Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 121(6): 1095-1111, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38574236

RESUMO

The protozoan parasite Plasmodium, the causative agent of malaria, undergoes an obligatory stage of intra-hepatic development before initiating a blood-stage infection. Productive invasion of hepatocytes involves the formation of a parasitophorous vacuole (PV) generated by the invagination of the host cell plasma membrane. Surrounded by the PV membrane (PVM), the parasite undergoes extensive replication. During intracellular development in the hepatocyte, the parasites provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterized by a long-lasting association of the autophagy marker protein, and ATG8 family member, LC3B with the PVM. LC3B localization at the PVM does not follow the canonical autophagy pathway since upstream events specific to canonical autophagy are dispensable. Here, we describe that LC3B localization at the PVM of Plasmodium parasites requires the V-ATPase and its interaction with ATG16L1. The WD40 domain of ATG16L1 is crucial for its recruitment to the PVM. Thus, we provide new mechanistic insight into the previously described PAAR response targeting Plasmodium liver stage parasites.


Assuntos
Proteínas Relacionadas à Autofagia , Autofagia , Hepatócitos , Fígado , Proteínas Associadas aos Microtúbulos , Plasmodium berghei , ATPases Vacuolares Próton-Translocadoras , Vacúolos , Vacúolos/metabolismo , Vacúolos/parasitologia , Plasmodium berghei/genética , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/metabolismo , Plasmodium berghei/enzimologia , Animais , Proteínas Relacionadas à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fígado/parasitologia , Camundongos , Hepatócitos/parasitologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Malária/parasitologia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Humanos
2.
PLoS Pathog ; 19(3): e1011210, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36996035

RESUMO

Plasmodium parasites have a complex life cycle alternating between a mosquito and a vertebrate host. Following the bite of an Anopheles female mosquito, Plasmodium sporozoites are transmitted from the skin to the liver; their first place of replication within the host. Successfully invaded sporozoites undergo a massive replication and growth involving asynchronous DNA replication and division that results in the generation of tens of thousands or even hundreds of thousands of merozoites depending on the Plasmodium species. The generation of a high number of daughter parasites requires biogenesis and segregation of organelles to finally reach a relatively synchronous cytokinesis event. At the end of liver stage (LS) development, merozoites are packed into merosomes and released into the bloodstream. They are then liberated and infect red blood cells to again produce merozoites by schizogony for the erythrocytic stage of the life cycle. Although parasite LS and asexual blood stage (ABS) differ in many respects, important similarities exist between the two. This review focuses on the cell division of Plasmodium parasite LS in comparison with other life cycle stages especially the parasite blood stage.


Assuntos
Fígado , Plasmodium , Animais , Citocinese , Estágios do Ciclo de Vida , Fígado/parasitologia , Merozoítos , Plasmodium/fisiologia , Pele , Esporozoítos
3.
NPJ Vaccines ; 7(1): 139, 2022 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-36333336

RESUMO

Whole-sporozoite (WSp) malaria vaccines induce protective immune responses in animal malaria models and in humans. A recent clinical trial with a WSp vaccine comprising genetically attenuated parasites (GAP) which arrest growth early in the liver (PfSPZ-GA1), showed that GAPs can be safely administered to humans and immunogenicity is comparable to radiation-attenuated PfSPZ Vaccine. GAPs that arrest late in the liver stage (LA-GAP) have potential for increased potency as shown in rodent malaria models. Here we describe the generation of four putative P. falciparum LA-GAPs, generated by CRISPR/Cas9-mediated gene deletion. One out of four gene-deletion mutants produced sporozoites in sufficient numbers for further preclinical evaluation. This mutant, PfΔmei2, lacking the mei2-like RNA gene, showed late liver growth arrest in human liver-chimeric mice with human erythrocytes, absence of unwanted genetic alterations and sensitivity to antimalarial drugs. These features of PfΔmei2 make it a promising vaccine candidate, supporting further clinical evaluation. PfΔmei2 (GA2) has passed regulatory approval for safety and efficacy testing in humans based on the findings reported in this study.

4.
J Cell Sci ; 134(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34013963

RESUMO

The intracellular lifestyle represents a challenge for the rapidly proliferating liver stage Plasmodium parasite. In order to scavenge host resources, Plasmodium has evolved the ability to target and manipulate host cell organelles. Using dynamic fluorescence-based imaging, we here show an interplay between the pre-erythrocytic stages of Plasmodium berghei and the host cell Golgi during liver stage development. Liver stage schizonts fragment the host cell Golgi into miniaturized stacks, which increases surface interactions with the parasitophorous vacuolar membrane of the parasite. Expression of specific dominant-negative Arf1 and Rab GTPases, which interfere with the host cell Golgi-linked vesicular machinery, results in developmental delay and diminished survival of liver stage parasites. Moreover, functional Rab11a is critical for the ability of the parasites to induce Golgi fragmentation. Altogether, we demonstrate that the structural integrity of the host cell Golgi and Golgi-associated vesicular traffic is important for optimal pre-erythrocytic development of P. berghei. The parasite hijacks the Golgi structure of the hepatocyte to optimize its own intracellular development. This article has an associated First Person interview with the first author of the paper.


Assuntos
Malária , Parasitos , Animais , Hepatócitos , Fígado , Plasmodium berghei , Proteínas de Protozoários
5.
Sci Adv ; 7(13)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33762334

RESUMO

Circulating levels of the adipokine leptin are linked to neuropathology in experimental cerebral malaria (ECM), but its source and regulation mechanism remain unknown. Here, we show that sequestration of infected red blood cells (iRBCs) in white adipose tissue (WAT) microvasculature increased local vascular permeability and leptin production. Mice infected with parasite strains that fail to sequester in WAT displayed reduced leptin production and protection from ECM. WAT sequestration and leptin induction were lost in CD36KO mice; however, ECM susceptibility revealed sexual dimorphism. Adipocyte leptin was regulated by the mechanistic target of rapamycin complex 1 (mTORC1) and blocked by rapamycin. In humans, although Plasmodium falciparum infection did not increase circulating leptin levels, iRBC sequestration, tissue leptin production, and mTORC1 activity were positively correlated with CM in pediatric postmortem WAT. These data identify WAT sequestration as a trigger for leptin production with potential implications for pathogenesis of malaria infection, prognosis, and treatment.


Assuntos
Malária Cerebral , Parasitos , Tecido Adiposo/patologia , Animais , Criança , Humanos , Leptina , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos
6.
Cell Microbiol ; 23(1): e13271, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32979009

RESUMO

The protozoan parasite Plasmodium, causative agent of malaria, invades hepatocytes by invaginating the host cell plasma membrane and forming a parasitophorous vacuole membrane (PVM). Surrounded by this PVM, the parasite undergoes extensive replication. Parasites inside a PVM provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterised by a long-lasting association of the autophagy marker protein LC3 with the PVM, which is not preceded by phosphatidylinositol 3-phosphate (PI3P)-labelling. Prior to productive invasion, sporozoites transmigrate several cells and here we describe that a proportion of traversing sporozoites become trapped in a transient traversal vacuole, provoking a host cell response that clearly differs from the PAAR response. These trapped sporozoites provoke PI3P-labelling of the surrounding vacuolar membrane immediately after cell entry, followed by transient LC3-labelling and elimination of the parasite by lysosomal acidification. Our data suggest that this PI3P response is not only restricted to sporozoites trapped during transmigration but also affects invaded parasites residing in a compromised vacuole. Thus, host cells can employ a pathway distinct from the previously described PAAR response to efficiently recognise and eliminate Plasmodium parasites.


Assuntos
Autofagia , Hepatócitos/parasitologia , Fosfatos de Fosfatidilinositol/metabolismo , Plasmodium berghei/metabolismo , Plasmodium berghei/parasitologia , Esporozoítos/metabolismo , Vacúolos/parasitologia , Animais , Linhagem Celular , Feminino , Células HeLa , Interações Hospedeiro-Parasita , Humanos , Malária/parasitologia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Organismos Geneticamente Modificados
7.
mSphere ; 5(6)2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33361125

RESUMO

Toxoplasma gondii and members of the genus Plasmodium are obligate intracellular parasites that leave their infected host cell upon a tightly controlled process of egress. Intracellular replication of the parasites occurs within a parasitophorous vacuole, and its membrane as well as the host plasma membrane need to be disrupted during egress, leading to host cell lysis. While several parasite-derived factors governing egress have been identified, much less is known about host cell factors involved in this process. Previously, RNA interference (RNAi)-based knockdown and antibody-mediated depletion identified a host signaling cascade dependent on guanine nucleotide-binding protein subunit alpha q (GNAQ) to be required for the egress of Toxoplasma tachyzoites and Plasmodium blood stage merozoites. Here, we used CRISPR/Cas9 technology to generate HeLa cells deficient in GNAQ and tested their capacity to support the egress of T. gondii tachyzoites and Plasmodium berghei liver stage parasites. While we were able to confirm the importance of GNAQ for the egress of T. gondii, we found that the egress of P. berghei liver stages was unaffected in the absence of GNAQ. These results may reflect differences between the lytic egress process in apicomplexans and the formation of host cell-derived vesicles termed merosomes by P. berghei liver stages.IMPORTANCE The coordinated release of apicomplexan parasites from infected host cells prior to reinvasion is a critical process for parasite survival and the spread of infection. While Toxoplasma tachyzoites and Plasmodium blood stages induce a fast disruption of their surrounding membranes during their egress from host cells, Plasmodium liver stages keep the host cell membrane intact and leave their host cell in host cell-derived vesicles called merosomes. The knockout of GNAQ, a protein involved in G-protein-coupled receptor signaling, demonstrates the importance of this host factor for the lytic egress of T. gondii tachyzoites. Contrastingly, the egress of P. berghei is independent of GNAQ at the liver stage, indicating the existence of a mechanistically distinct strategy to exit the host cell.


Assuntos
Membrana Celular/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Hepatócitos/parasitologia , Plasmodium berghei/fisiologia , Animais , Sistemas CRISPR-Cas , Células HeLa , Humanos , Malária/parasitologia , Plasmodium berghei/genética , Toxoplasma/metabolismo
8.
Trends Parasitol ; 36(2): 85-87, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31883707

RESUMO

A recent report by Jennison et al. reveals an important role for plasmepsin V (PMV), an aspartyl protease, in the development of malaria transmission stages. The authors showed that PMV activity is critical for protein export in these stages and that specific PMV inhibitors block parasite transmission to mosquitoes.


Assuntos
Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Malária/parasitologia , Malária/transmissão , Plasmodium/enzimologia , Animais , Antimaláricos/farmacologia , Carbamatos/farmacologia , Humanos , Estágios do Ciclo de Vida/efeitos dos fármacos , Estágios do Ciclo de Vida/fisiologia , Malária/prevenção & controle , Oligopeptídeos/farmacologia , Plasmodium/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos
9.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31730853

RESUMO

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Assuntos
Genoma de Protozoário , Estágios do Ciclo de Vida/genética , Fígado/metabolismo , Fígado/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/genética , Alelos , Amino Açúcares/biossíntese , Animais , Culicidae/parasitologia , Eritrócitos/parasitologia , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/metabolismo , Técnicas de Inativação de Genes , Genótipo , Modelos Biológicos , Mutação/genética , Parasitos/genética , Parasitos/crescimento & desenvolvimento , Fenótipo , Plasmodium berghei/metabolismo , Ploidias , Reprodução
10.
Curr Opin Microbiol ; 46: 93-101, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30317152

RESUMO

The use of rodents as model organisms to study human disease is based on the genetic and physiological similarities between the species. Successful molecular methods to generate transgenic reporter or humanized rodents has rendered rodents as powerful tools for understanding biological processes and host-pathogen interactions relevant to humans. In malaria research, rodent models have been pivotal for the study of liver stages, syndromes arising from blood stages of infection, and malaria transmission to and from the mammalian host. Importantly, many in vivo findings are comparable to pathology observed in humans only when adequate combinations of rodent strains and Plasmodium parasites are used.


Assuntos
Modelos Animais de Doenças , Malária/parasitologia , Roedores , Animais , Humanos , Plasmodium/genética , Plasmodium/fisiologia , Roedores/parasitologia
11.
Sci Adv ; 4(5): eaat3775, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29806032

RESUMO

Transmission of Plasmodium parasites to the mosquito requires the formation and development of gametocytes. Studies in infected humans have shown that only the most mature forms of Plasmodium falciparum gametocytes are present in circulation, whereas immature forms accumulate in the hematopoietic environment of the bone marrow. We used the rodent model Plasmodium berghei to study gametocyte behavior through time under physiological conditions. Intravital microscopy demonstrated preferential homing of early gametocyte forms across the intact vascular barrier of the bone marrow and the spleen early during infection and subsequent development in the extravascular environment. During the acute phase of infection, we observed vascular leakage resulting in further parasite accumulation in this environment. Mature gametocytes showed high deformability and were found entering and exiting the intact vascular barrier. We suggest that extravascular gametocyte localization and mobility are essential for gametocytogenesis and transmission of Plasmodium to the mosquito.


Assuntos
Medula Óssea/parasitologia , Malária/patologia , Malária/parasitologia , Plasmodium/fisiologia , Migração Transendotelial e Transepitelial , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Parasita , Humanos , Camundongos , Imagem Molecular , Sistema Fagocitário Mononuclear/parasitologia
12.
mSphere ; 3(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29404413

RESUMO

During their development within the vertebrate host, Plasmodium parasites infect hepatocytes and red blood cells. Within these cells, parasites are surrounded by a parasitophorous vacuole membrane (PVM). The PVM plays an essential role for the interaction of parasites with their host cells; however, only a limited number of proteins of this membrane have been identified so far. This is partially because systematic proteomic analysis of the protein content of the PVM has been difficult in the past, due to difficulties encountered in attempts to separate the PVM from other membranes such as the parasite plasma membrane. In this study, we adapted the BioID technique to in vitro-cultivated Plasmodium berghei blood stage parasites and utilized the promiscuous biotin ligase BirA* fused to PVM-resident exported protein 1 to biotinylate proteins of the PVM. These we further processed by affinity purification, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and label-free quantitation, leading to a list of 61 known and candidate PVM proteins. Seven proteins were analyzed further during blood and liver stage development. This resulted in the identification of three novel PVM proteins, which were the serine/threonine protein phosphatase UIS2 (PlasmoDB accession no. PBANKA_1328000) and two conserved Plasmodium proteins with unknown functions (PBANKA_0519300 and PBANKA_0509000). In conclusion, our report expands the number of known PVM proteins and experimentally validates BioID as a powerful method to screen for novel constituents of specific cellular compartments in P. berghei. IMPORTANCE Intracellular pathogens are often surrounded by a host-cell derived membrane. This membrane is modified by the pathogens to their own needs and is crucial for their intracellular lifestyle. In Plasmodium parasites, this membrane is referred to as the PVM and only a limited number of its proteins are known so far. Here, we applied in rodent P. berghei parasites a method called BioID, which is based on biotinylation of proximal and interacting proteins by the promiscuous biotin ligase BirA*, and demonstrated its usefulness in identification of novel PVM proteins.

13.
Sci Rep ; 7(1): 9740, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28851956

RESUMO

During asexual replication within the Anopheles mosquito and their vertebrate host, Plasmodium parasites depend on the generation of a massive amount of new plasma membrane to produce thousands of daughter parasites. How the parasite plasma membrane (PPM) is formed has mostly been studied by electron microscopy, which does not allow an insight into the dynamics of this process. We generated a Plasmodium berghei reporter parasite line by GFP-tagging of a non-essential PPM-localized protein, and followed plasma membrane development in living parasites through the entire Plasmodium life cycle. By generating double-fluorescent parasites in which the PPM is visualized in combination with the parasite endoplasmic reticulum, we show that membrane contact sites are formed between both membrane systems during oocyst and liver stage development that might be used to deliver lipids to the dramatically expanding PPM. In conclusion, we have established a powerful tool to follow PPM development in living parasites, which promises to greatly expand our knowledge of membrane biology in the Plasmodium parasite.


Assuntos
Membrana Celular/metabolismo , Microscopia Intravital/métodos , Plasmodium berghei/fisiologia , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Microscopia de Fluorescência , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Coloração e Rotulagem/métodos
14.
Cell Microbiol ; 19(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28573684

RESUMO

Eukaryotic cells can employ autophagy to defend themselves against invading pathogens. Upon infection by Plasmodium berghei sporozoites, the host hepatocyte targets the invader by labelling the parasitophorous vacuole membrane (PVM) with the autophagy marker protein LC3. Until now, it has not been clear whether LC3 recruitment to the PVM is mediated by fusion of autophagosomes or by direct incorporation. To distinguish between these possibilities, we knocked out genes that are essential for autophagosome formation and for direct LC3 incorporation into membranes. The CRISPR/Cas9 system was employed to generate host cell lines deficient for either FIP200, a member of the initiation complex for autophagosome formation, or ATG5, responsible for LC3 lipidation and incorporation of LC3 into membranes. Infection of these knockout cell lines with P. berghei sporozoites revealed that LC3 recruitment to the PVM indeed depends on functional ATG5 and the elongation machinery, but not on FIP200 and the initiation complex, suggesting a direct incorporation of LC3 into the PVM. Importantly, in P. berghei-infected ATG5-/- host cells, lysosomes still accumulated at the PVM, indicating that the recruitment of lysosomes follows an LC3-independent pathway.


Assuntos
Fígado/fisiopatologia , Plasmodium berghei/metabolismo , Plasmodium berghei/patogenicidade , Vacúolos/metabolismo , Autofagossomos/metabolismo , Autofagia/fisiologia , Sistemas CRISPR-Cas/fisiologia , Lisossomos/metabolismo , Transdução de Sinais/fisiologia , Esporozoítos/metabolismo
15.
Sci Rep ; 7(1): 2191, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28526861

RESUMO

The hepatic stage of the malaria parasite Plasmodium is accompanied by an autophagy-mediated host response directly targeting the parasitophorous vacuolar membrane (PVM) harbouring the parasite. Removal of the PVM-associated autophagic proteins such as ubiquitin, p62, and LC3 correlates with parasite survival. Yet, it is unclear how Plasmodium avoids the deleterious effects of selective autophagy. Here we show that parasites trap host autophagic factors in the tubovesicular network (TVN), an expansion of the PVM into the host cytoplasm. In proliferating parasites, PVM-associated LC3 becomes immediately redirected into the TVN, where it accumulates distally from the parasite's replicative centre. Finally, the host factors are shed as vesicles into the host cytoplasm. This strategy may enable the parasite to balance the benefits of the enhanced host catabolic activity with the risk of being eliminated by the cell's cytosolic immune defence.


Assuntos
Autofagia , Interações Hospedeiro-Parasita , Malária/metabolismo , Malária/parasitologia , Plasmodium berghei/fisiologia , Vacúolos/metabolismo , Animais , Linhagem Celular , Citoplasma/metabolismo , Genes Reporter , Humanos , Fígado/metabolismo , Fígado/parasitologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Transporte Proteico , Imagem com Lapso de Tempo
16.
mBio ; 8(2)2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28400525

RESUMO

A crucial step in the life cycle of Plasmodium parasites is the transition from the liver stage to the blood stage. Hepatocyte-derived merozoites reach the blood vessels of the liver inside host cell-derived vesicles called merosomes. The molecular basis of merosome formation is only partially understood. Here we show that Plasmodium berghei liver stage merozoites, upon rupture of the parasitophorous vacuole membrane, destabilize the host cell membrane (HCM) and induce separation of the host cell actin cytoskeleton from the HCM. At the same time, the phospholipid and protein composition of the HCM appears to be substantially altered. This includes the loss of a phosphatidylinositol 4,5-bisphosphate (PIP2) reporter and the PIP2-dependent actin-plasma membrane linker ezrin from the HCM. Furthermore, transmembrane domain-containing proteins and palmitoylated and myristoylated proteins, as well as glycosylphosphatidylinositol-anchored proteins, lose their HCM localization. Collectively, these findings provide an explanation of HCM destabilization during Plasmodium liver stage egress and thereby contribute to our understanding of the molecular mechanisms that lead to merosome formation.IMPORTANCE Egress from host cells is an essential process for intracellular pathogens, allowing successful infection of other cells and thereby spreading the infection. Here we describe the molecular details of a novel egress strategy of Plasmodium parasites infecting hepatocytes. We show that toward the end of the liver stage, parasites induce a breakdown of the host cell actin cytoskeleton, leading to destabilization of the host cell plasma membrane. This, in turn, results in the formation of membrane vesicles (merosomes), in which parasites can safely migrate from liver tissue to the bloodstream to infect red blood cells and start the pathogenic phase of malaria.


Assuntos
Membrana Celular/química , Hepatócitos/parasitologia , Proteínas de Membrana/análise , Fosfolipídeos/análise , Plasmodium berghei/fisiologia , Animais , Células Cultivadas , Camundongos Endogâmicos BALB C
17.
Nat Rev Microbiol ; 15(1): 37-54, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27890922

RESUMO

Over the past decade, major advances in imaging techniques have enhanced our understanding of Plasmodium spp. parasites and their interplay with mammalian hosts and mosquito vectors. Cryoelectron tomography, cryo-X-ray tomography and super-resolution microscopy have shifted paradigms of sporozoite and gametocyte structure, the process of erythrocyte invasion by merozoites, and the architecture of Maurer's clefts. Intravital time-lapse imaging has been revolutionary for our understanding of pre-erythrocytic stages of rodent Plasmodium parasites. Furthermore, high-speed imaging has revealed the link between sporozoite structure and motility, and improvements in time-lapse microscopy have enabled imaging of the entire Plasmodium falciparum erythrocytic cycle and the complete Plasmodium berghei pre-erythrocytic stages for the first time. In this Review, we discuss the contribution of key imaging tools to these and other discoveries in the malaria field over the past 10 years.


Assuntos
Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Merozoítos/fisiologia , Plasmodium berghei/fisiologia , Plasmodium falciparum/fisiologia , Esporozoítos/fisiologia , Animais , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Humanos , Malária/parasitologia , Malária/patologia , Merozoítos/ultraestrutura , Plasmodium berghei/ultraestrutura , Plasmodium falciparum/ultraestrutura , Esporozoítos/ultraestrutura , Imagem com Lapso de Tempo
18.
Mol Microbiol ; 102(5): 775-791, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27566438

RESUMO

To fuel the tremendously fast replication of Plasmodium liver stage parasites, the endoplasmic reticulum (ER) must play a critical role as a major site of protein and lipid biosynthesis. In this study, we analysed the parasite's ER morphology and function. Previous studies exploring the parasite ER have mainly focused on the blood stage. Visualizing the Plasmodium berghei ER during liver stage development, we found that the ER forms an interconnected network throughout the parasite with perinuclear and peripheral localizations. Surprisingly, we observed that the ER additionally generates huge accumulations. Using stimulated emission depletion microscopy and serial block-face scanning electron microscopy, we defined ER accumulations as intricate dense networks of ER tubules. We provide evidence that these accumulations are functional subdivisions of the parasite ER, presumably generated in response to elevated demands of the parasite, potentially consistent with ER stress. Compared to higher eukaryotes, Plasmodium parasites have a fundamentally reduced unfolded protein response machinery for reacting to ER stress. Accordingly, parasite development is greatly impaired when ER stress is applied. As parasites appear to be more sensitive to ER stress than are host cells, induction of ER stress could potentially be used for interference with parasite development.


Assuntos
Retículo Endoplasmático/ultraestrutura , Plasmodium berghei/ultraestrutura , Animais , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Fígado/parasitologia , Malária/parasitologia , Microscopia/métodos , Microscopia Eletrônica de Varredura , Plasmodium berghei/metabolismo , Proteínas de Protozoários/metabolismo , Resposta a Proteínas não Dobradas
19.
Malar J ; 15: 232, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27102897

RESUMO

BACKGROUND: Bioluminescence imaging is widely used for cell-based assays and animal imaging studies, both in biomedical research and drug development. Its main advantages include its high-throughput applicability, affordability, high sensitivity, operational simplicity, and quantitative outputs. In malaria research, bioluminescence has been used for drug discovery in vivo and in vitro, exploring host-pathogen interactions, and studying multiple aspects of Plasmodium biology. While the number of fluorescent proteins available for imaging has undergone a great expansion over the last two decades, enabling simultaneous visualization of multiple molecular and cellular events, expansion of available luciferases has lagged behind. The most widely used bioluminescent probe in malaria research is the Photinus pyralis firefly luciferase, followed by the more recently introduced Click-beetle and Renilla luciferases. Ultra-sensitive imaging of Plasmodium at low parasite densities has not been previously achieved. With the purpose of overcoming these challenges, a Plasmodium berghei line expressing the novel ultra-bright luciferase enzyme NanoLuc, called PbNLuc has been generated, and is presented in this work. RESULTS: NanoLuc shows at least 150 times brighter signal than firefly luciferase in vitro, allowing single parasite detection in mosquito, liver, and sexual and asexual blood stages. As a proof-of-concept, the PbNLuc parasites were used to image parasite development in the mosquito, liver and blood stages of infection, and to specifically explore parasite liver stage egress, and pre-patency period in vivo. CONCLUSIONS: PbNLuc is a suitable parasite line for sensitive imaging of the entire Plasmodium life cycle. Its sensitivity makes it a promising line to be used as a reference for drug candidate testing, as well as the characterization of mutant parasites to explore the function of parasite proteins, host-parasite interactions, and the better understanding of Plasmodium biology. Since the substrate requirements of NanoLuc are different from those of firefly luciferase, dual bioluminescence imaging for the simultaneous characterization of two lines, or two separate biological processes, is possible, as demonstrated in this work.


Assuntos
Medições Luminescentes/métodos , Malária/parasitologia , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Animais , Culicidae/parasitologia , Interações Hospedeiro-Parasita , Humanos , Fígado/parasitologia , Luciferases/genética , Organismos Geneticamente Modificados/genética , Organismos Geneticamente Modificados/metabolismo , Plasmodium berghei/isolamento & purificação
20.
J Arthroplasty ; 31(1): 156-61, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26260783

RESUMO

Screws, pegs and hydroxyapatite-coating are used to enhance the primary stability of uncemented cups. We present a 14-year follow-up of 48 hips randomized to four groups: press-fit only, press-fit plus screws, press-fit plus pegs and hydroxyapatite-coated cups. Radiostereometric migration measurements showed equally good stability regardless cup augmentation. The mean wear rate was high, 0.21 mm/year, with no differences between the groups. Seven hips had radiographical osteolysis but only in hips with augmented cups. Cups without screw-holes compared with cups with screw-holes resulted in better clinical outcome at the 14-year follow-up. Thus, augmentation of uncemented cups with screws, pegs, or hydroxyapatite did not appear to improve the long-term stability compared with press-fit only.


Assuntos
Artroplastia de Quadril/estatística & dados numéricos , Prótese de Quadril/estatística & dados numéricos , Adulto , Idoso , Artroplastia de Quadril/instrumentação , Parafusos Ósseos/estatística & dados numéricos , Durapatita , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Desenho de Prótese , Falha de Prótese , Análise Radioestereométrica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...