Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 16(12): e1009067, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33383579

RESUMEN

Inorganic ions such as phosphate, are essential nutrients required for a broad spectrum of cellular functions and regulation. During infection, pathogens must obtain inorganic phosphate (Pi) from the host. Despite the essentiality of phosphate for all forms of life, how the intracellular parasite Toxoplasma gondii acquires Pi from the host cell is still unknown. In this study, we demonstrated that Toxoplasma actively internalizes exogenous Pi by exploiting a gradient of Na+ ions to drive Pi uptake across the plasma membrane. The Na+-dependent phosphate transport mechanism is electrogenic and functionally coupled to a cipargarmin sensitive Na+-H+-ATPase. Toxoplasma expresses one transmembrane Pi transporter harboring PHO4 binding domains that typify the PiT Family. This transporter named TgPiT, localizes to the plasma membrane, the inward buds of the endosomal organelles termed VAC, and many cytoplasmic vesicles. Upon Pi limitation in the medium, TgPiT is more abundant at the plasma membrane. We genetically ablated the PiT gene, and ΔTgPiT parasites are impaired in importing Pi and synthesizing polyphosphates. Interestingly, ΔTgPiT parasites accumulate 4-times more acidocalcisomes, storage organelles for phosphate molecules, as compared to parental parasites. In addition, these mutants have a reduced cell volume, enlarged VAC organelles, defects in calcium storage and a slightly alkaline pH. Overall, these mutants exhibit severe growth defects and have reduced acute virulence in mice. In survival mode, ΔTgPiT parasites upregulate several genes, including those encoding enzymes that cleave or transfer phosphate groups from phosphometabolites, transporters and ions exchangers localized to VAC or acidocalcisomes. Taken together, these findings point to a critical role of TgPiT for Pi supply for Toxoplasma and also for protection against osmotic stresses.


Asunto(s)
Osmorregulación/genética , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/fisiología , Toxoplasma , Animales , Animales Modificados Genéticamente , Transporte Biológico/genética , Células Cultivadas , Humanos , Ratones , Proteínas Cotransportadoras de Sodio-Fosfato/genética , Toxoplasma/genética , Toxoplasma/metabolismo
2.
J Cell Biol ; 216(12): 4235-4254, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29070609

RESUMEN

Many intracellular pathogens subvert host membrane trafficking pathways to promote their replication. Toxoplasma multiplies in a membrane-bound parasitophorous vacuole (PV) that interacts with mammalian host organelles and intercepts Golgi Rab vesicles to acquire sphingolipids. The mechanisms of host vesicle internalization and processing within the PV remain undefined. We demonstrate that Toxoplasma sequesters a broad range of Rab vesicles into the PV. Correlative light and electron microscopy analysis of infected cells illustrates that intravacuolar Rab1A vesicles are surrounded by the PV membrane, suggesting a phagocytic-like process for vesicle engulfment. Rab11A vesicles concentrate to an intravacuolar network (IVN), but this is reduced in Δgra2 and Δgra2Δgra6 parasites, suggesting that tubules stabilized by the TgGRA2 and TgGRA6 proteins secreted by the parasite within the PV contribute to host vesicle sequestration. Overexpression of a phospholipase TgLCAT, which is localized to the IVN, results in a decrease in the number of intravacuolar GFP-Rab11A vesicles, suggesting that TgLCAT controls lipolytic degradation of Rab vesicles for cargo release.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Interacciones Huésped-Parásitos , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Toxoplasma/metabolismo , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Células CHO , Chlorocebus aethiops , Cricetulus , Vesículas Citoplasmáticas/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/parasitología , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Genes Reporteros , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Fagocitosis , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esfingolípidos/metabolismo , Toxoplasma/ultraestructura , Vacuolas/ultraestructura , Células Vero , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab1/genética , Proteínas de Unión al GTP rab1/metabolismo
3.
mBio ; 7(3)2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27353755

RESUMEN

UNLABELLED: Plasmodium parasites undergo continuous cellular renovation to adapt to various environments in the vertebrate host and insect vector. In hepatocytes, Plasmodium berghei discards unneeded organelles for replication, such as micronemes involved in invasion. Concomitantly, intrahepatic parasites expand organelles such as the apicoplast that produce essential metabolites. We previously showed that the ATG8 conjugation system is upregulated in P. berghei liver forms and that P. berghei ATG8 (PbATG8) localizes to the membranes of the apicoplast and cytoplasmic vesicles. Here, we focus on the contribution of PbATG8 to the organellar changes that occur in intrahepatic parasites. We illustrated that micronemes colocalize with PbATG8-containing structures before expulsion from the parasite. Interference with PbATG8 function by overexpression results in poor development into late liver stages and production of small merosomes that contain immature merozoites unable to initiate a blood infection. At the cellular level, PbATG8-overexpressing P. berghei exhibits a delay in microneme compartmentalization into PbATG8-containing autophagosomes and elimination compared to parasites from the parental strain. The apicoplast, identifiable by immunostaining of the acyl carrier protein (ACP), undergoes an abnormally fast proliferation in mutant parasites. Over time, the ACP staining becomes diffuse in merosomes, indicating a collapse of the apicoplast. PbATG8 is not incorporated into the progeny of mutant parasites, in contrast to parental merozoites in which PbATG8 and ACP localize to the apicoplast. These observations reveal that Plasmodium ATG8 is a key effector in the development of merozoites by controlling microneme clearance and apicoplast proliferation and that dysregulation in ATG8 levels is detrimental for malaria infectivity. IMPORTANCE: Malaria is responsible for more mortality than any other parasitic disease. Resistance to antimalarial medicines is a recurring problem; new drugs are urgently needed. A key to the parasite's successful intracellular development in the liver is the metabolic changes necessary to convert the parasite from a sporozoite to a replication-competent, metabolically active trophozoite form. Our study reinforces the burgeoning concept that organellar changes during parasite differentiation are mediated by an autophagy-like process. We have identified ATG8 in Plasmodium liver forms as an important effector that controls the development and fate of organelles, e.g., the clearance of micronemes that are required for hepatocyte invasion and the expansion of the apicoplast that produces many metabolites indispensable for parasite replication. Given the unconventional properties and the importance of ATG8 for parasite development in hepatocytes, targeting the parasite's autophagic pathway may represent a novel approach to control malarial infections.


Asunto(s)
Familia de las Proteínas 8 Relacionadas con la Autofagia/genética , Hígado/parasitología , Proteínas de la Membrana/genética , Merozoítos/fisiología , Plasmodium berghei/genética , Plasmodium berghei/fisiología , Proteína Transportadora de Acilo/metabolismo , Animales , Apicoplastos , Autofagia , Hepatocitos/parasitología , Humanos , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Merozoítos/crecimiento & desarrollo , Ratones Transgénicos , Mutación , Orgánulos , Plasmodium berghei/citología , Plasmodium berghei/crecimiento & desarrollo , Proteínas Protozoarias/metabolismo
4.
J Biol Chem ; 291(8): 3725-46, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26694607

RESUMEN

The protozoan parasite Toxoplasma gondii develops within a parasitophorous vacuole (PV) in mammalian cells, where it scavenges cholesterol. When cholesterol is present in excess in its environment, the parasite expulses this lipid into the PV or esterifies it for storage in lipid bodies. Here, we characterized a unique T. gondii homologue of mammalian lecithin:cholesterol acyltransferase (LCAT), a key enzyme that produces cholesteryl esters via transfer of acyl groups from phospholipids to the 3-OH of free cholesterol, leading to the removal of excess cholesterol from tissues. TgLCAT contains a motif characteristic of serine lipases "AHSLG" and the catalytic triad consisting of serine, aspartate, and histidine (SDH) from LCAT enzymes. TgLCAT is secreted by the parasite, but unlike other LCAT enzymes it is cleaved into two proteolytic fragments that share the residues of the catalytic triad and need to be reassembled to reconstitute enzymatic activity. TgLCAT uses phosphatidylcholine as substrate to form lysophosphatidylcholine that has the potential to disrupt membranes. The released fatty acid is transferred to cholesterol, but with a lower transesterification activity than mammalian LCAT. TgLCAT is stored in a subpopulation of dense granule secretory organelles, and following secretion, it localizes to the PV and parasite plasma membrane. LCAT-null parasites have impaired growth in vitro, reduced virulence in animals, and exhibit delays in egress from host cells. Parasites overexpressing LCAT show increased virulence and faster egress. These observations demonstrate that TgLCAT influences the outcome of an infection, presumably by facilitating replication and egress depending on the developmental stage of the parasite.


Asunto(s)
Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/enzimología , Toxoplasma/patogenicidad , Toxoplasmosis/enzimología , Dominio Catalítico , Línea Celular , Humanos , Fosfatidilcolina-Esterol O-Aciltransferasa/química , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Toxoplasma/genética , Toxoplasmosis/genética , Toxoplasmosis/patología
5.
Autophagy ; 10(2): 269-84, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24342964

RESUMEN

Plasmodium parasites successfully colonize different habitats within mammals and mosquitoes, and adaptation to various environments is accompanied by changes in their organelle composition and size. Previously, we observed that during hepatocyte infection, Plasmodium discards organelles involved in invasion and expands those implicated in biosynthetic pathways. We hypothesized that this process is regulated by autophagy. Plasmodium spp. possess a rudimentary set of known autophagy-related proteins that includes the ortholog of yeast Atg8. In this study, we analyzed the activity of the ATG8-conjugation pathway over the course of the lifecycle of Plasmodium falciparum and during the liver stage of Plasmodium berghei. We engineered a transgenic P. falciparum strain expressing mCherry-PfATG8. These transgenic parasites expressed mCherry-PfATG8 in human hepatocytes and erythrocytes, and in the midgut and salivary glands of Anopheles mosquitoes. In all observed stages, mCherry-PfATG8 was localized to tubular structures. Our EM and colocalization studies done in P. berghei showed the association of PbATG8 on the limiting membranes of the endosymbiont-derived plastid-like organelle known as the apicoplast. Interestingly, during parasite replication in hepatocytes, the association of PbATG8 with the apicoplast increases as this organelle expands in size. PbATG3, PbATG7 and PbATG8 are cotranscribed in all parasitic stages. Molecular analysis of PbATG8 and PbATG3 revealed a novel mechanism of interaction compared with that observed for other orthologs. This is further supported by the inability of Plasmodium ATG8 to functionally complement atg8Δ yeast or localize to autophagosomes in starved mammalian cells. Altogether, these data suggests a unique role for the ATG8-conjugation system in Plasmodium parasites.


Asunto(s)
Apicoplastos/inmunología , Autofagia/inmunología , Hígado/microbiología , Parásitos/inmunología , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Animales , Antígenos de Protozoos/inmunología , Familia de las Proteínas 8 Relacionadas con la Autofagia , Femenino , Hepatocitos/metabolismo , Hígado/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/inmunología , Parásitos/metabolismo , Fagosomas/inmunología , Saccharomyces cerevisiae/inmunología , Proteínas de Saccharomyces cerevisiae/inmunología
6.
Cell Microbiol ; 15(7): 1182-97, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23311949

RESUMEN

Cryptosporidium spp. are responsible for devastating diarrhoea in immunodeficient individuals. In the intestinal tract, the developmental stages of the parasite are confined to the apical surfaces of epithelial cells. Upon invasion, Cryptosporidium incorporates the microvillous membrane of the enterocyte to form the parasitophorous vacuole (PV) and sequesters itself from the host cytoplasm by rearranging the host cytoskeleton. Cryptosporidium parvum has minimal anabolic capabilities and relies on transporters and salvage pathways to meet its basic metabolic requirements. The cholesterol salvage pathway is crucial for the development of protozoan parasites. In this study, we have examined the sources of cholesterol from C. parvum infecting enterocytes. We illustrated that the intracellular stages of Cryptosporidium as well as the oocysts shed by the host, contain cholesterol. Incubation of infected enterocytes in lipoprotein-free medium impairs parasite development and results in substantial decrease in cholesterol content associated with the PV. Among lipoproteins, LDL constitutes an important source of cholesterol for Cryptosporidium. Dietary cholesterol incorporated into micelles is internalized into enterocytes by the NPC1L1 transporter. We showed that C. parvum also obtains cholesterol from micelles in enterocytes.Pharmacological blockade of NPC1L1 function by ezetimibe or moderate downregulation of NPC1L1 expression decreases parasite infectivity. These observations indicate that, despite its dual sequestration from the intestinal lumen and the host cytoplasm, C. parvum can, in fact, obtain cholesterol both from the gut's lumen and the host cell. This study highlights the evolutionary advantages for epicellular pathogens to access to nutrients from the outside and inside of the host cell.


Asunto(s)
LDL-Colesterol/metabolismo , Cryptosporidium parvum/metabolismo , Enterocitos/metabolismo , Enterocitos/parasitología , Línea Celular , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Microscopía , Modelos Biológicos
7.
Eukaryot Cell ; 12(2): 265-77, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23243063

RESUMEN

The prokaryote Chlamydia trachomatis and the protozoan Toxoplasma gondii, two obligate intracellular pathogens of humans, have evolved a similar modus operandi to colonize their host cell and salvage nutrients from organelles. In order to gain fundamental knowledge on the pathogenicity of these microorganisms, we have established a cell culture model whereby single fibroblasts are coinfected by C. trachomatis and T. gondii. We previously reported that the two pathogens compete for the same nutrient pools in coinfected cells and that Toxoplasma holds a significant competitive advantage over Chlamydia. Here we have expanded our coinfection studies by examining the respective abilities of Chlamydia and Toxoplasma to co-opt the host cytoskeleton and recruit organelles. We demonstrate that the two pathogen-containing vacuoles migrate independently to the host perinuclear region and rearrange the host microtubular network around each vacuole. However, Toxoplasma outcompetes Chlamydia to the host microtubule-organizing center to the detriment of the bacterium, which then shifts to a stress-induced persistent state. Solely in cells preinfected with Chlamydia, the centrosomes become associated with the chlamydial inclusion, while the Toxoplasma parasitophorous vacuole displays growth defects. Both pathogens fragment the host Golgi apparatus and recruit Golgi elements to retrieve sphingolipids. This study demonstrates that the productive infection by both Chlamydia and Toxoplasma depends on the capability of each pathogen to successfully adhere to a finely tuned developmental program that aims to remodel the host cell for the pathogen's benefit. In particular, this investigation emphasizes the essentiality of host organelle interception by intravacuolar pathogens to facilitate access to nutrients.


Asunto(s)
Infecciones por Chlamydia/microbiología , Chlamydia/fisiología , Toxoplasma/fisiología , Toxoplasmosis/parasitología , Células Cultivadas , Centrosoma/metabolismo , Centrosoma/microbiología , Centrosoma/parasitología , Ceramidas/metabolismo , Infecciones por Chlamydia/parasitología , Infecciones por Chlamydia/patología , Coinfección , Fibroblastos/microbiología , Fibroblastos/parasitología , Fibroblastos/patología , Aparato de Golgi/microbiología , Aparato de Golgi/parasitología , Aparato de Golgi/patología , Interacciones Huésped-Parásitos , Interacciones Huésped-Patógeno , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/microbiología , Membranas Intracelulares/parasitología , Viabilidad Microbiana , Microtúbulos/metabolismo , Microtúbulos/microbiología , Microtúbulos/parasitología , Mitocondrias/microbiología , Mitocondrias/parasitología , Mitocondrias/patología , Toxoplasmosis/microbiología , Toxoplasmosis/patología , Vacuolas/microbiología , Vacuolas/parasitología
8.
Cell Microbiol ; 15(4): 619-46, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23107293

RESUMEN

Toxoplasma and Chlamydia trachomatis are obligate intracellular pathogens that have evolved analogous strategies to replicate within mammalian cells. Both pathogens are known to extensively remodel the cytoskeleton, and to recruit endocytic and exocytic organelles to their respective vacuoles. However, how important these activities are for infectivity by either pathogen remains elusive. Here, we have developed a novel co-infection system to gain insights into the developmental cycles of Toxoplasma and C. trachomatis by infecting human cells with both pathogens, and examining their respective ability to replicate and scavenge nutrients. We hypothesize that the common strategies used by Toxoplasma and Chlamydia to achieve development results in direct competition of the two pathogens for the same pool of nutrients. We show that a single human cell can harbour Chlamydia and Toxoplasma. In co-infected cells, Toxoplasma is able to divert the content of host organelles, such as cholesterol. Consequently, the infectious cycle of Toxoplasma progresses unimpeded. In contrast, Chlamydia's ability to scavenge selected nutrients is diminished, and the bacterium shifts to a stress-induced persistent growth. Parasite killing engenders an ordered return to normal chlamydial development. We demonstrate that C. trachomatis enters a stress-induced persistence phenotype as a direct result from being barred from its normal nutrient supplies as addition of excess nutrients, e.g. amino acids, leads to substantial recovery of Chlamydia growth and infectivity. Co-infection of C. trachomatis with slow growing strains of Toxoplasma or a mutant impaired in nutrient acquisition does not restrict chlamydial development. Conversely, Toxoplasma growth is halted in cells infected with the highly virulent Chlamydia psittaci. This study illustrates the key role that cellular remodelling plays in the exploitation of host intracellular resources by Toxoplasma and Chlamydia. It further highlights the delicate balance between success and failure of infection by intracellular pathogens in a co-infection system at the cellular level.


Asunto(s)
Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/metabolismo , Alimentos , Toxoplasma/crecimiento & desarrollo , Toxoplasma/metabolismo , Células Cultivadas , Humanos , Interacciones Microbianas , Estrés Fisiológico
9.
PLoS Pathog ; 7(12): e1002410, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22174676

RESUMEN

Several proteins that play key roles in cholesterol synthesis, regulation, trafficking and signaling are united by sharing the phylogenetically conserved 'sterol-sensing domain' (SSD). The intracellular parasite Toxoplasma possesses at least one gene coding for a protein containing the canonical SSD. We investigated the role of this protein to provide information on lipid regulatory mechanisms in the parasite. The protein sequence predicts an uncharacterized Niemann-Pick, type C1-related protein (NPC1) with significant identity to human NPC1, and it contains many residues implicated in human NPC disease. We named this NPC1-related protein, TgNCR1. Mammalian NPC1 localizes to endo-lysosomes and promotes the movement of sterols and sphingolipids across the membranes of these organelles. Miscoding patient mutations in NPC1 cause overloading of these lipids in endo-lysosomes. TgNCR1, however, lacks endosomal targeting signals, and localizes to flattened vesicles beneath the plasma membrane of Toxoplasma. When expressed in mammalian NPC1 mutant cells and properly addressed to endo-lysosomes, TgNCR1 restores cholesterol and GM1 clearance from these organelles. To clarify the role of TgNCR1 in the parasite, we genetically disrupted NCR1; mutant parasites were viable. Quantitative lipidomic analyses on the ΔNCR1 strain reveal normal cholesterol levels but an overaccumulation of several species of cholesteryl esters, sphingomyelins and ceramides. ΔNCR1 parasites are also characterized by abundant storage lipid bodies and long membranous tubules derived from their parasitophorous vacuoles. Interestingly, these mutants can generate multiple daughters per single mother cell at high frequencies, allowing fast replication in vitro, and they are slightly more virulent in mice than the parental strain. These data suggest that the ΔNCR1 strain has lost the ability to control the intracellular levels of several lipids, which subsequently results in the stimulation of lipid storage, membrane biosynthesis and parasite division. Based on these observations, we ascribe a role for TgNCR1 in lipid homeostasis in Toxoplasma.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Femenino , Técnicas de Inactivación de Genes , Humanos , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteína Niemann-Pick C1 , Reacción en Cadena de la Polimerasa , Proteínas Protozoarias/genética , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Toxoplasma/genética , Toxoplasma/patogenicidad , Toxoplasmosis Animal/genética , Toxoplasmosis Animal/metabolismo
10.
Mol Microbiol ; 76(5): 1232-49, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20487267

RESUMEN

Toxoplasma is a protozoan parasite proficiently adapted to thrive in a parasitophorous vacuole (PV) formed in the cytoplasm of a large variety of mammalian cells. As an actively dividing organism, the parasite must adjust the lipid composition of its membranes to preserve organelle vitality and expand the size of the PV membrane to accommodate growing progeny. We showed that Toxoplasma takes up host lipids and can expel major lipids in an ATP-dependent process. In order to provide detailed mechanistic insights into lipid trafficking phenomena relevant to Toxoplasma biology, we characterized six parasite ATP-binding cassette (ABC) G family transporters and investigated their potential contribution to lipid homeostatic processes. All these transporters are expressed in the parasite and five of them are upregulated upon exposure to sterols. Four ABCG are localized to secretory organelles and the plasma membrane, and promote cholesterol and phospholipid efflux, reflecting the importance in exportation of large amounts of lipids into the PV. Interestingly, one ABCG that is associated with vesicles in the PV and the plasma membrane acts as a cholesterol importer. This last finding expands our current view on the role of some ABCG transporters in eukaryotic sterol influx.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Metabolismo de los Lípidos , Isoformas de Proteínas/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Secuencia de Aminoácidos , Animales , Transporte Biológico/fisiología , Células COS , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Interacciones Huésped-Parásitos , Humanos , Datos de Secuencia Molecular , Organismos Modificados Genéticamente , Fosfolípidos/metabolismo , Isoformas de Proteínas/genética , Proteínas Protozoarias/genética , Alineación de Secuencia , Esteroles/metabolismo , Toxoplasma/citología , Toxoplasma/genética
11.
J Biol Chem ; 279(2): 837-47, 2004 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-14551196

RESUMEN

Developing Dictyostelium cells form structures containing approximately 20,000 cells. The size regulation mechanism involves a secreted counting factor (CF) repressing cytosolic glucose levels. Glucose or a glucose metabolite affects cell-cell adhesion and motility; these in turn affect whether a group stays together, loses cells, or even breaks up. NADPH-coupled aldehyde reductase reduces a wide variety of aldehydes to the corresponding alcohols, including converting glucose to sorbitol. The levels of this enzyme previously appeared to be regulated by CF. We find that disrupting alrA, the gene encoding aldehyde reductase, results in the loss of alrA mRNA and AlrA protein and a decrease in the ability of cell lysates to reduce both glyceraldehyde and glucose in an NADPH-coupled reaction. Counterintuitively, alrA- cells grow normally and have decreased glucose levels compared with parental cells. The alrA- cells form long unbroken streams and huge groups. Expression of AlrA in alrA- cells causes cells to form normal fruiting bodies, indicating that AlrA affects group size. alrA- cells have normal adhesion but a reduced motility, and computer simulations suggest that this could indeed result in the formation of large groups. alrA- cells secrete low levels of countin and CF50, two components of CF, and this could partially account for why alrA- cells form large groups. alrA- cells are responsive to CF and are partially responsive to recombinant countin and CF50, suggesting that disrupting alrA inhibits but does not completely block the CF signal transduction pathway. Gas chromatography/mass spectroscopy indicates that the concentrations of several metabolites are altered in alrA- cells, suggesting that the Dictyostelium aldehyde reductase affects several metabolic pathways in addition to converting glucose to sorbitol. Together, our data suggest that disrupting alrA affects CF secretion, causes many effects on cellular metabolism, and has a major effect on group size.


Asunto(s)
Aldehído Reductasa/genética , Agregación Celular , Dictyostelium/enzimología , Aldehído Reductasa/metabolismo , Secuencia de Aminoácidos , Animales , Northern Blotting , Western Blotting , Adhesión Celular , Línea Celular , Movimiento Celular , Simulación por Computador , AMP Cíclico/metabolismo , ADN/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Glucosa/metabolismo , Gliceraldehído/metabolismo , Datos de Secuencia Molecular , NADP/metabolismo , Proteínas Protozoarias/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Recombinación Genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sorbitol/metabolismo , Fracciones Subcelulares , Factores de Tiempo
12.
J Biol Chem ; 278(52): 52262-72, 2003 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-14557265

RESUMEN

A secreted 450-kDa complex of proteins called counting factor (CF) is part of a negative feedback loop that regulates the size of the groups formed by developing Dictyostelium cells. Two components of CF are countin and CF50. Both recombinant countin and recombinant CF50 decrease group size in Dictyostelium. countin- cells have a decreased cAMP-stimulated cAMP pulse, whereas recombinant countin potentiates the cAMP pulse. We find that CF50 cells have an increased cAMP pulse, whereas recombinant CF50 decreases the cAMP pulse, suggesting that countin and CF50 have opposite effects on cAMP signal transduction. In addition, countin and CF50 have opposite effects on cAMP-stimulated Erk2 activation. However, like recombinant countin, recombinant CF50 increases cell motility. We previously found that cells bind recombinant countin with a Hill coefficient of approximately 2, a KH of 60 pm, and approximately 53 sites/cell. We find here that cells also bind 125I-recombinant CF50, with a Hill coefficient of approximately 2, a KH of approximately 15 ng/ml (490 pm), and approximately 56 sites/cell. Countin and CF50 require each other's presence to affect group size, but the presence of countin is not necessary for CF50 to bind to cells, and CF50 is not necessary for countin to bind to cells. Our working hypothesis is that a signal transduction pathway activated by countin binding to cells modulates a signal transduction pathway activated by CF50 binding to cells and vice versa and that these two pathways can be distinguished by their effects on cAMP signal transduction.


Asunto(s)
AMP Cíclico/metabolismo , Dictyostelium/fisiología , Proteínas Protozoarias/fisiología , Transducción de Señal , Animales , Movimiento Celular , Células Cultivadas , GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Cinética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Unión Proteica , Proteínas Protozoarias/metabolismo , Receptores de AMP Cíclico/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Ultracentrifugación
13.
J Biol Chem ; 277(36): 32596-605, 2002 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-12070154

RESUMEN

In Dictyostelium discoideum counting factor (CF), a secreted approximately 450-kDa complex of polypeptides, inhibits group and fruiting body size. When the gene encoding countin (a component of CF) was disrupted, cells formed large groups. We find that recombinant countin causes developing cells to form small groups, with an EC(50) of approximately 3 ng/ml, and affects cAMP signal transduction in the same manner as semipurified CF. Recombinant countin increases cell motility, decreases cell-cell adhesion, and regulates gene expression in a manner similar to the effect of CF. However, countin does not decrease adhesion or group size to the extent that semipurified CF does. A 1-min exposure of developing cells to countin causes an increase in F-actin polymerization and myosin phosphorylation and a decrease in myosin polymerization, suggesting that countin activates a rapid signal transduction pathway. (125)I-Labeled countin has countin bioactivity, and binding experiments suggest that vegetative and developing cells have approximately 53 cell-surface sites that bind countin with a K(D) of approximately 1.5 ng/ml or 60 pm. We hypothesize that countin regulates cell development through the same pathway as CF and that other proteins within the complex may modify the activity of countin and/or have independent size-regulating activities.


Asunto(s)
Dictyostelium/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/fisiología , Actinas/metabolismo , Animales , Sitios de Unión , Adhesión Celular , Movimiento Celular , Cromatografía en Gel , Clonación Molecular , AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , ADN Complementario/metabolismo , Dimerización , Relación Dosis-Respuesta a Droga , Yodo/farmacología , Cinética , Miosinas/metabolismo , Fosforilación , Unión Proteica , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Tiempo , Ultracentrifugación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA