos/parasitología , Proteínas de Choque Térmico/metabolismo , Interacciones Huésped-Parásitos , Plasmodium berghei/genética , Desplegamiento Proteico , Proteínas Protozoarias/metabolismo , Animales , Femenino , Proteínas de Choque Térmico/genética , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/metabolismo , Transporte de Proteínas , Proteínas Protozoarias/genética , Solubilidad
3.
BMC Mol Cell Biol ; 20(1): 12, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31138100

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) are increasingly being identified as modulatory molecules for physiological and pathological processes in muscle. Here, we investigated whether miRNAs influenced the expression of the stress-responsive gene N-myc downstream-regulated gene 2 (Ndrg2) in skeletal muscle cells through the targeted degradation or translation inhibition of NDRG2 mRNA transcripts during basal or catabolic stress conditions. RESULTS: Three miRNAs, mmu-miR-23a-3p (miR-23a), mmu-miR-23b-3p (miR-23b) and mmu-miR-28-5p (miR-28), were identified using an in silico approach and confirmed to target the 3' untranslated region of the mouse Ndrg2 gene through luciferase reporter assays. However, miR-23a, -23b or -28 overexpression had no influence on NDRG2 mRNA or protein levels up to 48 h post treatment in mouse C2C12 myotubes under basal conditions. Interestingly, a compensatory decrease in the endogenous levels of the miRNAs in response to each other's overexpression was measured. Furthermore, dexamethasone, a catabolic stress agent that induces NDRG2 expression, decreased miR-23a and miR-23b endogenous levels at 24 h post treatment suggesting an interplay between these miRNAs and NDRG2 regulation under similar stress conditions. Accordingly, when overexpressed simultaneously, miR-23a, -23b and -28 attenuated the dexamethasone-induced increase of NDRG2 protein translation but did not affect Ndrg2 gene expression. CONCLUSION: These findings highlight modulatory and co-regulatory roles for miR-23a, -23b and -28 and their novel regulation of NDRG2 during stress conditions in muscle.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , MicroARNs/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Estrés Fisiológico/genética , Regiones no Traducidas 3'/genética , Animales , Sitios de Unión , Línea Celular , Simulación por Computador , Dexametasona/farmacología , Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Biosíntesis de Proteínas , Estrés Fisiológico/efectos de los fármacos , Transfección
4.
FEBS J ; 285(11): 2037-2055, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29637707

RESUMEN

The pathogenic nature of malaria infections is due in part to the export of hundreds of effector proteins that actively remodel the host erythrocyte. The Plasmodium translocon of exported proteins (PTEX) has been shown to facilitate the trafficking of proteins into the host cell, a process that is essential for the survival of the parasite. The role of the auxiliary PTEX component PTEX88 remains unclear, as previous attempts to elucidate its function through reverse genetic approaches showed that in contrast to the core components PTEX150 and HSP101, knockdown of PTEX88 did not give rise to an export phenotype. Here, we have used biochemical approaches to understand how PTEX88 assembles within the translocation machinery. Proteomic analysis of the PTEX88 interactome showed that PTEX88 interacts closely with HSP101 but has a weaker affinity with the other core constituents of PTEX. PTEX88 was also found to associate with other PV-resident proteins, including chaperones and members of the exported protein-interacting complex that interacts with the major virulence factor PfEMP1, the latter contributing to cytoadherence and parasite virulence. Despite being expressed for the duration of the blood-stage life cycle, PTEX88 was only discretely observed at the parasitophorous vacuole membrane during ring stages and could not always be detected in the major high molecular weight complex that contains the other core components of PTEX, suggesting that its interaction with the PTEX complex may be dynamic. Together, these data have enabled the generation of an updated model of PTEX that now includes how PTEX88 assembles within the complex.


Asunto(s)
Interacciones Huésped-Parásitos/genética , Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Animales , Eritrocitos/parasitología , Humanos , Estadios del Ciclo de Vida/genética , Malaria Falciparum/parasitología , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Plasmodium falciparum/patogenicidad , Transporte de Proteínas/genética , Proteómica
5.
Am J Physiol Endocrinol Metab ; 313(3): E335-E343, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28465283

RESUMEN

Impairments in mitochondrial function and substrate metabolism are implicated in the etiology of obesity and Type 2 diabetes. MicroRNAs (miRNAs) can degrade mRNA or repress protein translation and have been implicated in the development of such disorders. We used a contrasting rat model system of selectively bred high- (HCR) or low- (LCR) intrinsic running capacity with established differences in metabolic health to investigate the molecular mechanisms through which miRNAs regulate target proteins mediating mitochondrial function and substrate oxidation processes. Quantification of select miRNAs using the rat miFinder miRNA PCR array revealed differential expression of 15 skeletal muscles (musculus tibialis anterior) miRNAs between HCR and LCR rats (14 with higher expression in LCR; P < 0.05). Ingenuity Pathway Analysis predicted these altered miRNAs to collectively target multiple proteins implicated in mitochondrial dysfunction and energy substrate metabolism. Total protein abundance of citrate synthase (CS; miR-19 target) and voltage-dependent anion channel 1 (miR-7a target) were higher in HCR compared with LCR cohorts (~57 and ~26%, respectively; P < 0.05). A negative correlation was observed for miR-19a-3p and CS (r = 0.32, P = 0.015) protein expression. To determine whether miR-19a-3p can regulate CS in vitro, we performed luciferase reporter and transfection assays in C2C12 myotubes. MiR-19a-3p binding to the CS untranslated region did not change luciferase reporter activity; however, miR-19a-3p transfection decreased CS protein expression (∼70%; P < 0.05). The differential miRNA expression targeting proteins implicated in mitochondrial dysfunction and energy substrate metabolism may contribute to the molecular basis, mediating the divergent metabolic health profiles of LCR and HCR rats.


Asunto(s)
Tolerancia al Ejercicio/genética , MicroARNs/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Carrera , Animales , Western Blotting , Línea Celular , Citrato (si)-Sintasa/metabolismo , Metabolismo Energético/genética , Técnicas In Vitro , Ratones , Fibras Musculares Esqueléticas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
6.
Genetics ; 205(1): 185-199, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28049706

RESUMEN

Methylation of histone H3 lysine 4 (H3K4) by Set1 complex/COMPASS is a hallmark of eukaryotic chromatin, but it remains poorly understood how this post-translational modification contributes to the regulation of biological processes like the cell cycle. Here, we report a H3K4 methylation-dependent pathway in Saccharomyces cerevisiae that governs toxicity toward benomyl, a microtubule destabilizing drug. Benomyl-sensitive growth of wild-type cells required mono- and dimethylation of H3K4 and Pho23, a PHD-containing subunit of the Rpd3L complex. Δset1 and Δpho23 deletions suppressed defects associated with ipl1-2 aurora kinase mutant, an integral component of the spindle assembly checkpoint during mitosis. Benomyl resistance of Δset1 strains was accompanied by deregulation of all four tubulin genes and the phenotype was suppressed by tub2-423 and Δtub3 mutations, establishing a genetic link between H3K4 methylation and microtubule function. Most interestingly, sine wave fitting and clustering of transcript abundance time series in synchronized cells revealed a requirement for Set1 for proper cell-cycle-dependent gene expression and Δset1 cells displayed delayed entry into S phase. Disruption of G1/S regulation in Δmbp1 and Δswi4 transcription factor mutants duplicated both benomyl resistance and suppression of ipl1-2 as was observed with Δset1 Taken together our results support a role for H3K4 methylation in the coordination of cell-cycle progression and proper assembly of the mitotic spindle during mitosis.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Huso Acromático/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Lisina/metabolismo , Metilación , Mitosis/fisiología , Procesamiento Proteico-Postraduccional , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Ubiquitinación
7.
PLoS One ; 11(6): e0158174, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27348424

RESUMEN

Over-expression of a GFP-PfRab1A fusion protein in Plasmodium falciparum schizonts produces a punctate pattern of fluorescence typical of rhoptries, secretory organelles involved in host cell invasion. The GFP-positive bodies were purified by a combination of differential and density gradient centrifugation and their protein content determined by MS/MS sequencing. Consistent with the GFP rhoptry-like pattern of transgenic parasites, four of the 19 proteins identified have been previously described to be rhoptry-associated and another four are ER or ER-associated proteins. Confirmation that GFP-PfRab1A decorates rhoptries was obtained by its co-localization with Rap1 and Ron4 in late phase schizonts. We conclude that PfRab1A potentially regulates vesicular traffic from the endoplasmic reticulum to the rhoptries in Apicomplexa parasites.


Asunto(s)
Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Esquizontes/metabolismo , Proteínas de Unión al GTP rab1/metabolismo , Clonación Molecular , Plasmodium falciparum/genética , Transporte de Proteínas , Proteómica/métodos , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión al GTP rab1/genética
8.
Cell Microbiol ; 18(11): 1551-1569, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27019089

RESUMEN

The Plasmodium translocon for exported proteins (PTEX) has been established as the machinery responsible for the translocation of all classes of exported proteins beyond the parasitophorous vacuolar membrane of the intraerythrocytic malaria parasite. Protein export, particularly in the asexual blood stage, is crucial for parasite survival as exported proteins are involved in remodelling the host cell, an essential process for nutrient uptake, waste removal and immune evasion. Here, we have truncated the conserved C-terminus of one of the essential PTEX components, PTEX150, in Plasmodium falciparum in an attempt to create mutants of reduced functionality. Parasites tolerated C-terminal truncations of up to 125 amino acids with no reduction in growth, protein export or the establishment of new permeability pathways. Quantitative proteomic approaches however revealed a decrease in other PTEX subunits associating with PTEX150 in truncation mutants, suggesting a role for the C-terminus of PTEX150 in regulating PTEX stability. Our analyses also reveal three previously unreported PTEX-associated proteins, namely PV1, Pf113 and Hsp70-x (respective PlasmoDB numbers; PF3D7_1129100, PF3D7_1420700 and PF3D7_0831700) and demonstrate that core PTEX proteins exist in various distinct multimeric forms outside the major complex.


Asunto(s)
Eritrocitos/parasitología , Proteínas de Transporte de Membrana/fisiología , Plasmodium falciparum/fisiología , Proteoma/metabolismo , Proteínas Protozoarias/fisiología , Células Cultivadas , Humanos , Complejos Multiproteicos/metabolismo , Dominios Proteicos , Mapas de Interacción de Proteínas , Estabilidad Proteica , Transporte de Proteínas
9.
PLoS One ; 11(2): e0149296, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26886275

RESUMEN

Pathogenesis of malaria infections is linked to remodeling of erythrocytes, a process dependent on the trafficking of hundreds of parasite-derived proteins into the host erythrocyte. Recent studies have demonstrated that the Plasmodium translocon of exported proteins (PTEX) serves as the central gateway for trafficking of these proteins, as inducible knockdown of the core PTEX constituents blocked the trafficking of all classes of cargo into the erythrocyte. However, the role of the auxiliary component PTEX88 in protein export remains less clear. Here we have used inducible knockdown technologies in P. falciparum and P. berghei to assess the role of PTEX88 in parasite development and protein export, which reveal that the in vivo growth of PTEX88-deficient parasites is hindered. Interestingly, we were unable to link this observation to a general defect in export of a variety of known parasite proteins, suggesting that PTEX88 functions in a different fashion to the core PTEX components. Strikingly, PTEX88-deficient P. berghei were incapable of causing cerebral malaria despite a robust pro-inflammatory response from the host. These parasites also exhibited a reduced ability to sequester in peripheral tissues and were removed more readily from the circulation by the spleen. In keeping with these findings, PTEX88-deficient P. falciparum-infected erythrocytes displayed reduced binding to the endothelial cell receptor, CD36. This suggests that PTEX88 likely plays a specific direct or indirect role in mediating parasite sequestration rather than making a universal contribution to the trafficking of all exported proteins.


Asunto(s)
Técnicas de Silenciamiento del Gen , Parásitos/patogenicidad , Plasmodium berghei/patogenicidad , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Animales , Antígenos CD36/metabolismo , Adhesión Celular , Femenino , Glucosamina/metabolismo , Inmunidad , Inflamación/inmunología , Inflamación/patología , Ratones Endogámicos C57BL , Parásitos/crecimiento & desarrollo , Unión Proteica , Transporte de Proteínas , Virulencia
10.
Cell Microbiol ; 18(3): 399-412, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26347246

RESUMEN

Export of most malaria proteins into the erythrocyte cytosol requires the Plasmodium translocon of exported proteins (PTEX) and a cleavable Plasmodium export element (PEXEL). In contrast, the contribution of PTEX in the liver stages and export of liver stage proteins is unknown. Here, using the FLP/FRT conditional mutatagenesis system, we generate transgenic Plasmodium berghei parasites deficient in EXP2, the putative pore-forming component of PTEX. Our data reveal that EXP2 is important for parasite growth in the liver and critical for parasite transition to the blood, with parasites impaired in their ability to generate a patent blood-stage infection. Surprisingly, whilst parasites expressing a functional PTEX machinery can efficiently export a PEXEL-bearing GFP reporter into the erythrocyte cytosol during a blood stage infection, this same reporter aggregates in large accumulations within the confines of the parasitophorous vacuole membrane during hepatocyte growth. Notably HSP101, the putative molecular motor of PTEX, could not be detected during the early liver stages of infection, which may explain why direct protein translocation of this soluble PEXEL-bearing reporter or indeed native PEXEL proteins into the hepatocyte cytosol has not been observed. This suggests that PTEX function may not be conserved between the blood and liver stages of malaria infection.


Asunto(s)
Malaria/parasitología , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/metabolismo , Animales , Animales Modificados Genéticamente , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Choque Térmico/metabolismo , Interacciones Huésped-Parásitos , Hígado/parasitología , Ratones , Plasmodium berghei/genética , Transporte de Proteínas/genética , Proteínas Protozoarias/genética , Tetraciclinas/farmacología
11.
Nature ; 511(7511): 587-91, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25043043

RESUMEN

During the blood stages of malaria, several hundred parasite-encoded proteins are exported beyond the double-membrane barrier that separates the parasite from the host cell cytosol. These proteins have a variety of roles that are essential to virulence or parasite growth. There is keen interest in understanding how proteins are exported and whether common machineries are involved in trafficking the different classes of exported proteins. One potential trafficking machine is a protein complex known as the Plasmodium translocon of exported proteins (PTEX). Although PTEX has been linked to the export of one class of exported proteins, there has been no direct evidence for its role and scope in protein translocation. Here we show, through the generation of two parasite lines defective for essential PTEX components (HSP101 or PTEX150), and analysis of a line lacking the non-essential component TRX2 (ref. 12), greatly reduced trafficking of all classes of exported proteins beyond the double membrane barrier enveloping the parasite. This includes proteins containing the PEXEL motif (RxLxE/Q/D) and PEXEL-negative exported proteins (PNEPs). Moreover, the export of proteins destined for expression on the infected erythrocyte surface, including the major virulence factor PfEMP1 in Plasmodium falciparum, was significantly reduced in PTEX knockdown parasites. PTEX function was also essential for blood-stage growth, because even a modest knockdown of PTEX components had a strong effect on the parasite's capacity to complete the erythrocytic cycle both in vitro and in vivo. Hence, as the only known nexus for protein export in Plasmodium parasites, and an essential enzymic machine, PTEX is a prime drug target.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Malaria/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Proteínas de Choque Térmico/genética , Humanos , Estadios del Ciclo de Vida/fisiología , Complejos Multiproteicos/metabolismo , Transporte de Proteínas/genética , Proteínas Protozoarias/genética , Vacuolas/metabolismo , Vacuolas/parasitología
12.
Mol Microbiol ; 89(6): 1167-86, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23869529

RESUMEN

Plasmodium parasites remodel their vertebrate host cells by translocating hundreds of proteins across an encasing membrane into the host cell cytosol via a putative export machinery termed PTEX. Previously PTEX150, HSP101 and EXP2 have been shown to be bona fide members of PTEX. Here we validate that PTEX88 and TRX2 are also genuine members of PTEX and provide evidence that expression of PTEX components are also expressed in early gametocytes, mosquito and liver stages, consistent with observations that protein export is not restricted to asexual stages. Although amenable to genetic tagging, HSP101, PTEX150, EXP2 and PTEX88 could not be genetically deleted in Plasmodium berghei, in keeping with the obligatory role this complex is postulated to have in maintaining normal blood-stage growth. In contrast, the putative thioredoxin-like protein TRX2 could be deleted, with knockout parasites displaying reduced grow-rates, both in vivo and in vitro, and reduced capacity to cause severe disease in a cerebral malaria model. Thus, while not essential for parasite survival, TRX2 may help to optimize PTEX activity. Importantly, the generation of TRX2 knockout parasites that display altered phenotypes provides a much-needed tool to dissect PTEX function.


Asunto(s)
Parasitemia/parasitología , Plasmodium berghei/enzimología , Plasmodium berghei/patogenicidad , Tiorredoxinas/metabolismo , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Análisis de Supervivencia , Tiorredoxinas/genética , Virulencia , Factores de Virulencia/genética
13.
PLoS One ; 8(4): e61482, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658610

RESUMEN

Protein export into the host red blood cell is one of the key processes in the pathobiology of the malaria parasite Plasmodiumtrl falciparum, which extensively remodels the red blood cell to ensure its virulence and survival. In this study, we aimed to shed further light on the protein export mechanisms in the rodent malaria parasite P. berghei and provide further proof of the conserved nature of host cell remodeling in Plasmodium spp. Based on the presence of an export motif (R/KxLxE/Q/D) termed PEXEL (Plasmodium export element), we have generated transgenic P. berghei parasite lines expressing GFP chimera of putatively exported proteins and analysed one of the newly identified exported proteins in detail. This essential protein, termed PbCP1 (P. berghei Cleft-like Protein 1), harbours an atypical PEXEL motif (RxLxY) and is further characterised by two predicted transmembrane domains (2TMD) in the C-terminal end of the protein. We have functionally validated the unusual PEXEL motif in PbCP1 and analysed the role of the 2TMD region, which is required to recruit PbCP1 to discrete membranous structures in the red blood cell cytosol that have a convoluted, vesico-tubular morphology by electron microscopy. Importantly, this study reveals that rodent malaria species also induce modifications to their host red blood cell.


Asunto(s)
Citosol/parasitología , Eritrocitos/parasitología , Plasmodium berghei/metabolismo , Proteínas Protozoarias/química , Secuencia de Aminoácidos , Animales , Citosol/ultraestructura , Eritrocitos/ultraestructura , Expresión Génica , Proteínas Fluorescentes Verdes , Estadios del Ciclo de Vida/genética , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Plasmodium berghei/genética , Plasmodium berghei/ultraestructura , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
14.
Proc Natl Acad Sci U S A ; 110(18): 7506-11, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589867

RESUMEN

The human malaria parasite Plasmodium falciparum harbors a relict, nonphotosynthetic plastid of algal origin termed the apicoplast. Although considerable progress has been made in defining the metabolic functions of the apicoplast, information on the composition and biogenesis of the four delimiting membranes of this organelle is limited. Here, we report an efficient method for preparing highly purified apicoplasts from red blood cell parasite stages and the comprehensive lipidomic analysis of this organelle. Apicoplasts were prepared from transgenic parasites expressing an epitope-tagged triosephosphate transporter and immunopurified on magnetic beads. Gas and liquid chromatography MS analyses of isolated apicoplast lipids indicated significant differences compared with total parasite lipids. In particular, apicoplasts were highly enriched in phosphatidylinositol, consistent with a suggested role for phosphoinositides in targeting membrane vesicles to apicoplasts. Apicoplast phosphatidylinositol and other phospholipids were also enriched in saturated fatty acids, which could reflect limited acyl exchange with other membrane phospholipids and/or a requirement for specific physical properties. Lipids atypical for plastids (sphingomyelins, ceramides, and cholesterol) were detected in apicoplasts. The presence of cholesterol in apicoplast membranes was supported by filipin staining of isolated apicoplasts. Galactoglycerolipids, dominant in plant and algal plastids, were not detected in P. falciparum apicoplasts, suggesting that these glycolipids are a hallmark of photosynthetic plastids and were lost when these organisms assumed a parasitic lifestyle. Apicoplasts thus contain an atypical melange of lipids scavenged from the human host alongside lipids remodeled by the parasite cytoplasm, and stable isotope labeling shows some apicoplast lipids are generated de novo by the organelle itself.


Asunto(s)
Lípidos/química , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Plastidios/química , Colesterol/metabolismo , Cromatografía Liquida , Ácidos Grasos/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Humanos , Metabolismo de los Lípidos , Plasmodium falciparum/ultraestructura , Plastidios/ultraestructura
15.
Trends Parasitol ; 29(5): 228-36, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23570755

RESUMEN

Apicomplexan parasites, including the Plasmodium species that cause malaria, contain three unusual apical secretory organelles (micronemes, rhoptries, and dense granules) that are required for the infection of new host cells. Because of their specialized nature, the majority of proteins secreted from these organelles are unique to Apicomplexans and are consequently poorly characterized. Although rhoptry proteins of Plasmodium have been implicated in events central to invasion, there is growing evidence to suggest that proteins originating from this organelle play key roles downstream of parasite entry into the host cell. Here we discuss recent work that has advanced our knowledge of rhoptry protein trafficking and function, and highlight areas of research that require further investigation.


Asunto(s)
Apicomplexa/patogenicidad , Orgánulos/metabolismo , Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Apicomplexa/metabolismo , Interacciones Huésped-Parásitos , Plasmodium/fisiología , Transporte de Proteínas
16.
J Biol Chem ; 287(11): 7871-84, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22253438

RESUMEN

To survive within its host erythrocyte, Plasmodium falciparum must export hundreds of proteins across both its parasite plasma membrane and surrounding parasitophorous vacuole membrane, most of which are likely to use a protein complex known as PTEX (Plasmodium translocon of exported proteins). PTEX is a putative protein trafficking machinery responsible for the export of hundreds of proteins across the parasitophorous vacuole membrane and into the human host cell. Five proteins are known to comprise the PTEX complex, and in this study, three of the major stoichiometric components are investigated including HSP101 (a AAA(+) ATPase), a protein of no known function termed PTEX150, and the apparent membrane component EXP2. We show that these proteins are synthesized in the preceding schizont stage (PTEX150 and HSP101) or even earlier in the life cycle (EXP2), and before invasion these components reside within the dense granules of invasive merozoites. From these apical organelles, the protein complex is released into the host cell where it resides with little turnover in the parasitophorous vacuole membrane for most of the remainder of the following cell cycle. At this membrane, PTEX is arranged in a stable macromolecular complex of >1230 kDa that includes an ∼600-kDa apparently homo-oligomeric complex of EXP2 that can be separated from the remainder of the PTEX complex using non-ionic detergents. Two different biochemical methods undertaken here suggest that PTEX components associate as EXP2-PTEX150-HSP101, with EXP2 associating with the vacuolar membrane. Collectively, these data support the hypothesis that EXP2 oligomerizes and potentially forms the putative membrane-spanning pore to which the remainder of the PTEX complex is attached.


Asunto(s)
Membranas Intracelulares/metabolismo , Proteínas de la Membrana/biosíntesis , Complejos Multiproteicos/biosíntesis , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/biosíntesis , Vacuolas/metabolismo , Humanos , Proteínas de la Membrana/genética , Complejos Multiproteicos/genética , Plasmodium falciparum/genética , Transporte de Proteínas/fisiología , Proteínas Protozoarias/genética , Esquizontes/metabolismo , Vacuolas/genética
17.
Biochem Soc Trans ; 38(3): 775-82, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20491664

RESUMEN

Malaria, which is caused by species of the parasite genus Plasmodium, remains a major global health problem. A vestigial plastid homologous with the chloroplasts of plants and algae was discovered in malaria and related parasites from the phylum Apicomplexa and has radically changed our view of the evolutionary origins of these disease-causing protists. We now recognize that this large group of parasites had a photosynthetic ancestry and were converted into parasitism early in the evolution of animals. Apicomplexans have probably been parasitizing the animal kingdom for more than 500 million years. The relic plastid persists in most apicomplexans and is an essential component. Perturbation of apicoplast function or inheritance results in parasite death, making the organelle a promising target for chemotherapy. Plastids, including those of malaria parasites, are essentially reduced endosymbiotic bacteria living inside a eukaryotic host. This means that plastids have bacterial-type metabolic pathways and housekeeping processes, all of which are vulnerable to antibacterial compounds. Indeed, many antibacterials kill malaria parasites by blocking essential processes in the plastid. Furthermore, a range of herbicides that target plastid metabolism of undesired plants are also parasiticidal, making them potential new leads for antimalarial drugs. In the present review, we examine the evolutionary origins of the malaria parasite's plastid by endosymbiosis and outline the recent findings on how the organelle imports nuclear-encoded proteins through a set of translocation machineries in the membranes that bound the organelle.


Asunto(s)
Malaria Falciparum/parasitología , Plasmodium falciparum/citología , Plastidios , Simbiosis , Animales , Evolución Biológica , Humanos , Filogenia , Plasmodium falciparum/clasificación , Plasmodium falciparum/fisiología , Plastidios/metabolismo , Plastidios/ultraestructura
18.
Eukaryot Cell ; 8(8): 1146-54, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19502580

RESUMEN

Protein trafficking to the stroma of the apicoplast of Plasmodium falciparum requires translocation across several membranes. To further elucidate the mechanisms responsible, we investigated two proteins: P. falciparum Tic22 (PfTic22), a putative component of the translocon of the inner chloroplast membrane; and PfsDer1-1, one of two homologues of the P. falciparum symbiont-derived Der1 (sDer1) protein, a putative component of an endoplasmic reticulum-associated degradation (ERAD) complex in the periplastid membrane. We constructed parasites expressing hemagglutinin (HA)-tagged PfTic22 and PfsDer1-1 under the control of their endogenous promoters using the 3' replacement strategy. We show that both PfTic22-HA and PfsDer1-1-HA are expressed predominantly during the trophozoite stage of the asexual replication cycle, which corresponds to the most dynamic stages of apicoplast activity. Although both proteins localize to the periphery of the apicoplast, PfTic22-HA is a membrane-associated protein while PfsDer1-1-HA is an integral membrane protein. Phylogenetic analysis indicates that PfsDer1-1 is one of two Der1 paralogues predicted to localize to the apicoplast in P. falciparum and that it has orthologues in diatom algae, supporting the chromalveolate hypothesis. These observations are consistent with putative roles for PfTic22 and PfsDer1-1 in protein translocation into the apicoplast of P. falciparum.


Asunto(s)
Plasmodium falciparum/metabolismo , Plastidios/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Plasmodium falciparum/genética , Plastidios/genética , Transporte de Proteínas , Proteínas Protozoarias/genética
19.
Trends Parasitol ; 25(5): 197-200, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19346163

RESUMEN

Several apicomplexan parasites harbour an essential plastid known as the apicoplast. Apicoplasts import proteins and metabolites for several biological functions, but how import is achieved is largely unknown. Two recent reports have identified novel proteins in the apicoplast membranes, providing new perspectives on how proteins traffic to this organelle. The first report contributes to a newly recognized apicoplast-targeting pathway for membrane proteins, and the second identifies the first member of the protein-translocation complex in apicoplasts.


Asunto(s)
Membranas Intracelulares/metabolismo , Plasmodium falciparum/metabolismo , Plastidios/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Animales , Proteínas de la Membrana/metabolismo , Plasmodium falciparum/ultraestructura , Plastidios/ultraestructura , Transporte de Proteínas , Toxoplasma/ultraestructura
20.
Genetics ; 179(1): 95-112, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18493043

RESUMEN

The recently completed genome of Chlamydomonas reinhardtii was surveyed for components of the chloroplast protein translocation complexes. Putative components were identified using reciprocal BlastP searches with the protein sequences of Arabidopsis thaliana as queries. As a comparison, we also surveyed the new genomes of the bryophyte Physcomitrella patens, two prasinophyte green algae (Ostreococcus lucimarinus and Ostreococcus tauri), the red alga Cyanidioschizon merolae, and several cyanobacteria. Overall, we found that the components of the import pathway are remarkably well conserved, particularly among the Viridiplantae lineages. Specifically, C. reinhardtii contained almost all the components found in A. thaliana, with two exceptions. Missing from C. reinhardtii are the C-terminal ferredoxin-NADPH-reductase (FNR) binding domain of Tic62 and a full-length, TPR-bearing Toc64. Further, the N-terminal domain of C. reinhardtii Toc34 is highly acidic, whereas the analogous region in C. reinhardtii Toc159 is not. This reversal of the vascular plant model may explain the similarity of C. reinhardtii chloroplast transit peptides to mitochondrial-targeting peptides. Other findings from our genome survey include the absence of Tic22 in both Ostreococcus genomes; the presence of only one Toc75 homolog in C. merolae; and, finally, a distinctive propensity for gene duplication in P. patens.


Asunto(s)
Chlamydomonas reinhardtii/genética , Cloroplastos/genética , Evolución Molecular , Proteínas de Transporte de Membrana/genética , Modelos Moleculares , Complejos Multiproteicos/genética , Filogenia , Secuencia de Aminoácidos , Animales , Biología Computacional , Genómica , Funciones de Verosimilitud , Modelos Genéticos , Transporte de Proteínas/genética , Análisis de Secuencia , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...