Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 611(7936): 563-569, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36352220

RESUMO

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Assuntos
Regulação da Expressão Gênica , Hepatócitos , Fígado , Malária , Parasitos , Plasmodium berghei , Análise de Célula Única , Animais , Hepatócitos/citologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Fígado/anatomia & histologia , Fígado/citologia , Fígado/imunologia , Fígado/parasitologia , Malária/genética , Malária/imunologia , Malária/parasitologia , Parasitos/genética , Parasitos/imunologia , Parasitos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Plasmodium berghei/metabolismo , Imagem Individual de Molécula , Análise de Sequência de RNA , Ferro/metabolismo , Ácidos Graxos/metabolismo , Transcrição Gênica , Genes de Protozoários/genética , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia
2.
Proc Natl Acad Sci U S A ; 116(20): 9979-9988, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31028144

RESUMO

Cerebral malaria (CM) is a major cause of death due to Plasmodium infection. Both parasite and host factors contribute to the onset of CM, but the precise cellular and molecular mechanisms that contribute to its pathogenesis remain poorly characterized. Unlike conventional αß-T cells, previous studies on murine γδ-T cells failed to identify a nonredundant role for this T cell subset in experimental cerebral malaria (ECM). Here we show that mice lacking γδ-T cells are resistant to ECM when infected with Plasmodium berghei ANKA sporozoites, the liver-infective form of the parasite and the natural route of infection, in contrast with their susceptible phenotype if challenged with P. berghei ANKA-infected red blood cells that bypass the liver stage of infection. Strikingly, the presence of γδ-T cells enhanced the expression of Plasmodium immunogenic factors and exacerbated subsequent systemic and brain-infiltrating inflammatory αß-T cell responses. These phenomena were dependent on the proinflammatory cytokine IFN-γ, which was required during liver stage for modulation of the parasite transcriptome, as well as for downstream immune-mediated pathology. Our work reveals an unanticipated critical role of γδ-T cells in the development of ECM upon Plasmodium liver-stage infection.


Assuntos
Linfócitos Intraepiteliais/fisiologia , Fígado/imunologia , Malária Cerebral/imunologia , Plasmodium berghei/patogenicidade , Esporozoítos/patogenicidade , Animais , Fígado/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esporozoítos/crescimento & desenvolvimento
4.
Cell Microbiol ; 18(3): 437-50, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26399761

RESUMO

Autophagy plays an important role in the defence against intracellular pathogens. However, some microorganisms can manipulate this host cell pathway to their advantage. In this study, we addressed the role of host cell autophagy during Plasmodium berghei liver infection. We show that vesicles containing the autophagic marker LC3 surround parasites from early time-points after invasion and throughout infection and colocalize with the parasitophorous vacuole membrane. Moreover, we show that the LC3-positive vesicles that surround Plasmodium parasites are amphisomes that converge from the endocytic and autophagic pathways, because they contain markers of both pathways. When the host autophagic pathway was inhibited by silencing several of its key regulators such as LC3, Beclin1, Vps34 or Atg5, we observed a reduction in parasite size. We also found that LC3 surrounds parasites in vivo and that parasite load is diminished in a mouse model deficient for autophagy. Together, these results show the importance of the host autophagic pathway for parasite development during the liver stage of Plasmodium infection.


Assuntos
Autofagia/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Fígado/parasitologia , Malária/patologia , Plasmodium berghei/patogenicidade , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Fígado/patologia , Malária/parasitologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo
5.
EMBO Rep ; 16(8): 955-64, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26113366

RESUMO

Upon infection of a mammalian host, Plasmodium parasites first replicate inside hepatocytes, generating thousands of new parasites. Although Plasmodium intra-hepatic development represents a substantial metabolic challenge to the host hepatocyte, how infected cells respond to and integrate this stress remains poorly understood. Here, we present proteomic and transcriptomic analyses, revealing that the endoplasmic reticulum (ER)-resident unfolded protein response (UPR) is activated in host hepatocytes upon Plasmodium berghei infection. The expression of XBP1s--the active form of the UPR mediator XBP1--and the liver-specific UPR mediator CREBH is induced by P. berghei infection in vivo. Furthermore, this UPR induction increases parasite liver burden. Altogether, our data suggest that ER stress is a central feature of P. berghei intra-hepatic development, contributing to the success of infection.


Assuntos
Estresse do Retículo Endoplasmático , Hepatócitos/parasitologia , Interações Hospedeiro-Parasita , Malária/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Resposta a Proteínas não Dobradas , Animais , Proteínas de Ligação a DNA/genética , Perfilação da Expressão Gênica , Hepatócitos/fisiologia , Hepatócitos/ultraestrutura , Estágios do Ciclo de Vida , Malária/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Carga Parasitária , Plasmodium berghei/patogenicidade , Proteômica , Fatores de Transcrição de Fator Regulador X , Transdução de Sinais/genética , Fatores de Transcrição/genética , Proteína 1 de Ligação a X-Box
6.
Chembiochem ; 15(13): 1920-30, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25111632

RESUMO

Malaria, an infectious disease caused by eukaryotic parasites of the genus Plasmodium, afflicts hundreds of millions of people every year. Both the parasite and its host utilize protein kinases to regulate essential cellular processes. Bioinformatic analyses of parasite genomes predict at least 65 protein kinases, but their biological functions and therapeutic potential are largely unknown. We profiled 1358 small-molecule kinase inhibitors to evaluate the role of both the human and the malaria kinomes in Plasmodium infection of liver cells, the parasites' obligatory but transient developmental stage that precedes the symptomatic blood stage. The screen identified several small molecules that inhibit parasite load in liver cells, some with nanomolar efficacy, and each compound was subsequently assessed for activity against blood-stage malaria. Most of the screening hits inhibited both liver- and blood-stage malaria parasites, which have dissimilar gene expression profiles and infect different host cells. Evaluation of existing kinase activity profiling data for the library members suggests that several kinases are essential to malaria parasites, including cyclin-dependent kinases (CDKs), glycogen synthase kinases, and phosphoinositide-3-kinases. CDK inhibitors were found to bind to Plasmodium protein kinase 5, but it is likely that these compounds target multiple parasite kinases. The dual-stage inhibition of the identified kinase inhibitors makes them useful chemical probes and promising starting points for antimalarial development.


Assuntos
Genoma de Protozoário/genética , Malária/genética , Plasmodium/genética , Proteínas Quinases/genética , Animais , Antimaláricos/química , Biologia Computacional , Avaliação Pré-Clínica de Medicamentos , Humanos , Fígado/parasitologia , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium/enzimologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas
7.
Malar J ; 13: 15, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24400642

RESUMO

BACKGROUND: The first phase of malaria infection occurs in the liver and is clinically silent. Inside hepatocytes each Plasmodium sporozoite replicate into thousands of erythrocyte-infectious merozoites that when released into the blood stream result in clinical symptoms of the disease. The time between sporozoite inoculation and the appearance of parasites in the blood is defined as the pre-patent period, which is classically analysed by time-consuming and labor-intensive techniques, such as microscopy and PCR. METHODS: Luciferase-expressing Plasmodium berghei parasites were used to measure pre-patent period of malaria infection in rodents using a bioluminescence assay that requires only one microliter of blood collected from the tail-vein. The accuracy and sensitivity of this new method was compared with conventional microscopy and PCR based techniques, and its capacity to measure the impact of anti-malarial interventions against the liver evaluated. RESULTS: The described method is very sensitive allowing the detection of parasites during the first cycles of blood stage replication. It accurately translates differences in liver load due to inoculation of different sporozoite doses as well as a result of treatment with different primaquine regimens. CONCLUSIONS: A novel, simple, fast, and sensitive method to measure pre-patent period of malaria infection in rodents is described here. The sensitivity and accuracy of this new method is comparable to standard PCR and microscopy-based techniques, respectively.


Assuntos
Medições Luminescentes/métodos , Malária/diagnóstico , Plasmodium berghei/isolamento & purificação , Animais , Sangue/parasitologia , Luciferases/metabolismo , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Reação em Cadeia da Polimerase , Estatísticas não Paramétricas
8.
Commun Biol ; 6(1): 205, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36810637

RESUMO

Eukaryotes have canonical pathways for responding to amino acid (AA) availability. Under AA-limiting conditions, the TOR complex is repressed, whereas the sensor kinase GCN2 is activated. While these pathways have been highly conserved throughout evolution, malaria parasites are a rare exception. Despite auxotrophic for most AA, Plasmodium does not have either a TOR complex nor the GCN2-downstream transcription factors. While Ile starvation has been shown to trigger eIF2α phosphorylation and a hibernation-like response, the overall mechanisms mediating detection and response to AA fluctuation in the absence of such pathways has remained elusive. Here we show that Plasmodium parasites rely on an efficient sensing pathway to respond to AA fluctuations. A phenotypic screen of kinase knockout mutant parasites identified nek4, eIK1 and eIK2-the last two clustering with the eukaryotic eIF2α kinases-as critical for Plasmodium to sense and respond to distinct AA-limiting conditions. Such AA-sensing pathway is temporally regulated at distinct life cycle stages, allowing parasites to actively fine-tune replication and development in response to AA availability. Collectively, our data disclose a set of heterogeneous responses to AA depletion in malaria parasites, mediated by a complex mechanism that is critical for modulating parasite growth and survival.


Assuntos
Aminoácidos , Plasmodium , Aminoácidos/deficiência , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo , Fosforilação , Fosfotransferases/metabolismo , Plasmodium/enzimologia , Plasmodium/genética
9.
Nat Microbiol ; 8(7): 1280-1292, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37277533

RESUMO

For Plasmodium falciparum, the most widespread and virulent malaria parasite that infects humans, persistence depends on continuous asexual replication in red blood cells, while transmission to their mosquito vector requires asexual blood-stage parasites to differentiate into non-replicating gametocytes. This decision is controlled by stochastic derepression of a heterochromatin-silenced locus encoding AP2-G, the master transcription factor of sexual differentiation. The frequency of ap2-g derepression was shown to be responsive to extracellular phospholipid precursors but the mechanism linking these metabolites to epigenetic regulation of ap2-g was unknown. Through a combination of molecular genetics, metabolomics and chromatin profiling, we show that this response is mediated by metabolic competition for the methyl donor S-adenosylmethionine between histone methyltransferases and phosphoethanolamine methyltransferase, a critical enzyme in the parasite's pathway for de novo phosphatidylcholine synthesis. When phosphatidylcholine precursors are scarce, increased consumption of SAM for de novo phosphatidylcholine synthesis impairs maintenance of the histone methylation responsible for silencing ap2-g, increasing the frequency of derepression and sexual differentiation. This provides a key mechanistic link that explains how LysoPC and choline availability can alter the chromatin status of the ap2-g locus controlling sexual differentiation.


Assuntos
Malária , Parasitos , Animais , Humanos , Parasitos/genética , Parasitos/metabolismo , Histonas/metabolismo , Diferenciação Sexual , Metilação , Epigênese Genética , Malária/parasitologia , Cromatina , Fosfatidilcolinas , Fosfolipídeos
10.
Biochem Biophys Res Commun ; 424(2): 295-300, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22749996

RESUMO

Body skeletal muscles formation starts with somite differentiation, due to signals from surrounding tissues. Somite ventral portion forms the sclerotome while its dorsal fraction constitutes the dermamyotome, and later the dermatome and myotome. Relative levels of BMP activity have been proposed to control several aspects of somite development, namely the time and location of myogenesis within the somite. The fine-tuning of BMP activity is primarily achieved via negative regulation by diffusible BMP inhibitors, such as Noggin and Chordin, and on a secondary level by proteins cleaving these inhibitors, such as BMP1/Tolloid metalloprotease family members. Herein, we carefully described the somitic expression of colloid-like1, one of the chick BMP1/Tolloid homologues, and found that this gene is specifically expressed in the 10 most anterior somites, suggesting that it may be involved in neck muscle formation. By using in ovo microsurgery and tridimensional embryo tissue culture techniques we assessed the function of surrounding structures, neural tube, notochord, surface ectoderm and lateral plate mesoderm, on the maintenance of somitic colloid-like1 gene expression. We unveil that a signal coming from the neural tube is responsible for this expression and rule out the main candidate pathway, Wnt. By comparing the somitic colloid-like1 gene expression with that of related signaling partners, such as BMP4, Noggin and Chordin, we propose that colloid-like1 plays a role in the reinforcement of BMP4 activity in the medial portion of the 10 most anterior dermomyotomes, thus belonging to the molecular machinery controlling neck muscle development in the chick.


Assuntos
Proteína Morfogenética Óssea 1/genética , Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Muscular/genética , Músculos do Pescoço/embriologia , Somitos/metabolismo , Metaloproteases Semelhantes a Toloide/genética , Animais , Proteína Morfogenética Óssea 4/genética , Proteínas de Transporte/genética , Embrião de Galinha , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Wnt/genética
11.
Nanomedicine ; 8(1): 17-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22033078

RESUMO

Infection of liver cells by Plasmodium, the malaria parasite, is a clinically silent, obligatory step of the parasite's life cycle. The authors studied the progression of Plasmodium infection in hepatic cells by atomic force microscopy, measuring both topographical and nanomechanical changes upon infection. In recent years, several studies have suggested that cellular nanomechanical properties can be correlated with disease progression. The authors' results show that infected cells exhibit considerable topographical changes, which can be correlated with the presence of the parasite, leading to a significant roughening of the cell membrane. The nanomechanical measurements showed that infected cells were significantly stiffer than noninfected cells. Furthermore, the stiffening of the cells appeared to be a cellular reaction to the Plasmodium infection, rather than a result of the stiffness of the invading parasites themselves. This article provides the first evidence of mechanical changes occurring in hepatic cells in response to Plasmodium infection. FROM THE CLINICAL EDITOR: The authors have studied the progression of Plasmodium infection in hepatic cells by atomic force microscopy, measuring topographical and nanomechanical changes upon infection. The nanomechanical measurements demonstrated that infected cells were significantly stiffer than noninfected cells.


Assuntos
Membrana Celular/ultraestrutura , Forma Celular , Hepatócitos/ultraestrutura , Fígado/ultraestrutura , Animais , Linhagem Celular Tumoral , Hepatócitos/parasitologia , Humanos , Fígado/parasitologia , Malária/parasitologia , Microscopia de Força Atômica , Plasmodium berghei/ultraestrutura
12.
Cell Rep ; 39(9): 110886, 2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35649358

RESUMO

Intracellular pathogens manipulate host cells to survive and thrive. Cellular sensing and signaling pathways are among the key host machineries deregulated to favor infection. In this study, we show that liver-stage Plasmodium parasites compete with the host to sequester a host endosomal-adaptor protein (APPL1) known to regulate signaling in response to endocytosis. The enrichment of APPL1 at the parasitophorous vacuole membrane (PVM) involves an atypical Plasmodium Rab5 isoform (Rab5b). Depletion of host APPL1 alters neither the infection nor parasite development; however, upon overexpression of a GTPase-deficient host Rab5 mutant (hRab5_Q79L), the parasites are smaller and their PVM is stripped of APPL1. Infection with the GTPase-deficient Plasmodium berghei Rab5b mutant (PbRab5b_Q91L) in this case rescues the PVM APPL1 signal and parasite size. In summary, we observe a robust correlation between the level of APPL1 retention at the PVM and parasite size during exoerythrocytic development.


Assuntos
Parasitos , Plasmodium berghei , Animais , Endocitose , GTP Fosfo-Hidrolases/metabolismo , Fígado/metabolismo
13.
Nat Commun ; 11(1): 5654, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-33159090

RESUMO

Plasmodium parasites possess a translocon that exports parasite proteins into the infected erythrocyte. Although the translocon components are also expressed during the mosquito and liver stage of infection, their function remains unexplored. Here, using a combination of genetic and chemical assays, we show that the translocon component Exported Protein 2 (EXP2) is critical for invasion of hepatocytes. EXP2 is a pore-forming protein that is secreted from the sporozoite upon contact with the host cell milieu. EXP2-deficient sporozoites are impaired in invasion, which can be rescued by the exogenous administration of recombinant EXP2 and alpha-hemolysin (an S. aureus pore-forming protein), as well as by acid sphingomyelinase. The latter, together with the negative impact of chemical and genetic inhibition of acid sphingomyelinase on invasion, reveals that EXP2 pore-forming activity induces hepatocyte membrane repair, which plays a key role in parasite invasion. Overall, our findings establish a novel and critical function for EXP2 that leads to an active participation of the host cell in Plasmodium sporozoite invasion, challenging the current view of the establishment of liver stage infection.


Assuntos
Hepatócitos/parasitologia , Fígado/parasitologia , Malária/parasitologia , Plasmodium berghei/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Humanos , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/crescimento & desenvolvimento , Transporte Proteico , Proteínas de Protozoários/genética , Esporozoítos/genética , Esporozoítos/metabolismo
14.
Sci Rep ; 10(1): 21026, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33273556

RESUMO

Iron homeostasis is an essential biological process that ensures the tissue distribution of iron for various cellular processes. As the major producer of hepcidin, the liver is central to the regulation of iron metabolism. The liver is also home to many immune cells, which upon activation may greatly impact iron metabolism. Here, we focus on the role of invariant natural killer T (iNKT) cells, a subset of T lymphocytes that, in mice, is most abundant in the liver. Activation of iNKT cells with the prototypical glycosphingolipid antigen, α-galactosylceramide, resulted in immune cell proliferation and biphasic changes in iron metabolism. This involved an early phase characterized by hypoferremia, hepcidin induction and ferroportin suppression, and a second phase associated with strong suppression of hepcidin despite elevated levels of circulating and tissue iron. We further show that these changes in iron metabolism are fully dependent on iNKT cell activation. Finally, we demonstrate that the biphasic regulation of hepcidin is independent of NK and Kupffer cells, and is initially driven by the STAT3 inflammatory pathway, whereas the second phase is regulated by repression of the BMP/SMAD signaling pathway. These findings indicate that iNKT activation and the resulting cell proliferation influence iron homeostasis.


Assuntos
Homeostase , Ferro/metabolismo , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Animais , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Proliferação de Células , Galactosilceramidas/imunologia , Hepcidinas/genética , Hepcidinas/metabolismo , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
15.
Sci Rep ; 9(1): 7575, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31110285

RESUMO

Malaria causes hepatic inflammation and damage, which contribute to disease severity. The pro-inflammatory cytokine interleukin (IL)-1α is released by non-hematopoietic or hematopoietic cells during liver injury. This study established the role of IL-1α in the liver pathology caused by blood-stage P. chabaudi malaria. During acute infection, hepatic inflammation and necrosis were accompanied by NLRP3 inflammasome-independent IL-1α production. Systemically, IL-1α deficiency attenuated weight loss and hypothermia but had minor effects on parasitemia control. In the liver, the absence of IL-1α reduced the number of TUNEL+ cells and necrotic lesions. This finding was associated with a lower inflammatory response, including TNF-α production. The main source of IL-1α in the liver of infected mice was inflammatory cells, particularly neutrophils. The implication of IL-1α in liver inflammation and necrosis caused by P. chabaudi infection, as well as in weight loss and hypothermia, opens up new perspectives for improving malaria outcomes by inhibiting IL-1 signaling.


Assuntos
Inflamação/imunologia , Interleucina-1alfa/imunologia , Fígado/patologia , Malária/imunologia , Plasmodium chabaudi/imunologia , Animais , Inflamação/parasitologia , Inflamação/patologia , Fígado/imunologia , Fígado/parasitologia , Malária/parasitologia , Malária/patologia , Masculino , Camundongos Endogâmicos C57BL , Necrose , Fator de Necrose Tumoral alfa/imunologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-32010639

RESUMO

Plasmodium parasites undergo a dramatic transformation during the liver stage of their life cycle, amplifying over 10,000-fold inside infected hepatocytes within a few days. Such a rapid growth requires large-scale interactions with, and manipulations of, host cell functions. Whereas hepatocyte polarity is well-known to be critical for liver function, little is presently known about its involvement during the liver stage of Plasmodium development. Apical domains of hepatocytes are critical components of their polarity machinery and constitute the bile canalicular network, which is central to liver function. Here, we employed high resolution 3-D imaging and advanced image analysis of Plasmodium-infected liver tissues to show that the parasite associates preferentially with the apical domain of hepatocytes and induces alterations in the organization of these regions, resulting in localized changes in the bile canalicular architecture in the liver tissue. Pharmacological perturbation of the bile canalicular network by modulation of AMPK activity reduces the parasite's association with bile canaliculi and arrests the parasite development. Our findings using Plasmodium-infected liver tissues reveal a host-Plasmodium interaction at the level of liver tissue organization. We demonstrate for the first time a role for bile canaliculi, a central component of the hepatocyte polarity machinery, during the liver stage of Plasmodium development.


Assuntos
Hepatócitos/parasitologia , Interações Hospedeiro-Patógeno/fisiologia , Fígado/parasitologia , Malária/parasitologia , Plasmodium berghei/fisiologia , Animais , Ácidos e Sais Biliares/análise , Canalículos Biliares/diagnóstico por imagem , Canalículos Biliares/parasitologia , Canalículos Biliares/patologia , Modelos Animais de Doenças , Imageamento Tridimensional , Estágios do Ciclo de Vida , Fígado/diagnóstico por imagem , Fígado/patologia , Malária/diagnóstico por imagem , Malária/patologia , Camundongos , Camundongos Endogâmicos C57BL
17.
Cell Host Microbe ; 23(6): 749-758, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29902440

RESUMO

Parasites undergo complex life cycles that comprise a wide variety of cellular differentiation events in different host compartments and transmission across multiple hosts. As parasites depend on host resources, it is not surprising they have developed efficient mechanisms to sense alterations and adapt to the available resources in a wide range of environments. Here we provide an overview of the nutritional needs of different parasites throughout their diverse life stages and highlight recent insights into strategies that both hosts and parasites have developed to meet these nutritional requirements needed for defense, survival, and replication. These studies will provide the foundation for a systems-level understanding of host-parasite interactions, which will require the integration of molecular, epidemiologic, and mechanistic data and the application of interdisciplinary approaches to model parasite regulatory networks that are triggered by alterations in host resources.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Nutrientes/metabolismo , Parasitos/metabolismo , Adaptação Biológica , Animais , Estágios do Ciclo de Vida , Estado Nutricional/fisiologia , Parasitos/patogenicidade , Plasmodium/metabolismo , Toxoplasma/metabolismo , Trypanosoma/metabolismo
18.
Cell Metab ; 27(4): 708-709, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29617637

RESUMO

Several vacuolar bacteria and parasites, such as Legionella, Chlamydia, and Toxoplasma, have been reported to grow associated with host mitochondria. The reason behind this phenomenon remains elusive. In this issue of Cell Metabolism, Pernas et al. (2018) propose that fusion of host mitochondria limits the availability of fatty acids needed for Toxoplasma gondii replication.


Assuntos
Parasitos , Toxoplasma , Animais , Ácidos Graxos , Interações Hospedeiro-Parasita , Mitocôndrias
19.
Nat Microbiol ; 3(1): 17-25, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29109477

RESUMO

The causative agent of malaria, Plasmodium, replicates inside a membrane-bound parasitophorous vacuole (PV), which shields this intracellular parasite from the cytosol of the host cell 1 . One common threat for intracellular pathogens is the homeostatic process of autophagy, through which cells capture unwanted intracellular material for lysosomal degradation 2 . During the liver stage of a malaria infection, Plasmodium parasites are targeted by the autophagy machinery of the host cell, and the PV membrane (PVM) becomes decorated with several autophagy markers, including LC3 (microtubule-associated protein 1 light chain 3) 3,4 . Here we show that Plasmodium berghei parasites infecting hepatic cells rely on the PVM transmembrane protein UIS3 to avoid elimination by host-cell-mediated autophagy. We found that UIS3 binds host LC3 through a non-canonical interaction with a specialized surface on LC3 where host proteins with essential functions during autophagy also bind. UIS3 acts as a bona fide autophagy inhibitor by competing with host LC3-interacting proteins for LC3 binding. Our work identifies UIS3, one of the most promising candidates for a genetically attenuated vaccine against malaria 5 , as a unique and potent mediator of autophagy evasion in Plasmodium. We propose that the protein-protein interaction between UIS3 and host LC3 represents a target for antimalarial drug development.


Assuntos
Autofagia/fisiologia , Hepatócitos/patologia , Malária/patologia , Malária/parasitologia , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Plasmodium berghei/genética , Animais , Autofagossomos/metabolismo , Linhagem Celular , Células HEK293 , Células Hep G2 , Hepatócitos/parasitologia , Hepatócitos/ultraestrutura , Interações Hospedeiro-Patógeno , Humanos , Malária/fisiopatologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Plasmodium berghei/metabolismo , Plasmodium berghei/patogenicidade , Ligação Proteica , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Vacúolos/metabolismo
20.
Nat Microbiol ; 2(12): 1600-1607, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28947801

RESUMO

The relevance of genetic factors in conferring protection to severe malaria has been demonstrated, as in the case of sickle cell trait and G6PD deficiency 1 . However, it remains unknown whether environmental components, such as dietary or metabolic variations, can contribute to the outcome of infection 2 . Here, we show that administration of a high-fat diet to mice for a period as short as 4 days impairs Plasmodium liver infection by over 90%. Plasmodium sporozoites can successfully invade and initiate replication but die inside hepatocytes, thereby are unable to cause severe disease. Transcriptional analyses combined with genetic and chemical approaches reveal that this impairment of infection is mediated by oxidative stress. We show that reactive oxygen species, probably spawned from fatty acid ß-oxidation, directly impact Plasmodium survival inside hepatocytes, and parasite load can be rescued by exogenous administration of the antioxidant N-acetylcysteine or the ß-oxidation inhibitor etomoxir. Together, these data reveal that acute and transient dietary alterations markedly impact the establishment of a Plasmodium infection and disease outcome.


Assuntos
Dieta Hiperlipídica/métodos , Interações Hospedeiro-Parasita/genética , Malária/dietoterapia , Acetilcisteína/metabolismo , Animais , Antioxidantes/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Células Hep G2 , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Humanos , Fígado/metabolismo , Fígado/parasitologia , Hepatopatias/metabolismo , Hepatopatias/parasitologia , Macrófagos/parasitologia , Macrófagos/patologia , Malária/sangue , Malária/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Carga Parasitária , Plasmodium berghei , Espécies Reativas de Oxigênio , Traço Falciforme/metabolismo , Esporozoítos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA