Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Mais filtros












Intervalo de ano de publicação
1.
Microbiol Spectr ; 10(1): e0141321, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34985339

RESUMO

Atmospheric Scanning Electron Microscopy (ASEM) is a powerful tool to observe a wet specimen at high resolution under atmospheric pressure. Here, we visualized a protozoan parasite Trypanosoma cruzi over the course of its infection cycle in the host mammalian cell. This is the first observation of intracellular parasite using a liquid-phase EM. Unlike regular SEM, aldehyde-fixed cell body of T. cruzi appears translucent, allowing the visualization of internal structures such as kinetoplast of trypomastigote and nucleus of amastigote. Plasma membrane of the host mammalian cell also appears translucent, which enabled direct observation of differentiating intracellular parasites and dynamic change of host cellular structures in their near-natural states. Various water-rich structures including micro- and macro- vesicles were visualized around T. cruzi. In addition, Correlative Light and Electron Microscopy exploiting open sample dish of ASEM allowed identification of parasite nucleus and transfected fluorescence-labeled parasites soon after internalization, while location of this morphological intermediate was otherwise obscure. Successful visualization of the differentiation of T. cruzi within the host cell demonstrated here opens up the possibility of using ASEM for observation of variety of intracellular parasites. IMPORTANCE Using Atmospheric Scanning Electron Microscopy (ASEM), we visualized interaction between infectious stage of Trypanosoma cruzi and completely intact host mammalian cell. Plasma membrane appears translucent under ASEM, which not only enables direct observation of T. cruzi within its host cell, but also reveals internal structures of the parasite itself. Sample deformation is minimal, since the specimen remains hydrated under atmospheric pressure at all times. This nature of ASEM, along with the open structure of ASEM sample dish, is suited for correlative light-electron microscopy, which can further be exploited in identification of fluorescent protein in the intracellular parasites.


Assuntos
Doença de Chagas/parasitologia , Trypanosoma cruzi/ultraestrutura , Animais , Membrana Celular/parasitologia , Membrana Celular/ultraestrutura , Humanos , Camundongos , Microscopia Eletrônica de Varredura , Trypanosoma cruzi/crescimento & desenvolvimento
2.
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
3.
mBio ; 12(3): e0098821, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34154418

RESUMO

Chagas' disease arises as a direct consequence of the lytic cycle of Trypanosoma cruzi in the mammalian host. While invasion is well studied for this pathogen, study of egress has been largely neglected. Here, we provide the first description of T. cruzi egress documenting a coordinated mechanism by which T. cruzi engineers its escape from host cells in which it has proliferated and which is essential for maintenance of infection and pathogenesis. Our results indicate that this parasite egress is a sudden event involving coordinated remodeling of host cell cytoskeleton and subsequent rupture of host cell plasma membrane. We document that host cells maintain plasma membrane integrity until immediately prior to parasite release and report the sequential transformation of the host cell's actin cytoskeleton from normal meshwork in noninfected cells to spheroidal cages-a process initiated shortly after amastigogenesis. Quantification revealed gradual reduction in F-actin over the course of infection, and using cytoskeletal preparations and electron microscopy, we were able to observe disruption of the F-actin proximal to intracellular trypomastigotes. Finally, Western blotting experiments suggest actin degradation driven by parasite proteases, suggesting that degradation of cytoskeleton is a principal component controlling the initiation of egress. Our results provide the first description of the cellular mechanism that regulates the lytic component of the T. cruzi lytic cycle. We show graphically how it is possible to preserve the envelope of host cell plasma membrane during intracellular proliferation of the parasite and how, in cells packed with amastigotes, differentiation into trypomastigotes may trigger sudden egress. IMPORTANCE Understanding how Trypanosoma cruzi interacts with host cells has been transformed by high-quality studies that have examined in detail the mechanisms of T. cruzi host cell invasion. In contrast, little is known about the latter stages of the parasite's lytic cycle: how parasites egress and thereby sustain round after round of infection. Our results show that once in the host cell cytosol and having undergone amastigogenesis, T. cruzi begins to alter the host cell cytoskeleton, remodeling normal F-actin meshworks into encapsulating spheroidal cages. Filamentous actin diminishes over the course of the lytic cycle, and just prior to egress, the filaments comprising the cages are severely degraded where adjacent to the parasites. We conclude that sudden egress follows breach of the containment afforded by the actin cytoskeleton and subsequent plasma membrane rupture-a process that when understood in molecular detail may serve as a target for future novel therapeutic interventions.


Assuntos
Citoesqueleto de Actina/fisiologia , Membrana Celular/patologia , Citoesqueleto/metabolismo , Citoesqueleto/parasitologia , Interações Hospedeiro-Parasita , Trypanosoma cruzi/fisiologia , Actinas/metabolismo , Animais , Membrana Celular/parasitologia , Doença de Chagas/parasitologia , Chlorocebus aethiops , Células Vero
4.
Nat Commun ; 12(1): 3788, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145271

RESUMO

Active host cell invasion by the obligate intracellular apicomplexan parasites relies on the formation of a moving junction, which connects parasite and host cell plasma membranes during entry. Invading Toxoplasma gondii tachyzoites secrete their rhoptry content and insert a complex of RON proteins on the cytoplasmic side of the host cell membrane providing an anchor to which the parasite tethers. Here we show that a rhoptry-resident kinase RON13 is a key virulence factor that plays a crucial role in host cell entry. Cryo-EM, kinase assays, phosphoproteomics and cellular analyses reveal that RON13 is a secretory pathway kinase of atypical structure that phosphorylates rhoptry proteins including the components of the RON complex. Ultimately, RON13 kinase activity controls host cell invasion by anchoring the moving junction at the parasite-host cell interface.


Assuntos
Membrana Celular/parasitologia , Proteínas de Protozoários/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/patologia , Transporte Biológico/fisiologia , Células Cultivadas , Interações Hospedeiro-Parasita , Humanos , Via Secretória/fisiologia , Fatores de Virulência
5.
Mol Biochem Parasitol ; 243: 111372, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33961918

RESUMO

Malaria parasites are obligate intracellular pathogens that live in human red blood cells harbored by a parasitophorous vacuole. The parasites need to exit from the red blood cell to continue life-cycle progression and ensure human-to-mosquito transmission. Two types of blood stages are able to lyse the enveloping red blood cell to mediate egress, the merozoites and the gametocytes. The intraerythrocytic parasites exit the red blood cell via an inside-out mode during which the membrane of the parasitophorous vacuole ruptures prior to the red blood cell membrane. Membrane rupture is initiated by the exocytosis of specialized secretory vesicles following the perception of egress triggers. The molecular mechanisms of red blood cell egress have particularly been studied in malaria gametocytes. Upon activation by external factors, gametocytes successively discharge at least two types of vesicles, the osmiophilic bodies needed to rupture the parasitophorous vacuole membrane and recently identified egress vesicles that are important for the perforation of the erythrocyte membrane. In recent years, important components of the signaling cascades leading to red blood cell egress have been investigated and several proteins of the osmiophilic bodies have been identified. We here report on the newest findings on the egress of gametocytes from the red blood cell. We further focus on the content and function of the egress-related vesicles and discuss the molecular machinery that might drive vesicle discharge.


Assuntos
Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Plasmodium/patogenicidade , Proteínas SNARE/metabolismo , Vesículas Secretórias/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Exocitose , Plasmodium/fisiologia , Proteínas de Protozoários/metabolismo
6.
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
7.
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
8.
Mol Immunol ; 129: 12-20, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33254075

RESUMO

Cryptocaryon irritans is an obligate parasitic ciliate protozoan that can infect various commercially important mariculture teleosts and cause high lethality and economic loss, especially Larimichthys crocea. Current methods of controlling or preventing this parasite with chemicals or antibiotics are widely considered to be environmentally harmful. The antiparasitic activity of some antimicrobial peptides (AMPs) attracted extensive attention of scholars. In the study, a novel piscidin 5-like type 4 (termed Lc-P5L4) excavated from comparative transcriptome of C. irritans - immuned L. crocea was identified and characterized. Sequence analysis shows the full-length cDNA of Lc-P5L4 is 539 bp containing an open reading frame (ORF) of 198 bp which encodes a peptide of 65 amino acid residues. The genome consists of three exons and two introns which exist in its ORF, and all the exon-intron boundaries are in accordance with classical GT-AG rule (GT/intron/AG). Multiple alignments indicate the signal peptides share highly conserved identity, while mature peptides are more diverse. Phylogenetic analysis displays Lc-P5L4 clusters together with other members of piscidin 5-like family. Next, quantitative Real-time PCR (qRT-PCR) detection found C. irritans infection could upregulate Lc-P5L4 expression level in all tested tissues significantly, it appeared earliest upregulation in the theronts infection stage in the head kidney; the expression contents reached to maximum level in the intestine, gill and muscle during trophonts falling off stage; while it was just upregulated during secondary bacterial infection stage in the liver and spleen. The data showed Lc-P5L4 upregulation time points were in accordance with different infection stages. With recombinant Lc-P5L4 (rLc-P5L4) obtained through Escherichia coli system, in vitro assay showed rLc-P5L4 could cause cilia deactivation, cell bodiesclumping and sticking to each other, then cell membrane rupture and contents leakage. The data illustrated Lc-P5L4 played critical roles in the immune defense against C. irritans infection, and provided another proof that piscidins exhibit multiple anti- C. irritans features.


Assuntos
Antiparasitários/metabolismo , Cilióforos/metabolismo , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Perciformes/genética , Perciformes/metabolismo , Aminoácidos/genética , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Infecções por Cilióforos/genética , Infecções por Cilióforos/metabolismo , Infecções por Cilióforos/parasitologia , DNA Complementar/genética , Éxons/genética , Doenças dos Peixes/genética , Doenças dos Peixes/metabolismo , Doenças dos Peixes/parasitologia , Genoma/genética , Íntrons/genética , Fígado/metabolismo , Fígado/parasitologia , Fases de Leitura Aberta/genética , Perciformes/parasitologia , Filogenia , Baço/metabolismo , Baço/parasitologia , Transcriptoma/genética , Regulação para Cima/genética
9.
Parasitol Int ; 75: 102003, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31669509

RESUMO

Plasmodium falciparum proteins involved in vascular endothelial cell adherence are transported to the surface of infected erythrocytes. These proteins are exported through parasite-derived membrane structures within the erythrocyte cytoplasm called Maurer's clefts. Skeleton binding protein 1 (SBP1) is localized in the Maurer's clefts and plays an important role in transporting molecules to the surface of infected erythrocytes. Details of the translocation pathway are unclear and in this study we focused on the subcellular localization of SBP1 at an early intraerythrocytic stage. We performed immunoelectron microscopy using specific anti-SBP1 antibodies generated by immunization with recombinant SBP1 of P. falciparum. At the early trophozoite (ring form) stage, SBP1 was detected within an electron dense material (EDM) found in the parasite cytoplasm and in the parasitophorous vacuolar (PV) space. These findings demonstrate that SBP1 accumulates in EDM in the early trophozoite cytoplasm and is transported to the PV space before translocation to the Maurer's clefts formed in the erythrocyte cytoplasm.


Assuntos
Eritrócitos/parasitologia , Proteínas de Membrana/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Vacúolos/parasitologia , Animais , Membrana Celular/parasitologia , Citoplasma/parasitologia , Proteínas de Membrana/metabolismo , Microscopia Imunoeletrônica , Plasmodium falciparum/metabolismo , Transporte Proteico , Proteínas de Protozoários/metabolismo , Coelhos
10.
Curr Opin Microbiol ; 52: 116-123, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31349210

RESUMO

Intracellular protozoans co-evolved with their mammalian host cells a range of strategies to cope with the composite and dynamic cell surface features they encounter during migration and infection. Therefore, these single-celled eukaryotic parasites represent a fascinating source of living probes for precisely capturing the dynamic coupling between the membrane and contractile cortex components of the cell surface. Such biomechanical changes drive a constant re-sculpting of the host cell surface, enabling rapid adjustments that contribute to cellular homeostasis. As emphasized in this review, through the design of specific molecular devices and stratagems to interfere with the biomechanics of the mammalian cell surface these parasitic microbes escape from dangerous or unfavourable microenvironments by breaching host cell membranes, directing the membrane repair machinery to wounded membrane areas, or minimizing membrane assault using discretion and speed when invading host cells for sustained residence.


Assuntos
Apicomplexa/patogenicidade , Membrana Celular/patologia , Citoplasma/parasitologia , Interações Hospedeiro-Parasita , Kinetoplastida/patogenicidade , Animais , Apicomplexa/genética , Membrana Celular/parasitologia , Humanos , Kinetoplastida/genética , Leishmania/genética , Leishmania/patogenicidade , Plasmodium/genética , Plasmodium/patogenicidade , Infecções por Protozoários , Toxoplasma/genética , Toxoplasma/patogenicidade , Trypanosoma/genética , Trypanosoma/patogenicidade
11.
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
12.
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
13.
Sci Rep ; 9(1): 5924, 2019 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-30976120

RESUMO

The propagation of Toxoplasma gondii is accomplished by repeated lytic cycles of parasite attachment to a host cell, invasion, replication within a parasitophorous vacuole, and egress from the cell. This lytic cycle is delicately regulated by calcium-dependent reversible phosphorylation of the molecular machinery that drives invasion and egress. While much progress has been made elucidating the protein kinases and substrates central to parasite propagation, little is known about the relevant protein phosphatases. In this study, we focused on the five protein phosphatases that are predicted to be membrane-associated either integrally or peripherally. We have determined that of these only PPM5C, a PP2C family member, localizes to the plasma membrane of Toxoplasma. Disruption of PPM5C results in a slow propagation phenotype in tissue culture. Interestingly, parasites lacking PPM5C divide and undergo egress at a normal rate, but have a deficiency in attaching to host cells. Both membrane localization and phosphatase activity are required for PPM5C's role in attachment. Phosphoproteomic analysis show relatively few phosphorylation sites being affected by PPM5C deletion in extracellular parasites of which several are found on proteins involved in signaling cascades. This implies that PPM5C is part of a wider regulatory network important for attachment to host cells.


Assuntos
Membrana Celular/metabolismo , Junções Célula-Matriz/metabolismo , Interações Hospedeiro-Parasita , Fosfoproteínas Fosfatases/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/metabolismo , Sinalização do Cálcio , Membrana Celular/parasitologia , Junções Célula-Matriz/parasitologia , Fibroblastos/metabolismo , Fibroblastos/parasitologia , Humanos , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteínas de Protozoários/genética , Toxoplasmose/parasitologia
14.
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
15.
Structure ; 26(8): 1059-1071.e6, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-29910186

RESUMO

Plasmodium falciparum and Toxoplasma gondii are obligate intracellular parasites that belong to the phylum of Apicomplexa and cause major human diseases. Their access to an intracellular lifestyle is reliant on the coordinated release of proteins from the specialized apical organelles called micronemes and rhoptries. A specific phosphatidic acid effector, the acylated pleckstrin homology domain-containing protein (APH) plays a central role in microneme exocytosis and thus is essential for motility, cell entry, and egress. TgAPH is acylated on the surface of the micronemes and recruited to phosphatidic acid (PA)-enriched membranes. Here, we dissect the atomic details of APH PA-sensing hub and its functional interaction with phospholipid membranes. We unravel the key determinant of PA recognition for the first time and show that APH inserts into and clusters multiple phosphate head-groups at the bilayer binding surface.


Assuntos
Fibroblastos/parasitologia , Ácidos Fosfatídicos/metabolismo , Plasmodium falciparum/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas de Protozoários/química , Toxoplasma/metabolismo , Acilação , Sequência de Aminoácidos , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Exocitose , Fibroblastos/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Interações Hospedeiro-Parasita , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Simulação de Dinâmica Molecular , Organelas/metabolismo , Organelas/ultraestrutura , Ácidos Fosfatídicos/química , Plasmodium falciparum/genética , Plasmodium falciparum/ultraestrutura , Domínios de Homologia à Plecstrina , Cultura Primária de Células , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Toxoplasma/genética , Toxoplasma/ultraestrutura
16.
Artigo em Inglês | MEDLINE | ID: mdl-29846473

RESUMO

Leptospirosis is an acute bacterial septicemic febrile disease caused by pathogenic leptospires, which affect humans and animals in all parts of the world. Transmission can occur by direct contact with infected animals or, more commonly, through indirect contact with water or soil contaminated with urine from infected animals. Leptospires enter the body by penetrating mucous membranes or skin abrasions and disseminate through the hematogenic route. In humans, leptospirosis may cause a wide spectrum of symptoms. Most cases have a biphasic clinical presentation, which begins with the septicemic phase followed by immune manifestations. The severe forms of the disease may be life threatening with multisystem damage including renal failure, hepatic dysfunction, vascular damage, pulmonary hemorrhage and muscle lesions. In this review, we present and discuss the pathogenesis of the human disease and the mechanisms of cell membrane injuries, which occur mainly due to the presence of leptospires and/or their antigen/s in the host tissues.


Assuntos
Caderinas/metabolismo , Membrana Celular/parasitologia , Rim/parasitologia , Leptospirose/etiologia , Leptospirose/patologia , Fígado/parasitologia , Doenças Musculares/parasitologia , Animais , Membrana Celular/patologia , Humanos , Rim/patologia , Leptospirose/metabolismo , Fígado/patologia , Doenças Musculares/patologia
17.
Microbes Infect ; 20(1): 57-62, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28951315

RESUMO

Previous studies have analysed the process of Toxoplasma gondii egress with the aid of inducers, such as calcium ionophores. Although calcium transients have been successful in triggering T. gondii egress, the structural panorama of "natural" and artificial events should match. The present study approaches the natural egress of this parasite using super-resolution and electron microscopy and reveals lytic and non-lytic events of individual egress; this corroborates the use of calcium ionophore as a reliable tool to trigger parasite egress. Altogether, our data suggest that different signalling routes can converge to similar structural aspects in natural and induced egress.


Assuntos
Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Linhagem Celular , Membrana Celular/parasitologia , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Células Epiteliais/parasitologia , Interações Hospedeiro-Parasita , Macaca mulatta , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia de Vídeo , Vacúolos/parasitologia , Vacúolos/fisiologia , Vacúolos/ultraestrutura
18.
Sci Rep ; 7(1): 17907, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29263399

RESUMO

Apicomplexa parasites, including Toxoplasma and Plasmodium species, possess a unique invasion mechanism that involves a tight apposition between the parasite and the host plasma membranes, called "moving junction" (MJ). The MJ is formed by the assembly of the microneme protein AMA1, exposed at the surface of the parasite, and the parasite rhoptry neck (RON) protein RON2, exposed at the surface of the host cell. In the host cell, RON2 is associated with three additional parasite RON proteins, RON4, RON5 and RON8. Here we describe RON4L1, an additional member of the MJ complex in Toxoplasma. RON4L1 displays some sequence similarity with RON4 and is cleaved at the C-terminal end before reaching the rhoptry neck. Upon secretion during invasion, RON4L1 is associated with MJ and targeted to the cytosolic face of the host membrane. We generated a RON4 L1 knock-out cell line and showed that it is not essential for the lytic cycle in vitro, although mutant parasites kill mice less efficiently. Similarly to RON8, RON4L1 is a coccidian-specific protein and its traffic to the MJ is not affected in absence of RON2, RON4 and RON5, suggesting the co-existence of independent MJ complexes in tachyzoite of Toxoplasma.


Assuntos
Membrana Celular/parasitologia , Interações Hospedeiro-Parasita , Junções Intercelulares/parasitologia , Proteínas de Protozoários/metabolismo , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia , Animais , Antígenos de Protozoários/metabolismo , Membrana Celular/metabolismo , Sobrevivência Celular , Células Cultivadas , Feminino , Junções Intercelulares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Toxoplasmose/metabolismo
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...