Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Cell Sci ; 134(5)2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33093240

RESUMEN

Endocytosis of caveolae has previously been implicated in the repair of plasma membrane wounds. Here, we show that caveolin-1-deficient fibroblasts lacking caveolae upregulate a tubular endocytic pathway and have a reduced capacity to reseal after permeabilization with pore-forming toxins compared with wild-type cells. Silencing endophilin-A2 expression inhibited fission of endocytic tubules and further reduced plasma membrane repair in cells lacking caveolin-1, supporting a role for tubular endocytosis as an alternative pathway for the removal of membrane lesions. Endophilin-A2 was visualized in association with cholera toxin B-containing endosomes and was recruited to recently formed intracellular vacuoles containing Trypanosoma cruzi, a parasite that utilizes the plasma membrane wounding repair pathway to invade host cells. Endophilin-A2 deficiency inhibited T. cruzi invasion, and fibroblasts deficient in both caveolin-1 and endophilin-A2 did not survive prolonged exposure to the parasites. These findings reveal a novel crosstalk between caveolin-1 and endophilin-A2 in the regulation of clathrin-independent endocytosis and plasma membrane repair, a process that is subverted by T. cruzi parasites for cell invasion.


Asunto(s)
Aciltransferasas/fisiología , Membrana Celular , Endocitosis , Trypanosoma cruzi , Células 3T3 , Animales , Caveolas , Clatrina , Ratones
2.
Nat Immunol ; 11(6): 495-502, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20473299

RESUMEN

Chemokines and other chemoattractants direct leukocyte migration and are essential for the development and delivery of immune and inflammatory responses. To probe the molecular mechanisms that underlie chemoattractant-guided migration, we did an RNA-mediated interference screen that identified several members of the synaptotagmin family of calcium-sensing vesicle-fusion proteins as mediators of cell migration: SYT7 and SYTL5 were positive regulators of chemotaxis, whereas SYT2 was a negative regulator of chemotaxis. SYT7-deficient leukocytes showed less migration in vitro and in a gout model in vivo. Chemoattractant-induced calcium-dependent lysosomal fusion was impaired in SYT7-deficient neutrophils. In a chemokine gradient, SYT7-deficient lymphocytes accumulated lysosomes in their uropods and had impaired uropod release. Our data identify a molecular pathway required for chemotaxis that links chemoattractant-induced calcium flux to exocytosis and uropod release.


Asunto(s)
Movimiento Celular/fisiología , Sinaptotagminas/metabolismo , Animales , Quimiocina CXCL12/metabolismo , Quimiotaxis , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores CXCR4/metabolismo , Sinaptotagmina II/metabolismo , Sinaptotagminas/genética , Linfocitos T/inmunología
3.
Hum Mol Genet ; 27(5): 811-822, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29301038

RESUMEN

Gaucher disease (GD) is caused by bi-allelic mutations in GBA1, the gene that encodes acid ß-glucocerebrosidase (GCase). Individuals affected by GD have hematologic, visceral and bone abnormalities, and in severe cases there is also neurodegeneration. To shed light on the mechanisms by which mutant GBA1 causes bone disease, we examined the ability of human induced pluripotent stem cells (iPSC) derived from patients with Types 1, 2 and 3 GD, to differentiate to osteoblasts and carry out bone deposition. Differentiation of GD iPSC to osteoblasts revealed that these cells had developmental defects and lysosomal abnormalities that interfered with bone matrix deposition. Compared with controls, GD iPSC-derived osteoblasts exhibited reduced expression of osteoblast differentiation markers, and bone matrix protein and mineral deposition were defective. Concomitantly, canonical Wnt/ß catenin signaling in the mutant osteoblasts was downregulated, whereas pharmacological Wnt activation with the GSK3ß inhibitor CHIR99021 rescued GD osteoblast differentiation and bone matrix deposition. Importantly, incubation with recombinant GCase (rGCase) rescued the differentiation and bone-forming ability of GD osteoblasts, demonstrating that the abnormal GD phenotype was caused by GCase deficiency. GD osteoblasts were also defective in their ability to carry out Ca2+-dependent exocytosis, a lysosomal function that is necessary for bone matrix deposition. We conclude that normal GCase enzymatic activity is required for the differentiation and bone-forming activity of osteoblasts. Furthermore, the rescue of bone matrix deposition by pharmacological activation of Wnt/ß catenin in GD osteoblasts uncovers a new therapeutic target for the treatment of bone abnormalities in GD.


Asunto(s)
Enfermedad de Gaucher/patología , Células Madre Pluripotentes Inducidas/patología , Lisosomas/patología , Osteoblastos/patología , Matriz Ósea/patología , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Exocitosis/genética , Enfermedad de Gaucher/genética , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Lisosomas/genética , Mutación , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Vía de Señalización Wnt , beta Catenina/metabolismo
4.
PLoS Pathog ; 14(6): e1007140, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29906288

RESUMEN

Iron is essential for many cellular processes, but can generate highly toxic hydroxyl radicals in the presence of oxygen. Therefore, intracellular iron accumulation must be tightly regulated, by balancing uptake with storage or export. Iron uptake in Leishmania is mediated by the coordinated action of two plasma membrane proteins, the ferric iron reductase LFR1 and the ferrous iron transporter LIT1. However, how these parasites regulate their cytosolic iron concentration to prevent toxicity remains unknown. Here we characterize Leishmania Iron Regulator 1 (LIR1), an iron responsive protein with similarity to membrane transporters of the major facilitator superfamily (MFS) and plant nodulin-like proteins. LIR1 localizes on the plasma membrane of L. amazonensis promastigotes and intracellular amastigotes. After heterologous expression in Arabidopsis thaliana, LIR1 decreases the iron content of leaves and worsens the chlorotic phenotype of plants lacking the iron importer IRT1. Consistent with a role in iron efflux, LIR1 deficiency does not affect iron uptake by L. amazonensis but significantly increases the amount of iron retained intracellularly in the parasites. LIR1 null parasites are more sensitive to iron toxicity and have drastically impaired infectivity, phenotypes that are reversed by LIR1 complementation. We conclude that LIR1 functions as a plasma membrane iron exporter with a critical role in maintaining iron homeostasis and promoting infectivity in L. amazonensis.


Asunto(s)
Membrana Celular/metabolismo , Hierro/farmacología , Leishmania/efectos de los fármacos , Leishmaniasis/prevención & control , Proteínas Protozoarias/metabolismo , Virulencia/efectos de los fármacos , Animales , Arabidopsis/citología , Arabidopsis/efectos de los fármacos , Arabidopsis/parasitología , Transporte Biológico , Células Cultivadas , Femenino , Homeostasis , Hierro/toxicidad , Leishmaniasis/metabolismo , Leishmaniasis/parasitología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Proteínas Protozoarias/genética
5.
Cell Microbiol ; 21(11): e13065, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31155842

RESUMEN

Acid sphingomyelinase (ASM) is a lysosomal enzyme that cleaves the phosphorylcholine head group of sphingomyelin, generating ceramide. Recessive mutations in SMPD1, the gene encoding ASM, cause Niemann-Pick Disease Types A and B. These disorders are attributed not only to lipid accumulation inside lysosomes but also to changes on the outer leaflet of the plasma membrane, highlighting an extracellular role for ASM. Secretion of ASM occurs under physiological conditions, and earlier studies proposed two forms of the enzyme, one resident in lysosomes and another form that would be diverted to the secretory pathway. Such differential intracellular trafficking has been difficult to explain because there is only one SMPD1 transcript that generates an active enzyme, found primarily inside lysosomes. Unexpectedly, studies of cell invasion by the protozoan parasite Trypanosoma cruzi revealed that conventional lysosomes can fuse with the plasma membrane in response to elevations in intracellular Ca2+ , releasing their contents extracellularly. ASM exocytosed from lysosomes remodels the outer leaflet of the plasma membrane, promoting parasite invasion and wound repair. Here, we discuss the possibility that ASM release during lysosomal exocytosis, in response to various forms of stress, may represent a major source of the secretory form of this enzyme.


Asunto(s)
Membrana Celular/parasitología , Lisosomas/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Trypanosoma cruzi/patogenicidad , Animales , Secreciones Corporales/efectos de la radiación , Calcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/patología , Ceramidas/metabolismo , Exocitosis , Humanos , Lisosomas/metabolismo , Enfermedad de Niemann-Pick Tipo A/enzimología , Enfermedad de Niemann-Pick Tipo B/enzimología , Transporte de Proteínas , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Esfingomielinas/metabolismo , Trypanosoma cruzi/metabolismo
6.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31527128

RESUMEN

The molecular mechanisms underlying biological differences between two Leishmania species that cause cutaneous disease, L. major and L. amazonensis, are poorly understood. In L. amazonensis, reactive oxygen species (ROS) signaling drives differentiation of nonvirulent promastigotes into forms capable of infecting host macrophages. Tight spatial and temporal regulation of H2O2 is key to this signaling mechanism, suggesting a role for ascorbate-dependent peroxidase (APX), which degrades mitochondrial H2O2 Earlier studies showed that APX-null L. major parasites are viable, accumulate higher levels of H2O2, generate a greater yield of infective metacyclic promastigotes, and have increased virulence. In contrast, we found that in L. amazonensis, the ROS-inducible APX is essential for survival of all life cycle stages. APX-null promastigotes could not be generated, and parasites carrying a single APX allele were impaired in their ability to infect macrophages and induce cutaneous lesions in mice. Similar to what was reported for L. major, APX depletion in L. amazonensis enhanced differentiation of metacyclic promastigotes and amastigotes, but the parasites failed to replicate after infecting macrophages. APX expression restored APX single-knockout infectivity, while expression of catalytically inactive APX drastically reduced virulence. APX overexpression in wild-type promastigotes reduced metacyclogenesis, but enhanced intracellular survival following macrophage infection or inoculation into mice. Collectively, our data support a role for APX-regulated mitochondrial H2O2 in promoting differentiation of virulent forms in both L. major and L. amazonensis Our results also uncover a unique requirement for APX-mediated control of ROS levels for survival and successful intracellular replication of L. amazonensis.


Asunto(s)
Ascorbato Peroxidasas/metabolismo , Leishmania major/patogenicidad , Leishmania mexicana/patogenicidad , Leishmaniasis Cutánea/patología , Macrófagos/parasitología , Especies Reactivas de Oxígeno/metabolismo , Animales , Ascorbato Peroxidasas/genética , Células Cultivadas , Leishmania major/genética , Leishmania major/metabolismo , Leishmania mexicana/genética , Leishmania mexicana/metabolismo , Leishmaniasis Cutánea/parasitología , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/fisiología , Virulencia
7.
J Biol Chem ; 292(29): 12324-12338, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28550086

RESUMEN

Leishmaniasis is one of the leading globally neglected diseases, affecting millions of people worldwide. Leishmania infection depends on the ability of insect-transmitted metacyclic promastigotes to invade mammalian hosts, differentiate into amastigotes, and replicate inside macrophages. To counter the hostile oxidative environment inside macrophages, these protozoans contain anti-oxidant systems that include iron-dependent superoxide dismutases (SODs) in mitochondria and glycosomes. Increasing evidence suggests that in addition to this protective role, Leishmania mitochondrial SOD may also initiate H2O2-mediated redox signaling that regulates gene expression and metabolic changes associated with differentiation into virulent forms. To investigate this hypothesis, we examined the specific role of SODA, the mitochondrial SOD isoform in Leishmania amazonensis Our inability to generate L. amazonensis SODA null mutants and the lethal phenotype observed following RNAi-mediated silencing of the Trypanosoma brucei SODA ortholog suggests that SODA is essential for trypanosomatid survival. L. amazonensis metacyclic promastigotes lacking one SODA allele failed to replicate in macrophages and were severely attenuated in their ability to generate cutaneous lesions in mice. Reduced expression of SODA also resulted in mitochondrial oxidative damage and failure of SODA/ΔsodA promastigotes to differentiate into axenic amastigotes. SODA expression above a critical threshold was also required for the development of metacyclic promastigotes, as SODA/ΔsodA cultures were strongly depleted in this infective form and more susceptible to reactive oxygen species (ROS)-induced stress. Collectively, our data suggest that SODA promotes Leishmania virulence by protecting the parasites against mitochondrion-generated oxidative stress and by initiating ROS-mediated signaling mechanisms required for the differentiation of infective forms.


Asunto(s)
Hierro/metabolismo , Leishmania mexicana/enzimología , Mitocondrias/enzimología , Proteínas Protozoarias/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/parasitología , Células de la Médula Ósea/patología , Línea Celular , Células Cultivadas , Células Clonales , Femenino , Técnicas de Inactivación de Genes , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Leishmania mexicana/crecimiento & desarrollo , Leishmania mexicana/patogenicidad , Leishmania mexicana/ultraestructura , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/metabolismo , Leishmaniasis Cutánea/parasitología , Leishmaniasis Cutánea/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/parasitología , Macrófagos/patología , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Carga de Parásitos , Transporte de Proteínas , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Interferencia de ARN , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/genética , Virulencia
8.
PLoS Pathog ; 12(1): e1005340, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26741360

RESUMEN

Iron, an essential co-factor of respiratory chain proteins, is critical for mitochondrial function and maintenance of its redox balance. We previously reported a role for iron uptake in differentiation of Leishmania amazonensis into virulent amastigotes, by a mechanism that involves reactive oxygen species (ROS) production and is independent of the classical pH and temperature cues. Iron import into mitochondria was proposed to be essential for this process, but evidence supporting this hypothesis was lacking because the Leishmania mitochondrial iron transporter was unknown. Here we describe MIT1, a homolog of the mitochondrial iron importer genes mrs3 (yeast) and mitoferrin-1 (human) that is highly conserved among trypanosomatids. MIT1 expression was essential for the survival of Trypanosoma brucei procyclic but not bloodstream forms, which lack functional respiratory complexes. L. amazonensis LMIT1 null mutants could not be generated, suggesting that this mitochondrial iron importer is essential for promastigote viability. Promastigotes lacking one LMIT1 allele (LMIT1/Δlmit1) showed growth defects and were more susceptible to ROS toxicity, consistent with the role of iron as the essential co-factor of trypanosomatid mitochondrial superoxide dismutases. LMIT1/Δlmit1 metacyclic promastigotes were unable to replicate as intracellular amastigotes after infecting macrophages or cause cutaneous lesions in mice. When induced to differentiate axenically into amastigotes, LMIT1/Δlmit1 showed strong defects in iron content and function of mitochondria, were unable to upregulate the ROS-regulatory enzyme FeSOD, and showed mitochondrial changes suggestive of redox imbalance. Our results demonstrate the importance of mitochondrial iron uptake in trypanosomatid parasites, and highlight the role of LMIT1 in the iron-regulated process that orchestrates differentiation of L. amazonensis into infective amastigotes.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Hierro/metabolismo , Leishmania/patogenicidad , Mitocondrias/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Leishmania/crecimiento & desarrollo , Leishmania/metabolismo , Leishmaniasis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Proteínas Protozoarias/genética , Virulencia
9.
Cell Microbiol ; 19(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27641840

RESUMEN

Leishmania is an intracellular protozoan parasite that causes a broad spectrum of clinical manifestations, ranging from self-healing skin lesions to fatal visceralizing disease. As the host cells of choice for all species of Leishmania, macrophages are critical for the establishment of infections. How macrophages contribute to parasite homing to specific tissues and how parasites modulate macrophage function are still poorly understood. In this study, we show that Leishmania amazonensis infection inhibits macrophage roaming motility. The reduction in macrophage speed is not dependent on particle load or on factors released by infected macrophages. L. amazonensis-infected macrophages also show reduced directional migration in response to the chemokine MCP-1. We found that infected macrophages have lower levels of total paxillin, phosphorylated paxillin, and phosphorylated focal adhesion kinase when compared to noninfected macrophages, indicating abnormalities in the formation of signaling adhesion complexes that regulate motility. Analysis of the dynamics of actin polymerization at peripheral sites also revealed a markedly enhanced F-actin turnover frequency in L. amazonensis-infected macrophages. Thus, Leishmania infection inhibits macrophage motility by altering actin dynamics and impairing the expression of proteins that function in plasma membrane-extracellular matrix interactions.


Asunto(s)
Actinas/metabolismo , Movimiento Celular , Leishmania mexicana/patogenicidad , Macrófagos/fisiología , Macrófagos/parasitología , Proteína-Tirosina Quinasas de Adhesión Focal/análisis , Macrófagos/química , Paxillin/análisis
10.
J Cell Sci ; 128(1): 27-32, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25380822

RESUMEN

The process of host cell invasion by Trypanosoma cruzi shares mechanistic elements with plasma membrane injury and repair. Both processes require Ca(2+)-triggered exocytosis of lysosomes, exocytosis of acid sphingomyelinase and formation of ceramide-enriched endocytic compartments. T. cruzi invades at peripheral sites, suggesting a need for spatial regulation of membrane traffic. Here, we show that Exo70 and Sec8 (also known as EXOC7 and EXOC4, respectively), components of the exocyst complex, accumulate in nascent T. cruzi vacuoles and at sites of mechanical wounding. Exo70 or Sec8 depletion inhibits T. cruzi invasion and Ca(2+)-dependent resealing of mechanical wounds, but does not affect the repair of smaller lesions caused by pore-forming toxins. Thus, T. cruzi invasion and mechanical lesion repair share a unique requirement for the exocyst, consistent with a dependence on targeted membrane delivery.


Asunto(s)
Membrana Celular/metabolismo , Enfermedad de Chagas/metabolismo , Endocitosis , Trypanosoma cruzi/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Calcio/metabolismo , Membrana Celular/patología , Enfermedad de Chagas/patología , Células HeLa , Humanos
11.
PLoS Pathog ; 10(1): e1003901, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24497831

RESUMEN

Leishmania parasites infect macrophages, cells that play an important role in organismal iron homeostasis. By expressing ferroportin, a membrane protein specialized in iron export, macrophages release iron stored intracellularly into the circulation. Iron is essential for the intracellular replication of Leishmania, but how the parasites compete with the iron export function of their host cell is unknown. Here, we show that infection with Leishmania amazonensis inhibits ferroportin expression in macrophages. In a TLR4-dependent manner, infected macrophages upregulated transcription of hepcidin, a peptide hormone that triggers ferroportin degradation. Parasite replication was inhibited in hepcidin-deficient macrophages and in wild type macrophages overexpressing mutant ferroportin that is resistant to hepcidin-induced degradation. Conversely, intracellular growth was enhanced by exogenously added hepcidin, or by expression of dominant-negative ferroportin. Importantly, dominant-negative ferroportin and macrophages from flatiron mice, a mouse model for human type IV hereditary hemochromatosis, restored the infectivity of mutant parasite strains defective in iron acquisition. Thus, inhibition of ferroportin expression is a specific strategy used by L. amazonensis to inhibit iron export and promote their own intracellular growth.


Asunto(s)
Hierro/metabolismo , Leishmania/metabolismo , Leishmaniasis/metabolismo , Macrófagos/metabolismo , Animales , Transporte Biológico Activo/genética , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hemocromatosis/parasitología , Hemocromatosis/patología , Hepcidinas/biosíntesis , Hepcidinas/genética , Humanos , Leishmaniasis/genética , Leishmaniasis/patología , Macrófagos/parasitología , Macrófagos/patología , Ratones , Ratones Noqueados , Mutación
12.
Traffic ; 13(3): 483-94, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22212686

RESUMEN

Cells permeabilized by the bacterial pore-forming toxin streptolysin O (SLO) reseal their plasma membrane in a Ca(2+) -dependent manner. Resealing involves Ca(2+) -dependent exocytosis of lysosomes, release of acid sphingomyelinase and rapid formation of endosomes that carry the transmembrane pores into the cell. The intracellular fate of the toxin-carrying endocytic vesicles, however, is still unknown. Here, we show that SLO pores removed from the plasma membrane by endocytosis are sorted into the lumen of lysosomes, where they are degraded. SLO-permeabilized cells contain elevated numbers of total endosomes, which increase gradually in size while transitioning from endosomes with flat clathrin coats to large multivesicular bodies (MVBs). Under conditions that allow endocytosis and plasma membrane repair, SLO is rapidly ubiquitinated and gradually degraded, in a process sensitive to inhibitors of lysosomal hydrolysis but not of proteasomes. The endosomes induced by SLO permeabilization become increasingly acidified and promote SLO degradation under normal conditions, but not in cells silenced for expression of Vps24, an ESCRT-III complex component required for the release of intraluminal vesicles into MVBs. Thus, cells dispose of SLO transmembrane pores by ubiquitination/ESCRT-dependent sorting into the lumen of late endosomes/lysosomes.


Asunto(s)
Toxinas Bacterianas/metabolismo , Permeabilidad de la Membrana Celular , Membrana Celular/metabolismo , Cuerpos Multivesiculares/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Western Blotting , Células Cultivadas , Endocitosis , Modelos Biológicos , Estreptolisinas/metabolismo
13.
PLoS Pathog ; 8(7): e1002795, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22807677

RESUMEN

Trypanosomatid protozoan parasites lack a functional heme biosynthetic pathway, so must acquire heme from the environment to survive. However, the molecular pathway responsible for heme acquisition by these organisms is unknown. Here we show that L. amazonensis LHR1, a homolog of the C. elegans plasma membrane heme transporter HRG-4, functions in heme transport. Tagged LHR1 localized to the plasma membrane and to endocytic compartments, in both L. amazonensis and mammalian cells. Heme deprivation in L. amazonensis increased LHR1 transcript levels, promoted uptake of the fluorescent heme analog ZnMP, and increased the total intracellular heme content of promastigotes. Conversely, deletion of one LHR1 allele reduced ZnMP uptake and the intracellular heme pool by approximately 50%, indicating that LHR1 is a major heme importer in L. amazonensis. Viable parasites with correct replacement of both LHR1 alleles could not be obtained despite extensive attempts, suggesting that this gene is essential for the survival of promastigotes. Notably, LHR1 expression allowed Saccharomyces cerevisiae to import heme from the environment, and rescued growth of a strain deficient in heme biosynthesis. Syntenic genes with high sequence identity to LHR1 are present in the genomes of several species of Leishmania and also Trypanosoma cruzi and Trypanosoma brucei, indicating that therapeutic agents targeting this transporter could be effective against a broad group of trypanosomatid parasites that cause serious human disease.


Asunto(s)
Hemo/metabolismo , Leishmania mexicana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Células HeLa , Hemo/deficiencia , Humanos , Leishmania mexicana/patogenicidad , Macrófagos/metabolismo , Macrófagos/parasitología , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Metaloporfirinas/metabolismo , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
14.
Infect Immun ; 81(10): 3620-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23876801

RESUMEN

The protozoan parasite Leishmania amazonensis is a heme auxotroph and must acquire this essential factor from the environment. Previous studies showed that L. amazonensis incorporates heme through the transmembrane protein LHR1 (Leishmania Heme Response 1). LHR1-null promastigotes were not viable, suggesting that the transporter is essential for survival. Here, we compared the growth, differentiation, and infectivity for macrophages and mice of wild-type, LHR1-single-knockout (LHR1/Δlhr1), and LHR1-complemented (LHR1/Δlhr1 plus LHR1) L. amazonensis strains. LHR1/Δlhr1 promastigotes replicated poorly in heme-deficient media and had lower intracellular heme content than wild-type parasites. LHR1/Δlhr1 promastigotes were also less effective in reducing ferric iron to ferrous iron, a reaction mediated by the heme-containing parasite enzyme LFR1 (Leishmania Ferric Reductase 1). LHR1/Δlhr1 parasites differentiated normally into aflagellated forms expressing amastigote-specific markers but were not able to replicate intracellularly after infecting macrophages. Importantly, the intracellular growth of LHR1/Δlhr1 amastigotes was fully restored when macrophages were allowed to phagocytose red blood cells prior to infection. LHR1/Δlhr1 parasites were also severely defective in the development of cutaneous lesions in mice. All phenotypes observed in LHR1/Δlhr1 L. amazonensis were rescued by expression of episomal LHR1. Our results reveal the importance of efficient heme uptake for L. amazonensis replication and vertebrate host infectivity, reinforcing the potential usefulness of LHR1 as a target for new antileishmanial drugs.


Asunto(s)
Hemo/metabolismo , Leishmania/patogenicidad , Macrófagos/parasitología , Proteínas Protozoarias/metabolismo , Animales , Eliminación de Gen , Leishmania/clasificación , Ratones , Proteínas Protozoarias/genética , Virulencia
15.
J Biol Chem ; 286(26): 23266-79, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558274

RESUMEN

The protozoan parasite Leishmania is the causative agent of serious human infections worldwide. The parasites alternate between insect and vertebrate hosts and cause disease by invading macrophages, where they replicate. Parasites lacking the ferrous iron transporter LIT1 cannot grow intracellularly, indicating that a plasma membrane-associated mechanism for iron uptake is essential for the establishment of infections. Here, we identify and functionally characterize a second member of the Leishmania iron acquisition pathway, the ferric iron reductase LFR1. The LFR1 gene is up-regulated under iron deprivation and accounts for all the detectable ferric reductase activity exposed on the surface of Leishmania amazonensis. LFR1 null mutants grow normally as promastigote insect stages but are defective in differentiation into the vertebrate infective forms, metacyclic promastigotes and amastigotes. LFR1 overexpression partially restores the abnormal morphology of infective stages but markedly reduces parasite viability, precluding its ability to rescue LFR1 null replication in macrophages. However, LFR1 overexpression is not toxic for amastigotes lacking the ferrous iron transporter LIT1 and rescues their growth defect. In addition, the intracellular growth of both LFR1 and LIT1 null parasites is rescued in macrophages loaded with exogenous iron. This indicates that the Fe(3+) reductase LFR1 functions upstream of LIT1 and suggests that LFR1 overexpression results in excessive Fe(2+) production, which impairs parasite viability after intracellular transport by LIT1.


Asunto(s)
FMN Reductasa/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Hierro/metabolismo , Leishmania/enzimología , Leishmania/patogenicidad , Leishmaniasis/enzimología , Proteínas Protozoarias/biosíntesis , Secuencia de Aminoácidos , Animales , Células Cultivadas , FMN Reductasa/genética , Humanos , Leishmania/genética , Leishmaniasis/genética , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Protozoarias/genética
16.
J Exp Med ; 203(10): 2363-75, 2006 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-17000865

RESUMEN

Infection of mammalian hosts with Leishmania amazonensis depends on the remarkable ability of these parasites to replicate within macrophage phagolysosomes. A critical adaptation for survival in this harsh environment is an efficient mechanism for gaining access to iron. In this study, we identify and characterize LIT1, a novel L. amazonensis membrane protein with extensive similarity to IRT1, a ZIP family ferrous iron transporter from Arabidopsis thaliana. The ability of LIT1 to promote iron transport was demonstrated after expression in yeast and in L. amazonensis LIT1-null amastigotes. Endogenous LIT1 was only detectable in amastigotes replicating intracellularly, and its intracellular expression was accelerated under conditions predicted to result in iron deprivation. Although L. amazonensis lacking LIT1 grew normally in axenic culture and had no defects differentiating into infective forms, replication within macrophages was abolished. Consistent with an essential role for LIT1 in intracellular growth as amastigotes, Deltalit1 parasites were avirulent. After inoculation into highly susceptible mice, no lesions were detected, even after extensive periods of time. Despite the absence of pathology, viable Deltalit1 parasites were recovered from the original sites of inoculation, indicating that L. amazonensis can persist in vivo independently of the ability to grow in macrophages. Our findings highlight the essential role played by intracellular iron acquisition in Leishmania virulence and identify this pathway as a promising target for therapeutic intervention.


Asunto(s)
Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Leishmania/metabolismo , Leishmania/patogenicidad , Lisosomas/parasitología , Macrófagos/parasitología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartilla de ADN , Hierro/metabolismo , Leishmania/fisiología , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Reproducción/fisiología , Alineación de Secuencia , Análisis de Secuencia de ADN , Virulencia , Levaduras
17.
Science ; 376(6591): 346-347, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35446648

RESUMEN

Removing membrane pores may help cancer cells survive T cell assault.

18.
Front Cell Infect Microbiol ; 12: 824494, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35186797

RESUMEN

Although macrophages have long been considered key players in the course of Leishmania infections, other non-professional phagocytes have lately been shown to maintain low levels of the parasite in safe intracellular niches. Recently, it was demonstrated that the adipose tissue is capable of harboring Old World L. (L.) infantum in mice. However, there is no evidence of experimental adipocyte infection with New World Leishmania species so far. In addition, it was not known whether adipocytes would be permissive for formation of the unique, large and communal parasitophorous vacuoles that are typical of L. (L.) amazonensis in macrophages. Here we evaluated the ability of L. (L.) amazonensis and L. (V.) braziliensis promastigotes and amastigotes to infect 3T3-L1 fibroblast-derived adipocytes (3T3-Ad) using light and transmission electron microscopy. Our results indicate that amastigotes and promastigotes of both species were capable of infecting and surviving inside pre- and fully differentiated 3T3-Ad for up to 144 h. Importantly, L. (L.) amazonensis amastigotes resided in large communal parasitophorous vacuoles in pre-adipocytes, which appeared to be compressed between large lipid droplets in mature adipocytes. In parallel, individual L. (V.) braziliensis amastigotes were detected in single vacuoles 144 h post-infection. We conclude that 3T3-Ad may constitute an environment that supports low loads of viable parasites perhaps contributing to parasite maintenance, since amastigotes of both species recovered from these cells differentiated into replicative promastigotes. Our findings shed light on the potential of a new host cell model that can be relevant to the persistence of New World Leishmania species.


Asunto(s)
Leishmania , Leishmaniasis Cutánea , Leishmaniasis , Células 3T3-L1 , Adipocitos , Animales , Leishmaniasis/parasitología , Ratones , Ratones Endogámicos BALB C
19.
J Cell Biol ; 174(7): 997-1007, 2006 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-16982801

RESUMEN

Synaptotagmin (Syt) VII is a ubiquitously expressed member of the Syt family of Ca2+ sensors. It is present on lysosomes in several cell types, where it regulates Ca2+-dependent exocytosis. Because [Ca2+]i and exocytosis have been associated with phagocytosis, we investigated the phagocytic ability of macrophages from Syt VII-/- mice. Syt VII-/- macrophages phagocytose normally at low particle/cell ratios but show a progressive inhibition in particle uptake under high load conditions. Complementation with Syt VII rescues this phenotype, but only when functional Ca2+-binding sites are retained. Reinforcing a role for Syt VII in Ca2+-dependent phagocytosis, particle uptake in Syt VII-/- macrophages is significantly less dependent on [Ca2+]i. Syt VII is concentrated on peripheral domains of lysosomal compartments, from where it is recruited to nascent phagosomes. Syt VII recruitment is rapidly followed by the delivery of Lamp1 to phagosomes, a process that is inhibited in Syt VII-/- macrophages. Thus, Syt VII regulates the Ca2+-dependent mobilization of lysosomes as a supplemental source of membrane during phagocytosis.


Asunto(s)
Calcio/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Macrófagos/metabolismo , Fagocitosis , Fagosomas/metabolismo , Sinaptotagminas/metabolismo , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Células Cultivadas , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Sinaptotagminas/genética
20.
Elife ; 102021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34704555

RESUMEN

B-cell receptor (BCR)-mediated antigen internalization and presentation are essential for humoral memory immune responses. Antigen encountered by B-cells is often tightly associated with the surface of pathogens and/or antigen-presenting cells. Internalization of such antigens requires myosin-mediated traction forces and extracellular release of lysosomal enzymes, but the mechanism triggering lysosomal exocytosis is unknown. Here, we show that BCR-mediated recognition of antigen tethered to beads, to planar lipid-bilayers or expressed on cell surfaces causes localized plasma membrane (PM) permeabilization, a process that requires BCR signaling and non-muscle myosin II activity. B-cell permeabilization triggers PM repair responses involving lysosomal exocytosis, and B-cells permeabilized by surface-associated antigen internalize more antigen than cells that remain intact. Higher affinity antigens cause more B-cell permeabilization and lysosomal exocytosis and are more efficiently presented to T-cells. Thus, PM permeabilization by surface-associated antigen triggers a lysosome-mediated B-cell resealing response, providing the extracellular hydrolases that facilitate antigen internalization and presentation.


Asunto(s)
Presentación de Antígeno/fisiología , Antígenos de Superficie , Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Linfocitos T/inmunología , Animales , Línea Celular , Membrana Celular , Exocitosis , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Permeabilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA