Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204285

RESUMO

Pathogenic intracellular bacteria, parasites and viruses have evolved sophisticated mechanisms to manipulate mammalian host cells to serve as niches for persistence and proliferation. The intracellular lifestyles of pathogens involve the manipulation of membrane-bound organellar compartments of host cells. In this review, we described how normal structural organization and cellular functions of endosomes, endoplasmic reticulum, Golgi apparatus, mitochondria, or lipid droplets are targeted by microbial virulence mechanisms. We focus on the specific interactions of Salmonella, Legionella pneumophila, Rickettsia rickettsii, Chlamydia spp. and Mycobacterium tuberculosis representing intracellular bacterial pathogens, and of Plasmodium spp. and Toxoplasma gondii representing intracellular parasites. The replication strategies of various viruses, i.e., Influenza A virus, Poliovirus, Brome mosaic virus, Epstein-Barr Virus, Hepatitis C virus, severe acute respiratory syndrome virus (SARS), Dengue virus, Zika virus, and others are presented with focus on the specific manipulation of the organelle compartments. We compare the specific features of intracellular lifestyle and replication cycles, and highlight the communalities in mechanisms of manipulation deployed.


Assuntos
Interações Hospedeiro-Patógeno , Organelas/metabolismo , Animais , Transporte Biológico , Biomarcadores , Metabolismo Energético , Interações Hospedeiro-Parasita , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Espaço Intracelular/parasitologia , Espaço Intracelular/virologia , Organelas/microbiologia , Organelas/parasitologia , Organelas/ultraestrutura , Fagocitose
2.
Parasitol Int ; 84: 102407, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34147682

RESUMO

Plasmodium parasites cause malaria in mammalian hosts and are transmitted by Anopheles mosquitoes. Activated gametocytes in the mosquito midgut egress from erythrocytes followed by fertilization and zygote formation. Zygotes differentiate into motile invasive ookinetes, which penetrate the midgut epithelium before forming oocysts beneath the basal lamina. Ookinete development and traversal across the mosquito midgut wall are major bottlenecks in the parasite life cycle. In ookinetes, surface proteins and proteins stored in apical organelles have been shown to be involved in parasite-host interactions. A group of ookinete proteins that are predicted to have such functions are named PSOPs (putative secreted ookinete protein). PSOP1 is possibly involved in migration through the midgut wall, and here its subcellular localization was examined in ookinetes by immunoelectron microscopy. PSOP1 localizes to the micronemes of Plasmodium yoelii and Plasmodium berghei ookinetes, indicating that it is stored and possibly apically secreted during ookinete penetration through the mosquito midgut wall.


Assuntos
Malária/parasitologia , Plasmodium berghei/genética , Plasmodium yoelii/genética , Proteínas de Protozoários/genética , Animais , Interações Hospedeiro-Parasita , Camundongos , Oocistos/fisiologia , Organelas/parasitologia , Proteínas de Protozoários/metabolismo
3.
Cell Microbiol ; 23(8): e13328, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33740320

RESUMO

Annulate lamellae (AL) have been observed many times over the years on electron micrographs of rapidly dividing cells, but little is known about these unusual organelles consisting of stacked sheets of endoplasmic reticulum-derived membranes with nuclear pore complexes (NPCs). Evidence is growing for a role of AL in viral infection. AL have been observed early in the life cycles of the hepatitis C virus (HCV) and, more recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), suggesting a specific induction of mechanisms potentially useful to these pathogens. Like other positive-strand RNA viruses, these viruses induce host cells membranes rearrangements. The NPCs of AL could potentially mediate exchanges between these partially sealed compartments and the cytoplasm. AL may also be involved in regulating Ca2+ homeostasis or cell cycle control. They were recently observed in cells infected with Theileria annulata, an intracellular protozoan parasite inducing cell proliferation. Further studies are required to clarify their role in intracellular pathogen/host-cell interactions.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Organelas/microbiologia , Organelas/parasitologia , Animais , COVID-19 , Citoplasma/virologia , Retículo Endoplasmático/microbiologia , Retículo Endoplasmático/parasitologia , Retículo Endoplasmático/ultraestrutura , Retículo Endoplasmático/virologia , Humanos , Organelas/ultraestrutura , Organelas/virologia , SARS-CoV-2/fisiologia
4.
Nat Commun ; 11(1): 1411, 2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32179747

RESUMO

The disease-causing blood-stage of the Plasmodium falciparum lifecycle begins with invasion of human erythrocytes by merozoites. Many vaccine candidates with key roles in binding to the erythrocyte surface and entry are secreted from the large bulb-like rhoptry organelles at the apical tip of the merozoite. Here we identify an essential role for the conserved protein P. falciparum Cytosolically Exposed Rhoptry Leaflet Interacting protein 1 (PfCERLI1) in rhoptry function. We show that PfCERLI1 localises to the cytosolic face of the rhoptry bulb membrane and knockdown of PfCERLI1 inhibits merozoite invasion. While schizogony and merozoite organelle biogenesis appear normal, biochemical techniques and semi-quantitative super-resolution microscopy show that PfCERLI1 knockdown prevents secretion of key rhoptry antigens that coordinate merozoite invasion. PfCERLI1 is a rhoptry associated protein identified to have a direct role in function of this essential merozoite invasion organelle, which has broader implications for understanding apicomplexan invasion biology.


Assuntos
Eritrócitos/parasitologia , Malária Falciparum/parasitologia , Merozoítos/metabolismo , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , Humanos , Merozoítos/genética , Merozoítos/crescimento & desenvolvimento , Organelas/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/genética
5.
PLoS Pathog ; 15(5): e1007670, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31121005

RESUMO

To elicit effective invasion and egress from infected cells, obligate intracellular parasites of the phylum Apicomplexa rely on the timely and spatially controlled exocytosis of specialized secretory organelles termed the micronemes. The effector molecules and signaling events underpinning this process are intricate; however, recent advances within the field of Toxoplasma gondii research have facilitated a broader understanding as well as a more integrated view of this complex cascade of events and have unraveled the importance of phosphatidic acid (PA) as a lipid mediator at multiple steps in this process.


Assuntos
Cálcio/metabolismo , GMP Cíclico/metabolismo , Exocitose/fisiologia , Organelas/metabolismo , Ácidos Fosfatídicos/metabolismo , Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Interações Hospedeiro-Parasita , Humanos , Organelas/parasitologia , Transporte Proteico , Proteínas de Protozoários/metabolismo , Transdução de Sinais
6.
Cell Microbiol ; 21(7): e13028, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30941868

RESUMO

The Plasmodium subtilisin-like serine protease SUB1 is expressed in hepatic and both asexual and sexual blood parasite stages. SUB1 is required for egress of invasive forms of the parasite from both erythrocytes and hepatocytes, but its subcellular localisation, function, and potential substrates in the sexual stages are unknown. Here, we have characterised the expression profile and subcellular localisation of SUB1 in Plasmodium berghei sexual stages. We show that the protease is selectively expressed in mature male gametocytes and localises to secretory organelles known to be involved in gamete egress, called male osmiophilic bodies. We have investigated PbSUB1 function in the sexual stages by generating P. berghei transgenic lines deficient in PbSUB1 expression or enzyme activity in gametocytes. Our results demonstrate that PbSUB1 plays a role in male gamete egress. We also show for the first time that the PbSUB1 substrate PbSERA3 is expressed in gametocytes and processed by PbSUB1 upon gametocyte activation. Taken together, our results strongly suggest that PbSUB1 is not only a promising drug target for asexual stages but could also be an attractive malaria transmission-blocking target.


Assuntos
Malária/genética , Plasmodium berghei/genética , Serina Endopeptidases/genética , Subtilisinas/genética , Animais , Eritrócitos/parasitologia , Células Germinativas/parasitologia , Hepatócitos/parasitologia , Malária/parasitologia , Masculino , Organelas/parasitologia , Plasmodium berghei/patogenicidade , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade
8.
Int J Parasitol ; 48(1): 83-95, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29100811

RESUMO

Plasmodium falciparum, the most lethal malaria parasite species for humans, vastly remodels the mature erythrocyte host cell upon invasion for its own survival. Maurer's clefts (MC) are membraneous structures established by the parasite in the cytoplasm of infected cells. These organelles are deemed essential for trafficking of virulence complex proteins. The display of the major virulence protein, P. falciparum erythrocyte membrane protein 1 (PfEMP1) on the surface of the infected red blood cell and the subsequent cytoadhesion of infected cells in the microvasculature of vital organs is the key mechanism that leads to the pathology associated with malaria infection. In a previous study we established that PFE60 (PIESP2) is one of the protein components of this complex. Here we demonstrate that PFE60 plays a role in MC lamella segmentation since in the absence of the protein, infected cells display a higher number of stacked MC compared with wild type infected red blood cells. Also, another exported parasite protein (Pf332) failed to localise correctly to the MC in cells lacking PFE60. Furthermore - unlike all other described resident MC membrane proteins - PFE60 does not require its transmembrane regions to be targeted to the organelle. We also provide further evidence that PFE60 is not a red blood cell surface antigen.


Assuntos
Membranas Intracelulares/química , Membranas Intracelulares/parasitologia , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Fatores de Virulência/metabolismo , Citoplasma/química , Citoplasma/parasitologia , Interações Hospedeiro-Parasita , Humanos , Malária Falciparum/metabolismo , Organelas/química , Organelas/metabolismo , Organelas/parasitologia , Plasmodium falciparum/genética , Transporte Proteico , Proteínas de Protozoários/genética , Fatores de Virulência/genética
9.
PLoS Genet ; 13(9): e1007008, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28922357

RESUMO

The macaque parasite Plasmodium knowlesi is a significant concern in Malaysia where cases of human infection are increasing. Parasites infecting humans originate from genetically distinct subpopulations associated with the long-tailed (Macaca fascicularis (Mf)) or pig-tailed macaques (Macaca nemestrina (Mn)). We used a new high-quality reference genome to re-evaluate previously described subpopulations among human and macaque isolates from Malaysian-Borneo and Peninsular-Malaysia. Nuclear genomes were dimorphic, as expected, but new evidence of chromosomal-segment exchanges between subpopulations was found. A large segment on chromosome 8 originating from the Mn subpopulation and containing genes encoding proteins expressed in mosquito-borne parasite stages, was found in Mf genotypes. By contrast, non-recombining organelle genomes partitioned into 3 deeply branched lineages, unlinked with nuclear genomic dimorphism. Subpopulations which diverged in isolation have re-connected, possibly due to deforestation and disruption of wild macaque habitats. The resulting genomic mosaics reveal traits selected by host-vector-parasite interactions in a setting of ecological transition.


Assuntos
Interações Hospedeiro-Patógeno/genética , Malária/genética , Organelas/genética , Plasmodium knowlesi/genética , Animais , Culicidae/genética , Culicidae/parasitologia , Genoma , Humanos , Insetos Vetores/genética , Macaca fascicularis/genética , Macaca fascicularis/parasitologia , Macaca nemestrina/genética , Macaca nemestrina/parasitologia , Malária/parasitologia , Malária/transmissão , Organelas/parasitologia , Plasmodium knowlesi/patogenicidade
10.
J Eukaryot Microbiol ; 64(2): 173-182, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27463732

RESUMO

Five amoeboid organisms of different origin (isolated from fish organs, soil and digestive tract of earthworm) that shared light microscopical and ultrastructural features including type and arrangement of mitochondrial cristae were subjected to phylogenetic analyses based on sequences of SSU rDNA and protein coding genes (actin, cytochrome oxidase I, and eukaryotic elongation factor 2). The reconstruction of multigene phylogeny of the strains studied (i) revealed that they belong to the same single-genus Copromyxa clade; (ii) strongly supported position of Copromyxa cantabrigiensis (syn. Hartmannella cantabrigiensis) within the genus; (iii) together with comparisons of light and electron microscopy data justified reclassification of Cashia limacoides (syn. Vexillifera expectata) to Copromyxa limacoides n. comb., and (iv) justified description of a new species, Copromyxa laresi n. sp.


Assuntos
Amebozoários/classificação , Amebozoários/genética , Amebozoários/ultraestrutura , Lobosea/classificação , Lobosea/genética , Lobosea/ultraestrutura , Filogenia , Actinas/genética , Amoeba , Amebozoários/isolamento & purificação , Animais , Sequência de Bases , República Tcheca , DNA de Protozoário/genética , DNA Ribossômico/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Quinase do Fator 2 de Elongação/genética , Brânquias/parasitologia , Lobosea/isolamento & purificação , Microscopia Eletrônica , Mitocôndrias/parasitologia , Mitocôndrias/ultraestrutura , Oligoquetos/parasitologia , Organelas/parasitologia , Organelas/ultraestrutura , Proteínas de Protozoários/genética , Alinhamento de Sequência , Solo/parasitologia , Espanha , Especificidade da Espécie , Peixe-Zebra/parasitologia
11.
Exp Parasitol ; 167: 7-16, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27132051

RESUMO

Toxoplasmosis is a globally spread zoonosis. The pathogen Toxoplasma gondii can hijack cellular organelles of host for replication. Although a number of important cellular life events are controlled by cell organelles, very little is known of the transcriptional changes of host cellular organelles after infection with T. gondii. Herein, we performed RNA-sequencing (RNA-seq) and bioinformatics analyses to study the global organelle component changes. It was found that many transcripts of the mouse spleen cellular organelle components were altered by acute T. gondii infection with the RH strain (Type I). Most differentially expressed transcripts of mitochondrial components were downregulated, especially those involved in biosynthetic and metabolic processes. Moreover, mitochondria based apoptosis process was downregulated. In terms of cytoskeleton, most differentially expressed transcript of cytoskeleton components were also downregulated, including septin cytoskeleton, cytoskeleton organization, centrosome and myosin. For endolysosomal system, ion transporters were downregulated at mRNA level, whereas the cytolytic components were increased, such as granzymes, Rab27a and perforin1 (Prf1). The main transcripts of Golgi apparatus components involved in sialylation or vesicle-mediated transportation were downregulated, while immune related components were upregulated. For endoplasmic reticulum (ER), posttranslational modification, drug metabolism and material transportation related transcripts were downregulated. In addition, T. gondii antigen cross-presentation by MHC-I complex could be downregulated by the downregulation of CD76 and ubiquitination related transcripts. The present study, for the first time, described the transcriptional changes of the mouse spleen cellular organelles following acute T. gondii infection, which provides a foundation to study the interaction between T. gondii and host cells at the sub-cellular level.


Assuntos
Organelas/metabolismo , Baço/metabolismo , Toxoplasmose Animal/metabolismo , Animais , Apoptose , Biologia Computacional , Citoesqueleto/metabolismo , Regulação para Baixo , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Endossomos/imunologia , Endossomos/metabolismo , Metabolismo Energético , Expressão Gênica , Complexo de Golgi/metabolismo , Lisossomos/imunologia , Lisossomos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Organelas/parasitologia , Organelas/patologia , RNA de Protozoário/química , RNA de Protozoário/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Baço/parasitologia , Baço/patologia , Baço/ultraestrutura , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/patologia , Transcriptoma , Regulação para Cima
12.
Pathog Dis ; 69(2): 72-86, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23821471

RESUMO

The bacterium Chlamydia trachomatis and the protozoan parasite Toxoplasma gondii are the causative agents of chlamydiosis and toxoplasmosis in humans, respectively. Both microorganisms are obligate intracellular pathogens and notorious for extensively modifying the cytoskeletal architecture and the endomembrane system of their host cells to establish productive infections. This review highlights the similar tactics developed by these two pathogens to manipulate their host cell despite their genetic unrelatedness. Using an in vitro cell culture model whereby single fibroblasts are infected by C. trachomatis and T. gondii simultaneously, thus setting up an intracellular competition, we demonstrate that the solutions to the problem of intracellular survival deployed by the parasite and the bacterium may represent an example of convergent evolution, driven by the necessity to acquire nutrients in a hostile environment.


Assuntos
Infecções por Chlamydia/complicações , Coinfecção/microbiologia , Coinfecção/parasitologia , Organelas/microbiologia , Organelas/parasitologia , Toxoplasmose/complicações , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/fisiologia , Fibroblastos/microbiologia , Fibroblastos/parasitologia , Humanos , Modelos Teóricos , Toxoplasma/fisiologia , Toxoplasmose/parasitologia
13.
Cell Microbiol ; 15(7): 1111-26, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23279197

RESUMO

The haemoglobinopathies S and C protect carriers from severe Plasmodium falciparum malaria. We have recently shown that haemoglobin S and C interfere with host-actin remodelling in parasitized erythrocytes and the generation of an actin network that seems to be required for vesicular protein trafficking from the Maurer's clefts (a parasite-derived intermediary protein secretory organelle) to the erythrocyte surface. Here we show that the actin network exerts skeletal functions by anchoring the Maurer's clefts within the erythrocyte cytoplasm. Using a customized tracking tool to investigate the motion of single Maurer's clefts, we found that a functional actin network restrains Brownian motion of this organelle. Maurer's clefts moved significantly faster in wild-type erythrocytes treated with the actin depolymerizing agent cytochalasin D and in erythrocytes containing the haemoglobin variants S and C. Our data support the model of an impaired actin network being an underpinning cause of cellular malfunctioning in parasitized erythrocytes containing haemoglobin S or C, and, possibly, for the protective role of these haemoglobin variants against severe malaria.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/parasitologia , Hemoglobina C/metabolismo , Hemoglobina Falciforme/metabolismo , Organelas/metabolismo , Organelas/parasitologia , Plasmodium falciparum/metabolismo , Actinas/metabolismo , Interações Hospedeiro-Patógeno
14.
Mediators Inflamm ; 2012: 478601, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22619483

RESUMO

The flagellated protozoa Trypanosoma cruzi is the causal agent of Chagas' disease, a significant public health issue and still a major cause of morbidity and mortality in Latin America. Acute Chagas' disease elicits a strong inflammatory response. In order to control the parasite multiplication, cells of the monocytic lineage are highly mobilized. Monocyte differentiation leads to the formation of phagocytosing macrophages, which are strongly activated and direct host defense. A distinguishing feature of Chagas' disease-triggered macrophages is the presence of increased numbers of distinct cytoplasmic organelles termed lipid bodies or lipid droplets. These organelles are actively formed in response to the parasite and are sites for synthesis and storage of inflammatory mediators. This review covers current knowledge on lipid bodies elicited by the acute Chagas' disease within inflammatory macrophages and discusses the role of these organelles in inflammation. The increased knowledge of lipid bodies in pathogenic mechanisms of infections may not only contribute to the understanding of pathogen-host interactions but may also identify new targets for intervention.


Assuntos
Doença de Chagas/parasitologia , Interações Hospedeiro-Patógeno , Monócitos/parasitologia , Trypanosoma cruzi/fisiologia , Animais , Apoptose , Doença de Chagas/imunologia , Técnicas de Cocultura , Citoplasma/metabolismo , Modelos Animais de Doenças , Humanos , Imunidade Inata , Inflamação , Lipídeos/química , Macrófagos/imunologia , Macrófagos/parasitologia , Camundongos , Microscopia de Fluorescência/métodos , Monócitos/citologia , Organelas/parasitologia , Ratos , Trypanosoma cruzi/metabolismo
15.
PLoS Pathog ; 7(2): e1001286, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21379336

RESUMO

Apicomplexan parasites cause devastating diseases including malaria and toxoplasmosis. They harbour a plastid-like, non-photosynthetic organelle of algal origin, the apicoplast, which fulfils critical functions for parasite survival. Because of its essential and original metabolic pathways, the apicoplast has become a target for the development of new anti-apicomplexan drugs. Here we show that the lipid phosphatidylinositol 3-monophosphate (PI3P) is involved in apicoplast biogenesis in Toxoplasma gondii. In yeast and mammalian cells, PI3P is concentrated on early endosomes and regulates trafficking of endosomal compartments. Imaging of PI3P in T. gondii showed that the lipid was associated with the apicoplast and apicoplast protein-shuttling vesicles. Interference with regular PI3P function by over-expression of a PI3P specific binding module in the parasite led to the accumulation of vesicles containing apicoplast peripheral membrane proteins around the apicoplast and, ultimately, to the loss of the organelle. Accordingly, inhibition of the PI3P-synthesising kinase interfered with apicoplast biogenesis. These findings point to an unexpected implication for this ubiquitous lipid and open new perspectives on how nuclear encoded proteins traffic to the apicoplast. This study also highlights the possibility of developing specific pharmacological inhibitors of the parasite PI3-kinase as novel anti-apicomplexan drugs.


Assuntos
Organelas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Toxoplasma/crescimento & desenvolvimento , Toxoplasma/metabolismo , Toxoplasmose/metabolismo , Animais , Animais Geneticamente Modificados , Apicomplexa , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/parasitologia , Prepúcio do Pênis/citologia , Prepúcio do Pênis/metabolismo , Prepúcio do Pênis/parasitologia , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Biogênese de Organelas , Organelas/parasitologia , Fosfatidilinositol 3-Quinases/metabolismo , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia
16.
APMIS ; 117(5-6): 458-76, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19400868

RESUMO

Toxoplasma gondii is an obligate intracellular protozoan parasite that can infect most warm-blooded animals and cause severe and life-threatening disease in developing fetuses and in immune-compromised patients. Although Toxoplasma was discovered over 100 years ago, we are only now beginning to appreciate the importance of the role that parasite modulation of its host has on parasite growth, bradyzoite development, immune evasion, and virulence. The goal of this review is to highlight these findings, to develop an integrated model for communication between Toxoplasma and its host, and to discuss new questions that arise out of these studies.


Assuntos
Interações Hospedeiro-Parasita , Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Adesão Celular , Ciclo Celular , Citocinas/fisiologia , Citoesqueleto/parasitologia , Progressão da Doença , Regulação da Expressão Gênica , Genótipo , Interações Hospedeiro-Parasita/imunologia , Humanos , Camundongos , Organelas/parasitologia , Proteínas de Protozoários/fisiologia , Toxoplasma/genética , Toxoplasma/imunologia , Toxoplasma/patogenicidade , Toxoplasmose/imunologia , Toxoplasmose Animal/imunologia , Transcrição Gênica , Vacúolos/metabolismo , Vacúolos/parasitologia , Virulência
17.
Annu Rev Microbiol ; 62: 471-87, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18785844

RESUMO

Intracellular pathogens such as viruses and bacteria subvert all the major cellular functions of their hosts. Targeted host processes include protein synthesis, membrane trafficking, modulation of gene expression, antigen presentation, and apoptosis. In recent years, it has become evident that protozoan pathogens, including members of the phylum Apicomplexa, also hijack their host cell's functions to access nutrients and to escape cellular defenses and immune responses. These obligate intracellular parasites provide superb illustrations of the subversion of host cell processes such as the recruitment and reorganization of host cell compartments without fusion around the parasitophorous vacuole of Toxoplasma gondii; the export of Plasmodium falciparum proteins on the surface of infected erythrocytes; and the induced transformation of the lymphocytes infected by Theileria parva, which leads to clonal extension.


Assuntos
Apicomplexa/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Animais , Apicomplexa/imunologia , Apoptose , Cryptosporidium parvum/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Humanos , Modelos Biológicos , Organelas/parasitologia , Plasmodium falciparum/patogenicidade , Infecções por Protozoários/parasitologia , Infecções por Protozoários/patologia , Infecções por Protozoários/fisiopatologia , Theileria parva/patogenicidade , Toxoplasma/patogenicidade
18.
Mol Microbiol ; 67(4): 687-91, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18179419

RESUMO

In 1902, the German physician Georg Maurer discovered a dotted staining pattern within the cytoplasm of Plasmodium falciparum infected erythrocytes that, according to the tradition at the time, was named in his honour. The significance of Georg Maurer's discovery remained unrecognized for almost a century. Only recently are Maurer's clefts appreciated as a novel type of secretory organelle. Established by the malaria parasite within its host cell, Maurer's clefts play an essential role in directing proteins from the parasite to the erythrocyte surface. In this issue of Molecular Microbiology, Hanssen et al. report on the three dimensional structure of Maurer's clefts, as determined by electron tomography. The data presented suggest that Maurer's clefts are connected to both the parasitophorous vacuolar and the erythrocyte plasma membrane, however, no continuum exists that would allow lipids or proteins to freely flow between these three compartments. This seminal work, which stands in the tradition of Georg Maurer's original discovery, represents a milestone in our understanding of the structure and function of this fascinating organelle.


Assuntos
Eritrócitos/parasitologia , Eritrócitos/ultraestrutura , Malária Falciparum/parasitologia , Organelas/ultraestrutura , Plasmodium falciparum , Animais , Citoplasma/parasitologia , Citoplasma/ultraestrutura , Vesículas Citoplasmáticas/ultraestrutura , Interações Hospedeiro-Parasita , Membranas Intracelulares/ultraestrutura , Organelas/parasitologia , Transporte Proteico
19.
Mol Microbiol ; 67(4): 703-18, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18067543

RESUMO

During intraerythrocytic development, the human malaria parasite, Plasmodium falciparum, establishes membrane-bound compartments, known as Maurer's clefts, outside the confines of its own plasma membrane. The Maurer's compartments are thought to be a crucial component of the machinery for protein sorting and trafficking; however, their ultrastructure is only partly defined. We have used electron tomography to image Maurer's clefts of 3D7 strain parasites. The compartments are revealed as flattened structures with a translucent lumen and a more electron-dense coat. They display a complex and convoluted morphology, and some regions are modified with surface nodules, each with a circular cross-section of approximately 25 nm. Individual 25 nm vesicle-like structures are also seen in the erythrocyte cytoplasm and associated with the red blood cell membrane. The Maurer's clefts are connected to the red blood cell membrane by regions with extended stalk-like profiles. Immunogold labelling with specific antibodies confirms differential labelling of the Maurer's clefts and the parasitophorous vacuole and erythrocyte membranes. Spot fluorescence photobleaching was used to demonstrate the absence of a lipid continuum between the Maurer's clefts and parasite membranes and the host plasma membrane.


Assuntos
Eritrócitos/parasitologia , Eritrócitos/ultraestrutura , Organelas/ultraestrutura , Plasmodium falciparum/fisiologia , Animais , Antígenos de Protozoários/metabolismo , Citoplasma/ultraestrutura , Vesículas Citoplasmáticas/ultraestrutura , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/ultraestrutura , Eritrócitos/metabolismo , Interações Hospedeiro-Parasita , Humanos , Imageamento Tridimensional/métodos , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Organelas/parasitologia , Fotodegradação , Plasmodium falciparum/ultraestrutura , Tomografia/métodos , Vacúolos/parasitologia , Vacúolos/ultraestrutura
20.
Nat Rev Microbiol ; 6(1): 79-88, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18059289

RESUMO

Toxoplasma gondii is a single-celled, eukaryotic parasite that can only reproduce inside a host cell. Upon entry, this Apicomplexan parasite co-opts host functions for its own purposes. An unusual set of apical organelles, named rhoptries, contain some of the machinery that is used by T. gondii both for invasion and to commandeer host functions. Of particular interest are a group of injected protein kinases that are among the most variable of all the T. gondii proteins. At least one of these kinases has a major effect on host-gene expression, including the modulation of key regulators of the immune response. Here, we discuss these recent findings and use them to propose a model in which an expansion of host range is a major force that drives rhoptry-protein evolution.


Assuntos
Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Organelas/parasitologia , Organelas/ultraestrutura , Proteínas de Protozoários/fisiologia , Toxoplasma/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA