Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
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
2.
Trends Parasitol ; 39(6): 445-460, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37061442

RESUMO

Malaria is a febrile illness caused by species of the protozoan parasite Plasmodium and is characterized by recursive infections of erythrocytes, leading to clinical symptoms and pathology. In mammals, Plasmodium parasites undergo a compulsory intrahepatic development stage before infecting erythrocytes. Liver-stage parasites have a metabolic configuration to facilitate the replication of several thousand daughter parasites. Their metabolism is of interest to identify cellular pathways essential for liver infection, to kill the parasite before onset of the disease. In this review, we summarize the current knowledge on nutrient acquisition and biosynthesis by liver-stage parasites mostly generated in murine malaria models, gaps in knowledge, and challenges to create a holistic view of the development and deficiencies in this field.


Assuntos
Malária , Parasitos , Plasmodium , Animais , Camundongos , Plasmodium/metabolismo , Fígado/parasitologia , Malária/parasitologia , Parasitos/metabolismo , Eritrócitos/parasitologia , Proteínas de Protozoários/metabolismo , Mamíferos
3.
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
4.
Immunity ; 56(3): 592-605.e8, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36804959

RESUMO

Plasmodium replicates within the liver prior to reaching the bloodstream and infecting red blood cells. Because clinical manifestations of malaria only arise during the blood stage of infection, a perception exists that liver infection does not impact disease pathology. By developing a murine model where the liver and blood stages of infection are uncoupled, we showed that the integration of signals from both stages dictated mortality outcomes. This dichotomy relied on liver stage-dependent activation of Vγ4+ γδ T cells. Subsequent blood stage parasite loads dictated their cytokine profiles, where low parasite loads preferentially expanded IL-17-producing γδ T cells. IL-17 drove extra-medullary erythropoiesis and concomitant reticulocytosis, which protected mice from lethal experimental cerebral malaria (ECM). Adoptive transfer of erythroid precursors could rescue mice from ECM. Modeling of γδ T cell dynamics suggests that this protective mechanism may be key for the establishment of naturally acquired malaria immunity among frequently exposed individuals.


Assuntos
Eritropoese , Malária Cerebral , Animais , Camundongos , Eritrócitos , Interleucina-17 , Fígado/parasitologia , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T gama-delta , Malária
5.
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
6.
Nat Commun ; 13(1): 4976, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-36008486

RESUMO

The development of next-generation antimalarials that are efficacious against the human liver and asexual blood stages is recognized as one of the world's most pressing public health challenges. In recent years, aminoacyl-tRNA synthetases, including prolyl-tRNA synthetase, have emerged as attractive targets for malaria chemotherapy. We describe the development of a single-step biochemical assay for Plasmodium and human prolyl-tRNA synthetases that overcomes critical limitations of existing technologies and enables quantitative inhibitor profiling with high sensitivity and flexibility. Supported by this assay platform and co-crystal structures of representative inhibitor-target complexes, we develop a set of high-affinity prolyl-tRNA synthetase inhibitors, including previously elusive aminoacyl-tRNA synthetase triple-site ligands that simultaneously engage all three substrate-binding pockets. Several compounds exhibit potent dual-stage activity against Plasmodium parasites and display good cellular host selectivity. Our data inform the inhibitor requirements to overcome existing resistance mechanisms and establish a path for rational development of prolyl-tRNA synthetase-targeted anti-malarial therapies.


Assuntos
Aminoacil-tRNA Sintetases , Antimaláricos , Plasmodium , Aminoacil-tRNA Sintetases/química , Antimaláricos/química , Antimaláricos/farmacologia , Humanos , Piperidinas , Plasmodium falciparum , Quinazolinonas , RNA de Transferência
7.
Nat Commun ; 13(1): 3747, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35768411

RESUMO

Severe malaria can manifest itself with a variety of well-recognized clinical phenotypes that are highly predictive of death - severe anaemia, coma (cerebral malaria), multiple organ failure, and respiratory distress. The reasons why an infected individual develops one pathology rather than another remain poorly understood. Here we use distinct rodent models of infection to show that the host microbiota is a contributing factor for the development of respiratory distress syndrome and host mortality in the context of malaria infections (malaria-associated acute respiratory distress syndrome, MA-ARDS). We show that parasite sequestration in the lung results in sustained immune activation. Subsequent production of the anti-inflammatory cytokine IL-10 by T cells compromises microbial control, leading to severe lung disease. Notably, bacterial clearance with linezolid, an antibiotic commonly used in the clinical setting to control lung-associated bacterial infections, prevents MA-ARDS-associated lethality. Thus, we propose that the host's anti-inflammatory response to limit tissue damage can result in loss of microbial control, which promotes MA-ARDS. This must be considered when intervening against life-threatening respiratory complications.


Assuntos
Malária , Microbiota , Síndrome do Desconforto Respiratório , Animais , Modelos Animais de Doenças , Pulmão/patologia , Malária/complicações , Malária/parasitologia , Plasmodium berghei/fisiologia
8.
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
9.
iScience ; 25(5): 104281, 2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35573190

RESUMO

Parasite-derived PVM-resident proteins are critical for complete parasite development inside hepatocytes, although the function of most of these proteins remains unknown. Here, we show that the upregulated in infectious sporozoites 4 (UIS4) protein, resident at the PVM, interacts with the host cell actin. By suppressing filamentous actin formation, UIS4 avoids parasite elimination. Host cell actin dynamics increases around UIS4-deficient parasites, which is associated with subsequent parasite elimination. Notably, parasite elimination is impaired significantly by the inhibition of host myosin-II, possibly through relieving the compression generated by actomyosin complexes at the host-parasite interface. Together, these data reveal that UIS4 has a critical role in the evasion of host defensive mechanisms, enabling hence EEF survival and development.

10.
Int J Parasitol ; 52(11): 711-715, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35367213

RESUMO

While the liver and blood stages of the Plasmodium life cycle are commonly regarded as two separate fields of malaria research, several studies have pointed towards the existence of a bidirectional cross-talk, where one stage of mammalian infection may impact the establishment and progression of the other. Despite the constraints in experimentally addressing concurrent liver and blood stage Plasmodium infections, animal models and clinical studies have unveiled a plethora of molecular interactions between the two. Here, we review the current knowledge on the reciprocal influence of hepatic and erythrocytic infection by malaria parasites, and discuss its impacts on immunity, pathology and vaccination against this deadly disease.


Assuntos
Malária , Parasitos , Plasmodium , Animais , Malária/parasitologia , Fígado/parasitologia , Estágios do Ciclo de Vida , Mamíferos
11.
Eur J Immunol ; 52(1): 149-160, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34695227

RESUMO

During the COVID-19 pandemic, Portugal has experienced three distinct SARS-CoV-2 infection waves. We previously documented the prevalence of SARS-CoV-2 immunity, measured by specific antibodies, in September 2020, 6 months after the initial moderate wave. Here, we show the seroprevalence changes 6 months later, up to the second week of March 2021, shortly following the third wave, which was one of the most severe in the world, and 2 months following the start of the vaccination campaign. A longitudinal epidemiological study was conducted, with a stratified quota sample of the Portuguese population. Serological testing was performed, including ELISA determination of antibody class and titers. The proportion of seropositives, which was 2.2% in September 2020, rose sharply to 17.3% (95% CI: 15.8-18.8%) in March 2021. Importantly, circulating IgG and IgA antibody levels were very stable 6 months after the initial determination and up to a year after initial infection, indicating long-lasting infection immunity against SARS-CoV-2. Moreover, vaccinated people had higher IgG levels from 3 weeks post-vaccination when compared with previously infected people at the same time post-infection.


Assuntos
Anticorpos Antivirais/imunologia , Teste Sorológico para COVID-19 , COVID-19 , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , SARS-CoV-2/imunologia , Adolescente , Adulto , COVID-19/epidemiologia , COVID-19/imunologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Portugal/epidemiologia , Prevalência , Fatores de Tempo
12.
Emerg Infect Dis ; 27(11): 2878-2881, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34437830

RESUMO

In September 2020, we tested 13,398 persons in Portugal for antibodies against severe acute respiratory syndrome coronavirus 2 by using a quota sample stratified by age and population density. We found a seroprevalence of 2.2%, 3-4 times larger than the official number of cases at the end of the first wave of the pandemic.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Pandemias , Portugal/epidemiologia , Prevalência , Estudos Soroepidemiológicos
13.
Commun Biol ; 3(1): 688, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33214643

RESUMO

The malaria parasite Plasmodium obligatorily infects and replicates inside hepatocytes surrounded by a parasitophorous vacuole membrane (PVM), which is decorated by the host-cell derived autophagy protein LC3. We have previously shown that the parasite-derived, PVM-resident protein UIS3 sequesters LC3 to avoid parasite elimination by autophagy from hepatocytes. Here we show that a small molecule capable of disrupting this interaction triggers parasite elimination in a host cell autophagy-dependent manner. Molecular docking analysis of more than 20 million compounds combined with a phenotypic screen identified one molecule, C4 (4-{[4-(4-{5-[3-(trifluoromethyl) phenyl]-1,2,4-oxadiazol-3-yl}benzyl)piperazino]carbonyl}benzonitrile), capable of impairing infection. Using biophysical assays, we established that this impairment is due to the ability of C4 to disrupt UIS3-LC3 interaction, thus inhibiting the parasite's ability to evade the host autophagy response. C4 impacts infection in autophagy-sufficient cells without harming the normal autophagy pathway of the host cell. This study, by revealing the disruption of a critical host-parasite interaction without affecting the host's normal function, uncovers an efficient anti-malarial strategy to prevent this deadly disease.


Assuntos
Antimaláricos/farmacologia , Proteínas de Membrana/metabolismo , Plasmodium berghei/fisiologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Animais , Antimaláricos/uso terapêutico , Autofagia , Adesão Celular , Bases de Dados de Compostos Químicos , Humanos , Malária/tratamento farmacológico , Malária/parasitologia , Masculino , Proteínas de Membrana/química , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Conformação Proteica , Proteínas de Protozoários/química
14.
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
15.
Stem Cell Reports ; 14(6): 1123-1134, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32442532

RESUMO

Asymptomatic and obligatory liver stage (LS) infection of Plasmodium parasites presents an attractive target for antimalarial vaccine and drug development. Lack of robust cellular models to study LS infection has hindered the discovery and validation of host genes essential for intrahepatic parasite development. Here, we present a chemically differentiated mouse embryonic stem cell (ESC)-based LS model, which supports complete development of Plasmodium berghei exoerythrocytic forms (EEFs) and can be used to define new host-parasite interactions. Using our model, we established that host Pnpla2, coding for adipose triglyceride lipase, is dispensable for P. berghei EEF development. In addition, we also evaluated in-vitro-differentiated human hepatocyte-like cells (iHLCs) to study LS of P. berghei and found it to be a sub-optimal infection model. Overall, our results present a new mouse ESC-based P. berghei LS infection model that can be utilized to study the impact of host genetic variation on parasite development.


Assuntos
Diferenciação Celular , Hepatócitos/parasitologia , Interações Hospedeiro-Parasita , Malária/parasitologia , Células-Tronco Embrionárias Murinas/citologia , Plasmodium berghei/patogenicidade , Animais , Linhagem Celular , Células Cultivadas , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Lipase/genética , Lipase/metabolismo , Malária/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo
16.
Science ; 368(6492): 746-753, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32409471

RESUMO

Malarial rhythmic fevers are the consequence of the synchronous bursting of red blood cells (RBCs) on completion of the malaria parasite asexual cell cycle. Here, we hypothesized that an intrinsic clock in the parasite Plasmodium chabaudi underlies the 24-hour-based rhythms of RBC bursting in mice. We show that parasite rhythms are flexible and lengthen to match the rhythms of hosts with long circadian periods. We also show that malaria rhythms persist even when host food intake is evenly spread across 24 hours, suggesting that host feeding cues are not required for synchrony. Moreover, we find that the parasite population remains synchronous and rhythmic even in an arrhythmic clock mutant host. Thus, we propose that parasite rhythms are generated by the parasite, possibly to anticipate its circadian environment.


Assuntos
Ritmo Circadiano/fisiologia , Febre/fisiopatologia , Febre/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Malária/fisiopatologia , Malária/parasitologia , Plasmodium chabaudi/fisiologia , Animais , Proteínas CLOCK/genética , Ritmo Circadiano/genética , Sinais (Psicologia) , Escuridão , Ingestão de Alimentos , Eritrócitos/parasitologia , Comportamento Alimentar , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita/genética , Camundongos , Camundongos Mutantes , Plasmodium chabaudi/genética , Transcrição Gênica
17.
Trends Parasitol ; 36(1): 11-18, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31787522

RESUMO

Plasmodium, the causative agent of malaria, is responsible for more than 200 million new infections and 400 000 deaths yearly. While in recent years the influence of the microbiota in homeostasis and a wide variety of disorders has taken center stage, its contribution during malaria infections has only now started to emerge. The few published studies suggest two distinct but complementary directions. Plasmodium infections can cause significant alterations in host (at least gut) microbiota, and host gut microbiota can influence the clinical outcome of malaria infections. In this opinion article, we highlight the most fundamental unanswered questions in the field that will, hopefully, point future research directions towards unveiling key mechanistic insights of the Plasmodium-host-microbiota axis.


Assuntos
Interações Hospedeiro-Parasita , Malária/microbiologia , Microbiota/fisiologia , Animais , Microbioma Gastrointestinal/fisiologia , Humanos , Plasmodium/fisiologia
18.
JCI Insight ; 4(24)2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31852843

RESUMO

Despite an unprecedented 2 decades of success, the combat against malaria - the mosquito-transmitted disease caused by Plasmodium parasites - is no longer progressing. Efforts toward eradication are threatened by the lack of an effective vaccine and a rise in antiparasite drug resistance. Alternative approaches are urgently needed. Repurposing of available, approved drugs with distinct modes of action are being considered as viable and immediate adjuncts to standard antimicrobial treatment. Such strategies may be well suited to the obligatory and clinically silent first phase of Plasmodium infection, where massive parasite replication occurs within hepatocytes in the liver. Here, we report that the widely used antidiabetic drug, metformin, impairs parasite liver stage development of both rodent-infecting Plasmodium berghei and human-infecting P. falciparum parasites. Prophylactic treatment with metformin curtails parasite intracellular growth in vitro. An additional effect was observed in mice with a decrease in the numbers of infected hepatocytes. Moreover, metformin provided in combination with conventional liver- or blood-acting antimalarial drugs further reduced the total burden of P. berghei infection and substantially lessened disease severity in mice. Together, our findings indicate that repurposing of metformin in a prophylactic regimen could be considered for malaria chemoprevention.


Assuntos
Antimaláricos/farmacologia , Malária/prevenção & controle , Metformina/farmacologia , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Animais , Antimaláricos/uso terapêutico , Células Cultivadas , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Quimioterapia Combinada/métodos , Hepatócitos , Humanos , Concentração Inibidora 50 , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/parasitologia , Malária/sangue , Malária/tratamento farmacológico , Malária/parasitologia , Masculino , Mefloquina/farmacologia , Mefloquina/uso terapêutico , Metformina/uso terapêutico , Camundongos , Carga Parasitária , Testes de Sensibilidade Parasitária , Plasmodium berghei/isolamento & purificação , Plasmodium falciparum/isolamento & purificação , Primaquina/farmacologia , Primaquina/uso terapêutico , Cultura Primária de Células
19.
Curr Biol ; 29(13): R632-R634, 2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31287981

RESUMO

Female Anopheles mosquitoes are the definitive hosts of Plasmodium parasites. A new study has found that successful establishment and development of Plasmodium in the Anopheles midgut requires mosquito oogenesis, without affecting egg production.


Assuntos
Anopheles , Parasitos , Plasmodium , Animais , Feminino
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...