Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
PLoS Pathog ; 17(11): e1010030, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34807955

RESUMO

The parasite Entamoeba histolytica is the etiological agent of amoebiasis, a major cause of morbidity and mortality due to parasitic diseases in developing countries. Phagocytosis is an essential mode of obtaining nutrition and has been associated with the virulence behaviour of E. histolytica. Signalling pathways involved in activation of cytoskeletal dynamics required for phagocytosis remains to be elucidated in this parasite. Our group has been studying initiation of phagocytosis and formation of phagosomes in E. histolytica and have described some of the molecules that play key roles in the process. Here we showed the involvement of non-Dbl Rho Guanine Nucleotide Exchange Factor, EhGEF in regulation of amoebic phagocytosis by regulating activation of EhRho1. EhGEF was found in the phagocytic cups during the progression of cups, until closure of phagosomes, but not in the phagosomes themselves. Our observation from imaging, pull down experiments and down regulating expression of different molecules suggest that EhGEF interacts with EhRho1 and it is required during initiation of phagocytosis and phagosome formation. Also, biophysical, and computational analysis reveals that EhGEF mediates GTP exchange on EhRho1 via an unconventional pathway. In conclusion, we describe a non-Dbl EhGEF of EhRho1 which is involved in endocytic processes of E. histolytica.


Assuntos
Entamoeba histolytica/fisiologia , Entamebíase/parasitologia , Fagocitose , Proteínas de Protozoários/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Membrana Celular/parasitologia , Entamebíase/genética , Entamebíase/metabolismo , Eritrócitos/parasitologia , Fagossomos , Proteínas de Protozoários/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Proteínas rho de Ligação ao GTP/genética
2.
Cell Mol Life Sci ; 78(10): 4545-4561, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33713154

RESUMO

Malaria is a vector-borne parasitic disease with a vast impact on human history, and according to the World Health Organisation, Plasmodium parasites still infect over 200 million people per year. Plasmodium falciparum, the deadliest parasite species, has a remarkable ability to undermine the host immune system and cause life-threatening disease during blood infection. The parasite's host cells, red blood cells (RBCs), generally maintain an asymmetric distribution of phospholipids in the two leaflets of the plasma membrane bilayer. Alterations to this asymmetry, particularly the exposure of phosphatidylserine (PS) in the outer leaflet, can be recognised by phagocytes. Because of the importance of innate immune defence numerous studies have investigated PS exposure in RBCs infected with P. falciparum, but have reached different conclusions. Here we review recent advancements in our understanding of the molecular mechanisms which regulate asymmetry in RBCs, and whether infection with the P. falciparum parasite results in changes to PS exposure. On the balance of evidence, it is likely that membrane asymmetry is disrupted in parasitised RBCs, though some methodological issues need addressing. We discuss the potential causes and consequences of altered asymmetry in parasitised RBCs, particularly for in vivo interactions with the immune system, and the role of host-parasite co-evolution. We also examine the potential asymmetric state of parasite membranes and summarise current knowledge on the parasite proteins, which could regulate asymmetry in these membranes. Finally, we highlight unresolved questions at this time and the need for interdisciplinary approaches to uncover the machinery which enables P. falciparum parasites to hide in mature erythrocytes.


Assuntos
Membrana Celular/metabolismo , Membrana Celular/parasitologia , Eritrócitos/metabolismo , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Fosfolipídeos/metabolismo , Plasmodium falciparum/patogenicidade , Animais , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/parasitologia
3.
J Cell Sci ; 132(6)2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30814331

RESUMO

Intracellular parasites of the genus Leishmania are the causative agents of leishmaniasis. The disease is transmitted by the bite of a sand fly vector, which inoculates the parasite into the skin of mammalian hosts, including humans. During chronic infection the parasite lives and replicates inside phagocytic cells, notably the macrophages. An interesting, but overlooked finding, is that other cell types and even non-phagocytic cells have been found to be infected by Leishmania spp. Nevertheless, the mechanisms by which Leishmania invades such cells had not been previously studied. Here, we show that L. amazonensis can induce their own entry into fibroblasts independently of actin cytoskeleton activity, and, thus, through a mechanism that is distinct from phagocytosis. Invasion involves subversion of host cell functions, such as Ca2+ signaling and recruitment and exocytosis of host cell lysosomes involved in plasma membrane repair.This article has an associated First Person interview with the first author of the paper.


Assuntos
Membrana Celular/parasitologia , Fibroblastos/parasitologia , Leishmania mexicana , Lisossomos/parasitologia , Citoesqueleto de Actina/parasitologia , Animais , Sinalização do Cálcio , Linhagem Celular , Membrana Celular/metabolismo , Exocitose , Interações Hospedeiro-Parasita , Leishmania mexicana/metabolismo , Leishmania mexicana/parasitologia , Macrófagos/parasitologia , Camundongos , Fagocitose
4.
J Mol Recognit ; 34(6): e2886, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33393093

RESUMO

Entamoeba histolytica (Eh), a parasitic protozoan and the causative agent of invasive Amoebiasis, invade the host tissue through an effective secretory pathway. There are several lines of evidence suggesting that amoebic trophozoite pore-forming complex amoebapore and a large class of proteases enzymes including rhomboid proteases, cysteine proteases, and metalloproteases are implicated in host tissue invasion. For successful delivery of these molecules/cargos, trophozoites heavily rely on sorting machinery from the endoplasmic reticulum, Golgi to plasma membrane. Although, sole secretion machinery in E. histolytica is not characterized yet. Therefore, here our aim is to understand the properties of key molecules N-ethylmaleimide-sensitive fusion protein attached to protein receptors (SNAREs) in E. histolytica. SNAREs proteins are an important component of the membrane-trafficking machinery and have been associated in a range of processes including vesicle tethering, fusion as well as specificity of vesicular transport in all eukaryotic cells. SNARE proteins are architecturally simple, categorized by the presence of one copy of a homologous coiled-coil forming motif. However, the structural information and protein-protein interaction study of Eh-associated syntaxin proteins are still not known. Here, we characterize the syntaxin 1 like molecule and VAMP from Eh through physiochemical profiling, modeling, atomistic simulation, protein-protein interaction, and docking approaches on the proteins containing SNARE and synaptobrevin domain. The modeled structures and the critical residues recognized through protein interaction and docking study may provide better structural and functional insights into these proteins and may aid in the development of newer diagnostic assays.


Assuntos
Entamoeba histolytica/metabolismo , Mapas de Interação de Proteínas/fisiologia , Proteínas Qa-SNARE/metabolismo , Sequência de Aminoácidos , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Células Eucarióticas/metabolismo , Células Eucarióticas/parasitologia , Canais Iônicos/metabolismo , Simulação de Acoplamento Molecular , Estudos Prospectivos , Proteínas de Protozoários/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo
5.
Plant Cell ; 29(6): 1460-1479, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28559475

RESUMO

A transient rise in cytosolic calcium ion concentration is one of the main signals used by plants in perception of their environment. The role of calcium in the detection of abiotic stress is well documented; however, its role during biotic interactions remains unclear. Here, we use a fluorescent calcium biosensor (GCaMP3) in combination with the green peach aphid (Myzus persicae) as a tool to study Arabidopsis thaliana calcium dynamics in vivo and in real time during a live biotic interaction. We demonstrate rapid and highly localized plant calcium elevations around the feeding sites of M. persicae, and by monitoring aphid feeding behavior electrophysiologically, we demonstrate that these elevations correlate with aphid probing of epidermal and mesophyll cells. Furthermore, we dissect the molecular mechanisms involved, showing that interplay between the plant defense coreceptor BRASSINOSTEROID INSENSITIVE-ASSOCIATED KINASE1 (BAK1), the plasma membrane ion channels GLUTAMATE RECEPTOR-LIKE 3.3 and 3.6 (GLR3.3 and GLR3.6), and the vacuolar ion channel TWO-PORE CHANNEL1 (TPC1) mediate these calcium elevations. Consequently, we identify a link between plant perception of biotic threats by BAK1, cellular calcium entry mediated by GLRs, and intracellular calcium release by TPC1 during a biologically relevant interaction.


Assuntos
Afídeos/patogenicidade , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Arabidopsis/parasitologia , Cálcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Citosol/metabolismo , Canais Iônicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Vacúolos/metabolismo , Animais , Proteínas de Arabidopsis/genética , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Regulação da Expressão Gênica de Plantas , Proteínas Serina-Treonina Quinases/genética , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo
6.
Cell Microbiol ; 21(11): e13065, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31155842

RESUMO

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.


Assuntos
Membrana Celular/parasitologia , Lisossomos/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Trypanosoma cruzi/patogenicidade , Animais , Secreções Corporais/efeitos da radiação , Cálcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/patologia , Ceramidas/metabolismo , Exocitose , Humanos , Lisossomos/metabolismo , Doença de Niemann-Pick Tipo A/enzimologia , Doença de Niemann-Pick Tipo B/enzimologia , Transporte Proteico , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética , Esfingomielinas/metabolismo , Trypanosoma cruzi/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(16): 4225-4230, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28373555

RESUMO

Invasion of the red blood cell (RBC) by the Plasmodium parasite defines the start of malaria disease pathogenesis. To date, experimental investigations into invasion have focused predominantly on the role of parasite adhesins or signaling pathways and the identity of binding receptors on the red cell surface. A potential role for signaling pathways within the erythrocyte, which might alter red cell biophysical properties to facilitate invasion, has largely been ignored. The parasite erythrocyte-binding antigen 175 (EBA175), a protein required for entry in most parasite strains, plays a key role by binding to glycophorin A (GPA) on the red cell surface, although the function of this binding interaction is unknown. Here, using real-time deformability cytometry and flicker spectroscopy to define biophysical properties of the erythrocyte, we show that EBA175 binding to GPA leads to an increase in the cytoskeletal tension of the red cell and a reduction in the bending modulus of the cell's membrane. We isolate the changes in the cytoskeleton and membrane and show that reduction in the bending modulus is directly correlated with parasite invasion efficiency. These data strongly imply that the malaria parasite primes the erythrocyte surface through its binding antigens, altering the biophysical nature of the target cell and thus reducing a critical energy barrier to invasion. This finding would constitute a major change in our concept of malaria parasite invasion, suggesting it is, in fact, a balance between parasite and host cell physical forces working together to facilitate entry.


Assuntos
Antígenos de Protozoários/metabolismo , Membrana Celular/patologia , Eritrócitos/patologia , Glicoforinas/metabolismo , Malária Falciparum/patologia , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Antígenos de Protozoários/genética , Biofísica , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Citoesqueleto , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Glicoforinas/genética , Interações Hospedeiro-Parasita , Humanos , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/isolamento & purificação , Ligação Proteica , Proteínas de Protozoários/genética , Transdução de Sinais
8.
PLoS Pathog ; 13(7): e1006453, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28683142

RESUMO

Egress of the malaria parasite Plasmodium falciparum from its host red blood cell is a rapid, highly regulated event that is essential for maintenance and completion of the parasite life cycle. Egress is protease-dependent and is temporally associated with extensive proteolytic modification of parasite proteins, including a family of papain-like proteins called SERA that are expressed in the parasite parasitophorous vacuole. Previous work has shown that the most abundant SERA, SERA5, plays an important but non-enzymatic role in asexual blood stages. SERA5 is extensively proteolytically processed by a parasite serine protease called SUB1 as well as an unidentified cysteine protease just prior to egress. However, neither the function of SERA5 nor the role of its processing is known. Here we show that conditional disruption of the SERA5 gene, or of both the SERA5 and related SERA4 genes simultaneously, results in a dramatic egress and replication defect characterised by premature host cell rupture and the failure of daughter merozoites to efficiently disseminate, instead being transiently retained within residual bounding membranes. SERA5 is not required for poration (permeabilization) or vesiculation of the host cell membrane at egress, but the premature rupture phenotype requires the activity of a parasite or host cell cysteine protease. Complementation of SERA5 null parasites by ectopic expression of wild-type SERA5 reversed the egress defect, whereas expression of a SERA5 mutant refractory to processing failed to rescue the phenotype. Our findings implicate SERA5 as an important regulator of the kinetics and efficiency of egress and suggest that proteolytic modification is required for SERA5 function. In addition, our study reveals that efficient egress requires tight control of the timing of membrane rupture.


Assuntos
Antígenos de Protozoários/metabolismo , Eritrócitos/parasitologia , Malária Falciparum/parasitologia , Peptídeo Hidrolases/metabolismo , Plasmodium falciparum/fisiologia , Animais , Antígenos de Protozoários/genética , Membrana Celular/parasitologia , Eritrócitos/química , Humanos , Cinética , Merozoítos/química , Merozoítos/genética , Merozoítos/crescimento & desenvolvimento , Merozoítos/fisiologia , Peptídeo Hidrolases/genética , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Proteólise
9.
Molecules ; 24(10)2019 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-31137574

RESUMO

Malaria is an infectious disease caused by Plasmodium group. The mechanisms of antimalarial drugs DHA/CQ are still unclear today. The inhibitory effects (IC50) of single treatments with DHA/CQ or V-ATPase inhibitor Baf-A1 or combination treatments by DHA/CQ combined with Baf-A1 on the growth of Plasmodium falciparum strain 3D7 was investigated. Intracellular cytoplasmic pH and labile iron pool (LIP) were labeled by pH probe BCECF, AM and iron probe calcein, AM, the fluorescence of the probes was measured by FCM. The effects of low doses of DHA (0.2 nM, 0.4 nM, 0.8 nM) on gene expression of V-ATPases (vapE, vapA, vapG) located in the membrane of DV were tested by RT-qPCR. DHA combined with Baf-A1 showed a synergism effect (CI = 0.524) on the parasite growth in the concentration of IC50. Intracellular pH and irons were effected significantly by different doses of DHA/Baf-A1. Intracellular pH was decreased by CQ combined with Baf-A1 in the concentration of IC50. Intracellular LIP was increased by DHA combined with Baf-A1 in the concentration of 20 IC50. The expression of gene vapA was down-regulated by all low doses of DHA (0.2/0.4/0.8 nM) significantly (p < 0.001) and the expression of vapG/vapE were up-regulated by 0.8 nM DHA significantly (p < 0.001). Interacting with ferrous irons, affecting the DV membrane proton pumping and acidic pH or cytoplasmic irons homeostasis may be the antimalarial mechanism of DHA while CQ showed an effect on cytoplasmic pH of parasite in vitro. Lastly, this article provides us preliminary results and a new idea for antimalarial drugs combination and new potential antimalarial combination therapies.


Assuntos
Artemisininas/farmacologia , Cloroquina/farmacologia , Eritrócitos/parasitologia , Homeostase , Estágios do Ciclo de Vida/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Animais , Antimaláricos/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/parasitologia , Quimioterapia Combinada , Eritrócitos/efeitos dos fármacos , Fluorescência , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Concentração Inibidora 50 , Ferro/metabolismo , Macrolídeos/farmacologia , Parasitos/efeitos dos fármacos , Parasitos/crescimento & desenvolvimento , Testes de Sensibilidade Parasitária , Plasmodium falciparum/efeitos dos fármacos , Trofozoítos/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo
10.
PLoS Pathog ; 11(11): e1005250, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26562790

RESUMO

The apicomplexan, Cryptosporidium parvum, possesses a bacterial-type lactate dehydrogenase (CpLDH). This is considered to be an essential enzyme, as this parasite lacks the Krebs cycle and cytochrome-based respiration, and mainly-if not solely, relies on glycolysis to produce ATP. Here, we provide evidence that in extracellular parasites (e.g., sporozoites and merozoites), CpLDH is localized in the cytosol. However, it becomes associated with the parasitophorous vacuole membrane (PVM) during the intracellular developmental stages, suggesting involvement of the PVM in parasite energy metabolism. We characterized the biochemical features of CpLDH and observed that, at lower micromolar levels, the LDH inhibitors gossypol and FX11 could inhibit both CpLDH activity (Ki = 14.8 µM and 55.6 µM, respectively), as well as parasite growth in vitro (IC50 = 11.8 µM and 39.5 µM, respectively). These observations not only reveal a new function for the poorly understood PVM structure in hosting the intracellular development of C. parvum, but also suggest LDH as a potential target for developing therapeutics against this opportunistic pathogen, for which fully effective treatments are not yet available.


Assuntos
Criptosporidiose/tratamento farmacológico , Cryptosporidium/enzimologia , L-Lactato Desidrogenase/metabolismo , Vacúolos/parasitologia , Sequência de Aminoácidos , Animais , Membrana Celular/parasitologia
11.
Exp Parasitol ; 174: 31-41, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28011167

RESUMO

Trypanosoma brucei are extracellular hemoflagellate protozoan parasites and one of the causative agents of a devastating zoonotic disease called African Trypanosomiasis. In humans, the disease is caused by Trypanosoma brucei rhodensiense and Trypanosoma brucei gambiense, which cross the blood brain barrier (BBB) causing neurological disorders which culminate in death if untreated. In some domestic animals and laboratory rodents, Trypanosoma brucei brucei causes a disease similar to that in humans. The mechanism by which Trypanosoma brucei brucei invade biological barriers including the BBB has not been fully elucidated. To further address this issue, Mardin Dardy Canine Kidney II (MDCKII) and Human dermal microvascular endothelial cell (HDMEC) monolayers were grown to confluence on transwell inserts to constitute in vitro biological barriers. MDCKII cells were chosen for their ability to form tight junctions similar to those formed by the BBB endothelial cells. Labeled trypanosomes were placed in the upper chamber of transwell inserts layered with confluent MDCKII/HDMEC monolayers and their ability to cross the monolayer over time evaluated. Our results show that only 0.5-1.25% of Trypanosoma brucei brucei were able to migrate across the monolayers after 3 h. By employing immune-staining and confocal microscopic analysis we observed that trypanosomes were located at the tight junctions and inside the cell in the MDCK II monolayers indicating that they crossed the monolayer using both the paracellular and transcellular routes. Our observations also showed that there seemed to be no obvious degradation of junction proteins Zonula Ocludens-1, Occludin and Ecadherin. In the HDMEC cell monolayer, our scanning electron microscopy data showed that Trypanosoma brucei brucei is able to modulate the plasma membrane to form invaginations similar to cuplike structures formed by Tlymphocytes. However these structures seemed to be independent of vascular adhesion molecules suggesting that they could be more like the membrane ruffles formed by certain intracellular bacteria during invasion. Taken together, our data reveal a mechanism by which Trypanosoma brucei brucei is able to cross different biological barriers including the BBB without causing any obvious damage.


Assuntos
Barreira Hematoencefálica/parasitologia , Células Madin Darby de Rim Canino/parasitologia , Trypanosoma brucei brucei/fisiologia , Tripanossomíase Africana/parasitologia , Animais , Barreira Hematoencefálica/ultraestrutura , Linhagem Celular , Membrana Celular/parasitologia , Membrana Celular/ultraestrutura , Cães , Flagelos/fisiologia , Flagelos/ultraestrutura , Imunofluorescência , Humanos , Células Madin Darby de Rim Canino/ultraestrutura , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Coelhos , Ratos , Proteínas de Junções Íntimas/química , Junções Íntimas/parasitologia , Trypanosoma brucei brucei/ultraestrutura , Tripanossomíase Africana/patologia
12.
PLoS Pathog ; 10(3): e1004025, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24651769

RESUMO

Apicomplexans facilitate host cell invasion through formation of a tight-junction interface between parasite and host plasma membranes called the moving junction (MJ). A complex of the rhoptry neck proteins RONs 2/4/5/8 localize to the MJ during invasion where they are believed to provide a stable anchoring point for host penetration. During the initiation of invasion, the preformed MJ RON complex is injected into the host cell where RON2 spans the host plasma membrane while RONs 4/5/8 localize to its cytosolic face. While much attention has been directed toward an AMA1-RON2 interaction supposed to occur outside the cell, little is known about the functions of the MJ RONs positioned inside the host cell. Here we provide a detailed analysis of RON5 to resolve outstanding questions about MJ complex organization, assembly and function during invasion. Using a conditional knockdown approach, we show loss of RON5 results in complete degradation of RON2 and mistargeting of RON4 within the parasite secretory pathway, demonstrating that RON5 plays a key role in organization of the MJ RON complex. While RON8 is unaffected by knockdown of RON5, these parasites are unable to invade new host cells, providing the first genetic demonstration that RON5 plays a critical role in host cell penetration. Although invasion is not required for injection of rhoptry effectors into the host cytosol, parasites lacking RON5 also fail to form evacuoles suggesting an intact MJ complex is a prerequisite for secretion of rhoptry bulb contents. Additionally, while the MJ has been suggested to function in egress, disruption of the MJ complex by RON5 depletion does not impact this process. Finally, functional complementation of our conditional RON5 mutant reveals that while proteolytic separation of RON5 N- and C-terminal fragments is dispensable, a portion of the C-terminal domain is critical for RON2 stability and function in invasion.


Assuntos
Membrana Celular/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Proteínas de Protozoários/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Toxoplasmose/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/microbiologia , Técnicas de Silenciamento de Genes , Humanos , Toxoplasma/metabolismo
13.
FASEB J ; 28(7): 3103-13, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24706359

RESUMO

The genomes of malaria parasites (Plasmodium spp.) contain a family of genes encoding proteins with a Plasmodium helical interspersed subtelomeric (PHIST) domain, most of which are predicted to be exported into the parasite-infected human red blood cell (iRBC). Here, using transgenic parasites and a combination of cellular, biochemical, and biophysical assays, we have characterized and determined the function of a novel member of the PHIST protein family in Plasmodium falciparum, termed lysine-rich membrane-associated PHISTb (LyMP). LyMP was shown to associate directly with the cytoskeleton of iRBCs where it plays a role in their abnormal ability to adhere to a protein expressed on vascular endothelial cells, resulting in sequestration. Deletion of LyMP dramatically reduced adhesion of iRBCs to CD36 by 55%, which was completely restored to wild-type levels on complementation. Intriguingly, in the absence of LyMP, formation of RBC membrane knobs and the level of surface exposure of the parasites' major cytoadhesive ligand, PfEMP1, were identical to those for the parental parasite line, demonstrating for the first time an additional mechanism that enhances cytoadherence of iRBCs beyond those already recognized. Our findings identify LyMP as a previously unknown RBC cytoskeletal-binding protein that is likely to be of major significance in the complex pathophysiology of falciparum malaria.-Proellocks, N. I., Herrmann, S., Buckingham, D. W., Hanssen, E., Hodges, E. K., Elsworth, B., Morahan, B. J., Coppel, R. L., Cooke, B. M. A lysine-rich membrane-associated PHISTb protein involved in alteration of the cytoadhesive properties of Plasmodium falciparum infected red blood cells.


Assuntos
Adesão Celular/fisiologia , Citoesqueleto/metabolismo , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Lisina/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Citoesqueleto/parasitologia , Endotélio Vascular/metabolismo , Endotélio Vascular/parasitologia , Humanos , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Proteínas de Membrana/metabolismo , Ligação Proteica/fisiologia
14.
Cell Microbiol ; 16(10): 1549-64, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24824158

RESUMO

The last step of Leishmania intracellular life cycle is the egress of amastigotes from the host cell and their uptake by adjacent cells. Using multidimensional live imaging of long-term-infected macrophage cultures we observed that Leishmania amazonensis amastigotes were transferred from cell to cell when the donor host macrophage delivers warning signs of imminent apoptosis. They were extruded from the macrophage within zeiotic structures (membrane blebs, an apoptotic feature) rich in phagolysosomal membrane components. The extrusions containing amastigotes were selectively internalized by vicinal macrophages and the rescued amastigotes remain viable in recipient macrophages. Host cell apoptosis induced by micro-irradiation of infected macrophage nuclei promoted amastigotes extrusion, which were rescued by non-irradiated vicinal macrophages. Using amastigotes isolated from LAMP1/LAMP2 knockout fibroblasts, we observed that the presence of these lysosomal components on amastigotes increases interleukin 10 production. Enclosed within host cell membranes, amastigotes can be transferred from cell to cell without full exposure to the extracellular milieu, what represents an important strategy developed by the parasite to evade host immune system.


Assuntos
Leishmania/patogenicidade , Leishmaniose/transmissão , Proteínas de Membrana Lisossomal/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Macrófagos/parasitologia , Animais , Apoptose , Linhagem Celular , Membrana Celular/parasitologia , Fibroblastos , Interleucina-10/biossíntese , Leishmaniose/patologia , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteínas de Membrana Lisossomal/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
15.
Eukaryot Cell ; 13(8): 965-76, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24951442

RESUMO

Intracellular pathogens can replicate efficiently only after they manipulate and modify their host cells to create an environment conducive to replication. While diverse cellular pathways are targeted by different pathogens, metabolism, membrane and cytoskeletal architecture formation, and cell death are the three primary cellular processes that are modified by infections. Toxoplasma gondii is an obligate intracellular protozoan that infects ∼30% of the world's population and causes severe and life-threatening disease in developing fetuses, in immune-comprised patients, and in certain otherwise healthy individuals who are primarily found in South America. The high prevalence of Toxoplasma in humans is in large part a result of its ability to modulate these three host cell processes. Here, we highlight recent work defining the mechanisms by which Toxoplasma interacts with these processes. In addition, we hypothesize why some processes are modified not only in the infected host cell but also in neighboring uninfected cells.


Assuntos
Toxoplasma/fisiologia , Toxoplasmose/imunologia , Animais , Apoptose , Metabolismo dos Carboidratos , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Núcleo Celular/parasitologia , Núcleo Celular/fisiologia , Metabolismo Energético , Interações Hospedeiro-Parasita , Humanos , Inflamassomos/fisiologia , Metabolismo dos Lipídeos , Toxoplasmose/parasitologia
16.
Proc Natl Acad Sci U S A ; 109(19): 7463-8, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22523242

RESUMO

Apical membrane antigen 1 (AMA1) is a conserved transmembrane adhesin of apicomplexan parasites that plays an important role in host-cell invasion. Toxoplasma gondii AMA1 (TgAMA1) is secreted onto the parasite surface and subsequently released by proteolytic cleavage within its transmembrane domain. To elucidate the function of TgAMA1 intramembrane proteolysis, we used a heterologous cleavage assay to characterize the determinants within the TgAMA1 transmembrane domain (ALIAGLAVGGVLLLALLGGGCYFA) that govern its processing. Quantitative analysis revealed that the TgAMA1(L/G) mutation enhanced cleavage by 13-fold compared with wild type. In contrast, the TgAMA1(AG/FF) mutation reduced cleavage by 30-fold, whereas the TgAMA1(GG/FF) mutation had a minor effect on proteolysis; mutating both motifs in a quadruple mutant blocked cleavage completely. We then complemented a TgAMA1 conditional knockout parasite line with plasmids expressing these TgAMA1 variants. Contrary to expectation, variants that increased or decreased TgAMA1 processing by >10-fold had no phenotypic consequences, revealing that the levels of rhomboid proteolysis in parasites are not delicately balanced. Only parasites transgenically expressing or carrying a true knock-in allele of the uncleavable TgAMA1(AG/FF+GG/FF) mutant showed a growth defect, which resulted from inhibiting invasion without perturbing intracellular replication. These data demonstrate that TgAMA1 cleavage plays a role in invasion, but refute a recently proposed model in which parasite replication within the host cell is regulated by intramembrane proteolysis of TgAMA1.


Assuntos
Antígenos de Protozoários/metabolismo , Membrana Celular/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/genética , Western Blotting , Células COS , Divisão Celular , Membrana Celular/parasitologia , Células Cultivadas , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interações Hospedeiro-Parasita , Humanos , Masculino , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Proteólise , Proteínas de Protozoários/genética , Homologia de Sequência de Aminoácidos , Toxoplasma/genética , Toxoplasma/fisiologia
17.
Biophys J ; 107(1): 43-54, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24988340

RESUMO

The blood stage malaria parasite, the merozoite, has a small window of opportunity during which it must successfully target and invade a human erythrocyte. The process of invasion is nonetheless remarkably rapid. To date, mechanistic models of invasion have focused predominantly on the parasite actomyosin motor contribution to the energetics of entry. Here, we have conducted a numerical analysis using dimensions for an archetypal merozoite to predict the respective contributions of the host-parasite interactions to invasion, in particular the role of membrane wrapping. Our theoretical modeling demonstrates that erythrocyte membrane wrapping alone, as a function of merozoite adhesive and shape properties, is sufficient to entirely account for the first key step of the invasion process, that of merozoite reorientation to its apex and tight adhesive linkage between the two cells. Next, parasite-induced reorganization of the erythrocyte cytoskeleton and release of parasite-derived membrane can also account for a considerable energetic portion of actual invasion itself, through membrane wrapping. Thus, contrary to the prevailing dogma, wrapping by the erythrocyte combined with parasite-derived membrane release can markedly reduce the expected contributions of the merozoite actomyosin motor to invasion. We therefore propose that invasion is a balance between parasite and host cell contributions, evolved toward maximal efficient use of biophysical forces between the two cells.


Assuntos
Membrana Celular/parasitologia , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Plasmodium falciparum/patogenicidade , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Merozoítos/fisiologia
18.
J Biol Chem ; 288(12): 8712-8725, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23376275

RESUMO

The recently discovered role of a perforin-like protein (PLP1) for rapid host cell egress by the protozoan parasite Toxoplasma gondii expanded the functional diversity of pore-forming proteins. Whereas PLP1 was found to be necessary for rapid egress and pathogenesis, the sufficiency for and mechanism of membrane attack were yet unknown. Here we further dissected the PLP1 knock-out phenotype, the mechanism of PLP1 pore formation, and the role of each domain by genetic complementation. We found that PLP1 is sufficient for membrane disruption and has a conserved mechanism of pore formation through target membrane binding and oligomerization to form large, multimeric membrane-embedded complexes. The highly conserved, central MACPF domain and the ß-sheet-rich C-terminal domain were required for activity. Loss of the unique N-terminal extension reduced lytic activity and led to a delay in rapid egress, but did not significantly decrease virulence, suggesting that small amounts of lytic activity are sufficient for pathogenesis. We found that both N- and C-terminal domains have membrane binding activity, with the C-terminal domain being critical for function. This dual mode of membrane association may promote PLP1 activity and parasite egress in the diverse cell types in which this parasite replicates.


Assuntos
Hemólise , Perforina/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/fisiologia , Animais , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Permeabilidade da Membrana Celular , Células Cultivadas , Eritrócitos/parasitologia , Feminino , Técnicas de Inativação de Genes , Interações Hospedeiro-Parasita , Humanos , Camundongos , Perforina/química , Perforina/genética , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Deleção de Sequência , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasmose/parasitologia
19.
Biochem J ; 452(3): 457-66, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23544851

RESUMO

Red blood cell invasion by the malaria parasite Plasmodium falciparum relies on a complex protein network that uses low and high affinity receptor-ligand interactions. Signal transduction through the action of specific kinases is a control mechanism for the orchestration of this process. In the present study we report on the phosphorylation of the CPD (cytoplasmic domain) of P. falciparum Rh2b (reticulocyte homologue protein 2b). First, we identified Ser3233 as the sole phospho-acceptor site in the CPD for in vitro phosphorylation by parasite extract. We provide several lines of evidence that this phosphorylation is mediated by PfCK2 (P. falciparum casein kinase 2): phosphorylation is cAMP independent, utilizes ATP as well as GTP as phosphate donors, is inhibited by heparin and tetrabromocinnamic acid, and is mediated by purified PfCK2. We raised a phospho-specific antibody and showed that Ser3233 phosphorylation occurs in the parasite prior to host cell egress. We analysed the spatiotemporal aspects of this phosphorylation using immunoprecipitated endogenous Rh2b and minigenes expressing the CPD either at the plasma or rhoptry membrane. Phosphorylation of Rh2b is not spatially restricted to either the plasma or rhoptry membrane and most probably occurs before Rh2b is translocated from the rhoptry neck to the plasma membrane.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/parasitologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Células Cultivadas , Eritrócitos/química , Humanos , Ligantes , Mutação/genética , Fosforilação/genética , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/sangue , Proteínas de Protozoários/genética
20.
Nanomedicine ; 10(2): 483-90, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24096030

RESUMO

Leishmaniasis is one of the most serious diseases in the world and can be lethal if untreated. This is especially the case for visceral leishmaniasis, which is commonly caused by Leishmania (L.) infantum and for which available medication is still inadequate. A recently described antimicrobial peptide DRS 01 has been reported to kill L. infantum promastigotes, but nothing is known about its mode of action or effect on the cell. In this paper we report the visualization of the interaction between DRS 01 and L. infantum promastigotes using two high resolution microscopic techniques: atomic force microscopy and scanning electron microscopy. The results show considerable morphological changes at and above the IC50 in the treated cells. Both membrane damage and flagella alterations were observed. The results strongly suggest a membrane-directed action for DRS 01 on the Leishmania species studied. FROM THE CLINICAL EDITOR: In this paper, the effects of DRS 01, an antimicrobial peptide, is studied in Leishmania infantum using atomic force microscopy as well as standard scanning electron microscopy techniques, with the conclusion of a membrane-based effect by DRS 01 on the parasites.


Assuntos
Proteínas de Anfíbios/química , Peptídeos Catiônicos Antimicrobianos/química , Antiprotozoários/uso terapêutico , Leishmania infantum/efeitos dos fármacos , Leishmaniose Visceral/tratamento farmacológico , Proteínas de Anfíbios/isolamento & purificação , Animais , Peptídeos Catiônicos Antimicrobianos/isolamento & purificação , Membrana Celular/parasitologia , Flagelos , Humanos , Concentração Inibidora 50 , Leishmaniose Visceral/parasitologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa