Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
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.
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
3.
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
4.
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
5.
Cell Rep ; 16(10): 2539-2545, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27568570

RESUMO

Manipulation of the master regulator of energy homeostasis AMP-activated protein kinase (AMPK) activity is a strategy used by many intracellular pathogens for successful replication. Infection by most pathogens leads to an activation of host AMPK activity due to the energetic demands placed on the infected cell. Here, we demonstrate that the opposite is observed in cells infected with rodent malaria parasites. Indeed, AMPK activity upon the infection of hepatic cells is suppressed and dispensable for successful infection. By contrast, an overactive AMPK is deleterious to intracellular growth and replication of different Plasmodium spp., including the human malaria parasite, P. falciparum. The negative impact of host AMPK activity on infection was further confirmed in mice under conditions that activate its function. Overall, this work establishes the role of host AMPK signaling as a suppressive pathway of Plasmodium hepatic infection and as a potential target for host-based antimalarial interventions.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Interações Hospedeiro-Parasita , Fígado/parasitologia , Malária/enzimologia , Malária/parasitologia , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Estágios do Ciclo de Vida , Fígado/patologia , Malária/patologia , Masculino , Camundongos Endogâmicos C57BL , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/patogenicidade
6.
Nat Commun ; 7: 10403, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26786069

RESUMO

Iron is an essential micronutrient but is also highly toxic. In yeast and plant cells, a key detoxifying mechanism involves iron sequestration into intracellular storage compartments, mediated by members of the vacuolar iron-transporter (VIT) family of proteins. Here we study the VIT homologue from the malaria parasites Plasmodium falciparum (PfVIT) and Plasmodium berghei (PbVIT). PfVIT-mediated iron transport in a yeast heterologous expression system is saturable (Km ∼ 14.7 µM), and selective for Fe(2+) over other divalent cations. PbVIT-deficient P. berghei lines (Pbvit(-)) show a reduction in parasite load in both liver and blood stages of infection in mice. Moreover, Pbvit(-) parasites have higher levels of labile iron in blood stages and are more sensitive to increased iron levels in liver stages, when compared with wild-type parasites. Our data are consistent with Plasmodium VITs playing a major role in iron detoxification and, thus, normal development of malaria parasites in their mammalian host.


Assuntos
Ferro/metabolismo , Plasmodium berghei/metabolismo , Plasmodium berghei/patogenicidade , Proteínas de Protozoários/metabolismo , Vacúolos/metabolismo , Animais , Linhagem Celular , Genótipo , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium berghei/genética , Proteínas de Protozoários/genética
7.
Nat Protoc ; 10(12): 2027-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26584444

RESUMO

The development of therapies and vaccines for human hepatropic pathogens requires robust model systems that enable the study of host-pathogen interactions. However, in vitro liver models of infection typically use either hepatoma cell lines that exhibit aberrant physiology or primary human hepatocytes in culture conditions in which they rapidly lose their hepatic phenotype. To achieve stable and robust in vitro primary human hepatocyte models, we developed micropatterned cocultures (MPCCs), which consist of primary human hepatocytes organized into 2D islands that are surrounded by supportive fibroblast cells. By using this system, which can be established over a period of days, and maintained over multiple weeks, we demonstrate how to recapitulate in vitro hepatic life cycles for the hepatitis B and C viruses and the Plasmodium pathogens P. falciparum and P. vivax. The MPCC platform can be used to uncover aspects of host-pathogen interactions, and it has the potential to be used for drug and vaccine development.


Assuntos
Técnicas de Cocultura/métodos , Hepacivirus/fisiologia , Vírus da Hepatite B/fisiologia , Hepatócitos/parasitologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno , Plasmodium/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Fibroblastos/citologia , Hepatite B/metabolismo , Hepatite C/metabolismo , Hepatócitos/citologia , Humanos , Malária/metabolismo , Camundongos , Plasmodium falciparum/fisiologia , Plasmodium vivax/fisiologia , Análise Serial de Tecidos/métodos
8.
Eukaryot Cell ; 14(1): 96-103, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25416236

RESUMO

Prior to invading nonreplicative erythrocytes, Plasmodium parasites undergo their first obligate step in the mammalian host inside hepatocytes, where each sporozoite replicates to generate thousands of merozoites. While normally quiescent, hepatocytes retain proliferative capacity and can readily reenter the cell cycle in response to diverse stimuli. Many intracellular pathogens, including protozoan parasites, manipulate the cell cycle progression of their host cells for their own benefit, but it is not known whether the hepatocyte cell cycle plays a role during Plasmodium liver stage infection. Here, we show that Plasmodium parasites can be observed in mitotic hepatoma cells throughout liver stage development, where they initially reduce the likelihood of mitosis and ultimately lead to significant acquisition of a binucleate phenotype. However, hepatoma cells pharmacologically arrested in S phase still support robust and complete Plasmodium liver stage development, which thus does not require cell cycle progression in the infected cell in vitro. Furthermore, murine hepatocytes remain quiescent throughout in vivo infection with either Plasmodium berghei or Plasmodium yoelii, as do Plasmodium falciparum-infected primary human hepatocytes, demonstrating that the rapid and prodigious growth of liver stage parasites is accomplished independent of host hepatocyte cell cycle progression during natural infection.


Assuntos
Hepatócitos/fisiologia , Mitose , Plasmodium/crescimento & desenvolvimento , Esporozoítos/fisiologia , Animais , Núcleo Celular/fisiologia , Células Hep G2 , Hepatócitos/citologia , Hepatócitos/parasitologia , Interações Hospedeiro-Parasita , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium/patogenicidade
9.
Bioorg Med Chem Lett ; 24(17): 4151-7, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25103602

RESUMO

The malaria parasite Plasmodium goes through two life stages in the human host, a non-symptomatic liver stage (LS) followed by a blood stage with all clinical manifestation of the disease. In this study, we investigated a series of 2-alkynoic fatty acids (2-AFAs) with chain lengths between 14 and 18 carbon atoms for dual in vitro activity against both life stages. 2-Octadecynoic acid (2-ODA) was identified as the best inhibitor of Plasmodium berghei parasites with ten times higher potency (IC50=0.34 µg/ml) than the control drug. In target determination studies, the same compound inhibited three Plasmodium falciparum FAS-II (PfFAS-II) elongation enzymes PfFabI, PfFabZ, and PfFabG with the lowest IC50 values (0.28-0.80 µg/ml, respectively). Molecular modeling studies provided insights into the molecular aspects underlying the inhibitory activity of this series of 2-AFAs and a likely explanation for the considerably different inhibition potentials. Blood stages of P. falciparum followed a similar trend where 2-ODA emerged as the most active compound, with 20 times less potency. The general toxicity and hepatotoxicity of 2-AFAs were evaluated by in vitro and in vivo methods in mammalian cell lines and zebrafish models, respectively. This study identifies 2-ODA as the most promising antiparasitic 2-AFA, particularly towards P. berghei parasites.


Assuntos
Antimaláricos/farmacologia , Ácido Graxo Sintase Tipo II/antagonistas & inibidores , Ácidos Graxos Insaturados/farmacologia , Malária/tratamento farmacológico , Malária/parasitologia , Plasmodium berghei/enzimologia , Plasmodium falciparum/enzimologia , Animais , Antimaláricos/síntese química , Antimaláricos/química , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ácido Graxo Sintase Tipo II/metabolismo , Ácidos Graxos Insaturados/síntese química , Ácidos Graxos Insaturados/química , Humanos , Modelos Moleculares , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Relação Estrutura-Atividade , Peixe-Zebra
10.
Nat Med ; 20(1): 47-53, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24362933

RESUMO

Before they infect red blood cells and cause malaria, Plasmodium parasites undergo an obligate and clinically silent expansion phase in the liver that is supposedly undetected by the host. Here, we demonstrate the engagement of a type I interferon (IFN) response during Plasmodium replication in the liver. We identified Plasmodium RNA as a previously unrecognized pathogen-associated molecular pattern (PAMP) capable of activating a type I IFN response via the cytosolic pattern recognition receptor Mda5. This response, initiated by liver-resident cells through the adaptor molecule for cytosolic RNA sensors, Mavs, and the transcription factors Irf3 and Irf7, is propagated by hepatocytes in an interferon-α/ß receptor-dependent manner. This signaling pathway is critical for immune cell-mediated host resistance to liver-stage Plasmodium infection, which we find can be primed with other PAMPs, including hepatitis C virus RNA. Together, our results show that the liver has sensor mechanisms for Plasmodium that mediate a functional antiparasite response driven by type I IFN.


Assuntos
Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Fígado/parasitologia , Plasmodium/imunologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Western Blotting , RNA Helicases DEAD-box/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Fígado/imunologia , Luciferases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise em Microsséries , Oligonucleotídeos/genética , Plasmodium/genética , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas
11.
Mar Drugs ; 11(10): 4019-34, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24152562

RESUMO

Terrestrial plants have proven to be a prolific producer of clinically effective antimalarial drugs, but the antimalarial potential of seaweeds has been little explored. The main aim of this study was to assess the in vitro chemotherapeutical and prophylactic potential of the extracts of twenty-three seaweeds collected from the south coast of England against blood stage (BS) and liver stage (LS) Plasmodium parasites. The majority (14) of the extracts were active against BS of P. falciparum, with brown seaweeds Cystoseira tamariscifolia, C. baccata and the green seaweed Ulva lactuca being the most active (IC(50)s around 3 µg/mL). The extracts generally had high selectivity indices (>10). Eight seaweed extracts inhibited the growth of LS parasites of P. berghei without any obvious effect on the viability of the human hepatoma (Huh7) cells, and the highest potential was exerted by U. lactuca and red seaweeds Ceramium virgatum and Halopitys incurvus (IC50 values 14.9 to 28.8 µg/mL). The LS-active extracts inhibited one or more key enzymes of the malarial type-II fatty acid biosynthesis (FAS-II) pathway, a drug target specific for LS. Except for the red seaweed Halopitys incurvus, all LS-active extracts showed dual activity versus both malarial intracellular stage parasites. This is the first report of LS antiplasmodial activity and dual stage inhibitory potential of seaweeds.


Assuntos
Antimaláricos/farmacologia , Fígado/efeitos dos fármacos , Phaeophyceae/química , Extratos Vegetais/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Alga Marinha/química , Antimaláricos/química , Carcinoma Hepatocelular/parasitologia , Linhagem Celular Tumoral , Humanos , Fígado/parasitologia
12.
J Med Chem ; 56(19): 7679-90, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24020770

RESUMO

Discovery of novel effective and safe antimalarials has been traditionally focused on targeting erythrocytic parasite stages that cause clinical symptoms. However, elimination of malaria parasites from the human population will be facilitated by intervention at different life-cycle stages of the parasite, including the obligatory developmental phase in the liver, which precedes the erythrocytic stage. We have previously reported that N-Mannich-based quinolon-4(1H)-imines are potent antiplasmodial agents but present several stability liabilities. We now report our efforts to optimize quinolon-4(1H)-imines as dual-stage antiplasmodial agents endowed with chemical and metabolic stability. We report compounds active against both the erythrocytic and exoerythrocytic forms of malaria parasites, such as the quinolon-4(1H)-imine 5p (IC50 values of 54 and 710 nM against the erythrocytic and exoerythrocytic forms), which constitute excellent starting points for further lead optimization as dual-stage antimalarials.


Assuntos
Antimaláricos/síntese química , Iminas/síntese química , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Quinolonas/síntese química , Animais , Animais Geneticamente Modificados , Antimaláricos/química , Antimaláricos/farmacologia , Linhagem Celular Tumoral , Cristalização , Complexo III da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Células HEK293 , Hemeproteínas/química , Humanos , Iminas/química , Iminas/farmacologia , Estágios do Ciclo de Vida/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Quinolonas/química , Quinolonas/farmacologia , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
13.
J Med Chem ; 56(11): 4811-5, 2013 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-23701465

RESUMO

We present a novel series of quinolin-4(1H)-imines as dual-stage antiplasmodials, several-fold more active than primaquine in vitro against Plasmodium berghei liver stage. Among those, compounds 5g and 5k presented low nanomolar IC50 values. The compounds are metabolically stable and modulate several drug targets. These results emphasize the value of quinolin-4(1H)-imines as a new chemotype and their suitable properties for further drug development.


Assuntos
Antimaláricos/síntese química , Iminas/síntese química , Fígado/efeitos dos fármacos , Malária/tratamento farmacológico , Quinolinas/síntese química , Antimaláricos/química , Antimaláricos/farmacologia , Linhagem Celular Tumoral , Humanos , Iminas/química , Iminas/farmacologia , Fígado/parasitologia , Testes de Sensibilidade Parasitária , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Quinolinas/química , Quinolinas/farmacologia , Relação Estrutura-Atividade
14.
Methods Mol Biol ; 923: 385-400, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22990793

RESUMO

There is an important role for in vitro assays to better understand the initial steps of malaria infection. In this section, we describe both microscopy-based and flow cytometry-based sporozoite invasion, migration and development assays with the rodent malaria parasites, Plasmodium berghei and Plasmodium yoelii, and the human malaria parasite, Plasmodium falciparum.


Assuntos
Movimento Celular , Citometria de Fluxo/métodos , Hepatócitos/parasitologia , Microscopia/métodos , Plasmodium/crescimento & desenvolvimento , Esporozoítos/metabolismo , Animais , Anopheles/parasitologia , Técnicas de Cultura de Células , Linhagem Celular , Células Hep G2 , Hepatócitos/patologia , Humanos , Malária/parasitologia , Camundongos
15.
Cell Immunol ; 277(1-2): 22-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22784562

RESUMO

Although the role of regulatory T cells (Tregs) during malaria infection has been studied extensively, such studies have focused exclusively on the role of Treg during the blood stage of infection; little is known about the detailed mechanisms of Tregs and sporozoite deposition in the dermis by mosquito bites. In this paper we show that sporozoites introduced into the skin by mosquito bites increase the mobility of skin Tregs and dendritic cells (DCs). We also show differences in MHC class II and/or CD86 expression on skin-resident dendritic cell subtypes and macrophages. From the observed decrease of the number of APCs into draining lymph nodes, suppression of CD28 expression in conventional CD4 T cells, and a low homeostatic proliferation of skin-migrated CD4 T found in nude mice indicate that Tregs may play a fundamental role during the initial phase of malaria parasite inoculation into the mammalian host.


Assuntos
Mordeduras e Picadas/imunologia , Culicidae/parasitologia , Malária/imunologia , Dermatopatias Parasitárias/imunologia , Pele/imunologia , Animais , Antígeno B7-2/biossíntese , Antígeno B7-2/imunologia , Mordeduras e Picadas/parasitologia , Proliferação de Células , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Genes MHC da Classe II/imunologia , Linfonodos/imunologia , Linfonodos/parasitologia , Macrófagos/imunologia , Macrófagos/parasitologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Pele/parasitologia , Esporozoítos/imunologia , Esporozoítos/parasitologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/parasitologia
16.
Proc Natl Acad Sci U S A ; 109(22): 8511-6, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22586124

RESUMO

Human malaria infection begins with a one-time asymptomatic liver stage followed by a cyclic symptomatic blood stage. All high-throughput malaria drug discovery efforts have focused on the cyclic blood stage, which has limited potential for the prophylaxis, transmission blocking, and eradication efforts that will be needed in the future. To address these unmet needs, a high-throughput phenotypic liver-stage Plasmodium parasite screen was developed to systematically identify molecules with liver-stage efficacy. The screen recapitulates liver-stage infection by isolating luciferase-expressing Plasmodium berghei parasites directly from the salivary glands of infected mosquitoes, adding them to confluent human liver cells in 384-well plates, and measuring luciferase activity after a suitable incubation period. Screening 5,375 known bioactive compounds identified 37 liver-stage malaria inhibitors with diverse modes of action, as shown by inhibition time course experiments. Further analysis of the hits in the Food and Drug Administration-approved drug subset revealed compounds that seem to act specifically on the liver stage of infection, suggesting that this phase of the parasite's life cycle presents a promising area for new drug discovery. Notably, many active compounds in this screen have molecular structures and putative targets distinctly different from those of known antimalarial agents.


Assuntos
Antimaláricos/farmacologia , Fígado/efeitos dos fármacos , Malária/prevenção & controle , Plasmodium berghei/efeitos dos fármacos , Animais , Anopheles/parasitologia , Antimaláricos/classificação , Avaliação Pré-Clínica de Medicamentos , Células Hep G2 , Humanos , Concentração Inibidora 50 , Insetos Vetores/parasitologia , Estágios do Ciclo de Vida , Fígado/parasitologia , Fígado/patologia , Malária/parasitologia , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/isolamento & purificação , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Resultado do Tratamento
17.
J Infect Dis ; 205(8): 1278-86, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22396598

RESUMO

Plasmodium parasites undergo a clinically silent and obligatory developmental phase in the host's liver cells before they are able to infect erythrocytes and cause malaria symptoms. To overcome the scarcity of compounds targeting the liver stage of malaria, we screened a library of 1037 existing drugs for their ability to inhibit Plasmodium hepatic development. Decoquinate emerged as the strongest inhibitor of Plasmodium liver stages, both in vitro and in vivo. Furthermore, decoquinate kills the parasite's replicative blood stages and is active against developing gametocytes, the forms responsible for transmission. The drug acts by selectively and specifically inhibiting the parasite's mitochondrial bc(1) complex, with little cross-resistance with the antimalarial drug atovaquone. Oral administration of a single dose of decoquinate effectively prevents the appearance of disease, warranting its exploitation as a potent antimalarial compound.


Assuntos
Antimaláricos/farmacologia , Hepatócitos/parasitologia , Malária/tratamento farmacológico , Malária/parasitologia , Plasmodium/efeitos dos fármacos , Animais , Atovaquona/farmacologia , Linhagem Celular Tumoral , Decoquinato/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Moleculares , Estrutura Molecular , Conformação Proteica
18.
PLoS One ; 7(1): e29408, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22238609

RESUMO

Plasmodium sporozoites are transmitted by Anopheles mosquitoes and infect hepatocytes, where a single sporozoite replicates into thousands of merozoites inside a parasitophorous vacuole. The nature of the Plasmodium-host cell interface, as well as the interactions occurring between these two organisms, remains largely unknown. Here we show that highly dynamic hepatocyte actin reorganization events occur around developing Plasmodium berghei parasites inside human hepatoma cells. Actin reorganization is most prominent between 10 to 16 hours post infection and depends on the actin severing and capping protein, gelsolin. Live cell imaging studies also suggest that the hepatocyte cytoskeleton may contribute to parasite elimination during Plasmodium development in the liver.


Assuntos
Actinas/metabolismo , Hepatócitos/parasitologia , Plasmodium/metabolismo , Multimerização Proteica/fisiologia , Citoesqueleto de Actina/metabolismo , Animais , Células Cultivadas , Gelsolina/metabolismo , Gelsolina/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hepatócitos/metabolismo , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/fisiologia , Humanos , Cinética , Fígado/metabolismo , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Organismos Geneticamente Modificados , Plasmodium/genética , Plasmodium/fisiologia , Tubulina (Proteína)/metabolismo
19.
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
20.
EMBO Rep ; 12(12): 1233-42, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22081142

RESUMO

After the bite of a malaria-infected mosquito, the Plasmodium sporozoite infects liver cells and produces thousands of merozoites, which then infect red blood cells, causing malaria. In malaria-endemic areas, several hundred infected mosquitoes can bite an individual each year, increasing the risk of superinfection. However, in infants that are yet to acquire immunity, superinfections are infrequent. We have recently shown that blood-stage parasitaemia, above a minimum threshold, impairs the growth of a subsequent sporozoite infection of liver cells. Blood-stage parasites stimulate the production of the host iron-regulatory factor hepcidin, which redistributes iron away from hepatocytes, reducing the development of the iron-dependent liver stage. This could explain why Plasmodium superinfection is not often found in young nonimmune children. Here, we discuss the impact that such protection from superinfection might have in epidemiological settings or in programmes for controlling malaria, as well as how the induction of hepcidin and redistribution of iron might influence anaemia and the outcome of non-Plasmodium co-infections.


Assuntos
Ferro/metabolismo , Malária/complicações , Malária/parasitologia , Plasmodium/fisiologia , Superinfecção/complicações , Superinfecção/parasitologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade , Malária/prevenção & controle , Plasmodium/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA