Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 3(1): 726, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33262483

RESUMO

To ensure the transport of nutrients necessary for their survival, Plasmodium falciparum parasites increase erythrocyte permeability to diverse solutes. These new permeation pathways (NPPs) have been extensively characterized in the pathogenic asexual parasite stages, however the existence of NPPs has never been investigated in gametocytes, the sexual stages responsible for transmission to mosquitoes. Here, we show that NPPs are still active in erythrocytes infected with immature gametocytes and that this activity declines along gametocyte maturation. Our results indicate that NPPs are regulated by cyclic AMP (cAMP) signaling cascade, and that the decrease in cAMP levels in mature stages results in a slowdown of NPP activity. We also show that NPPs facilitate the uptake of artemisinin derivatives and that phosphodiesterase (PDE) inhibitors can reactivate NPPs and increase drug uptake in mature gametocytes. These processes are predicted to play a key role in P. falciparum gametocyte biology and susceptibility to antimalarials.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Estágios do Ciclo de Vida/fisiologia , Plasmodium falciparum/patogenicidade , Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Células Cultivadas , AMP Cíclico/metabolismo , Humanos , Inibidores de Fosfodiesterase , Transdução de Sinais/fisiologia
2.
Blood ; 136(12): 1381-1393, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32589714

RESUMO

Plasmodium falciparum gametocytes, the sexual stage responsible for malaria parasite transmission from humans to mosquitoes, are key targets for malaria elimination. Immature gametocytes develop in the human bone marrow parenchyma, where they accumulate around erythroblastic islands. Notably though, the interactions between gametocytes and this hematopoietic niche have not been investigated. Here, we identify late erythroblasts as a new host cell for P falciparum sexual stages and show that gametocytes can fully develop inside these nucleated cells in vitro and in vivo, leading to infectious mature gametocytes within reticulocytes. Strikingly, we found that infection of erythroblasts by gametocytes and parasite-derived extracellular vesicles delay erythroid differentiation, thereby allowing gametocyte maturation to coincide with the release of their host cell from the bone marrow. Taken together, our findings highlight new mechanisms that are pivotal for the maintenance of immature gametocytes in the bone marrow and provide further insights on how Plasmodium parasites interfere with erythropoiesis and contribute to anemia in malaria patients.


Assuntos
Eritroblastos/parasitologia , Eritropoese , Interações Hospedeiro-Parasita , Malária Falciparum/fisiopatologia , Plasmodium falciparum/fisiologia , Adulto , Medula Óssea/parasitologia , Medula Óssea/fisiopatologia , Células Cultivadas , Eritroblastos/patologia , Feminino , Humanos , Malária Falciparum/parasitologia , Adulto Jovem
3.
Sci Rep ; 6: 35025, 2016 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-27731362

RESUMO

The development of new drugs to disrupt malaria transmission requires the establishment of an in vivo model to address the biology of Plasmodium falciparum sexual stages (gametocytes). Herein we show that chemically immune-modulated NSG mice grafted with human erythrocytes support complete sexual development of P. falciparum parasites and generate high gametocytemia. Immunohistochemistry and RT-qPCR analyses indicate an enrichment of immature gametocytes in the bone marrow and the spleen, suggesting a sequestration mechanism reminiscent to that observed in humans. Upon primaquine treatment, elimination of gametocytes from peripheral blood and from sequestration sites was observed, providing a proof of concept that these mice can be used for testing drugs. Therefore, this model allows the investigation of P. falciparum sexual commitment, gametocyte interactions with the bone marrow and spleen and provides the missing link between current in vitro assays and Phase I trials in humans for testing new malaria gametocytidal drugs.


Assuntos
Antimaláricos/administração & dosagem , Estágios do Ciclo de Vida/efeitos dos fármacos , Malária Falciparum/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Primaquina/administração & dosagem , Animais , Antimaláricos/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/parasitologia , Modelos Animais de Doenças , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Humanos , Injeções Intraperitoneais , Camundongos , Plasmodium falciparum/efeitos dos fármacos , Primaquina/farmacologia , Baço/efeitos dos fármacos , Baço/parasitologia
4.
Blood ; 127(24): e42-53, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27136945

RESUMO

Deformability of Plasmodium falciparum gametocyte-infected erythrocytes (GIEs) allows them to persist for several days in blood circulation and to ensure transmission to mosquitoes. Here, we investigate the mechanism by which the parasite proteins STEVOR (SubTElomeric Variable Open Reading frame) exert changes on GIE deformability. Using the microsphiltration method, immunoprecipitation, and mass spectrometry, we produce evidence that GIE stiffness is dependent on the cytoplasmic domain of STEVOR that interacts with ankyrin complex at the erythrocyte skeleton. Moreover, we show that GIE deformability is regulated by protein kinase A (PKA)-mediated phosphorylation of the STEVOR C-terminal domain at a specific serine residue (S324). Finally, we show that the increase of GIE stiffness induced by sildenafil (Viagra) is dependent on STEVOR phosphorylation status and on another independent mechanism. These data provide new insights into mechanisms by which phosphodiesterase inhibitors may block malaria parasite transmission.


Assuntos
Antígenos de Protozoários/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Deformação Eritrocítica , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Plasmodium falciparum , Animais , Células Cultivadas , Interações Hospedeiro-Parasita , Humanos , Malária Falciparum/sangue , Fosforilação , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo
5.
PLoS Pathog ; 11(5): e1004815, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25951195

RESUMO

Blocking Plasmodium falciparum transmission to mosquitoes has been designated a strategic objective in the global agenda of malaria elimination. Transmission is ensured by gametocyte-infected erythrocytes (GIE) that sequester in the bone marrow and at maturation are released into peripheral blood from where they are taken up during a mosquito blood meal. Release into the blood circulation is accompanied by an increase in GIE deformability that allows them to pass through the spleen. Here, we used a microsphere matrix to mimic splenic filtration and investigated the role of cAMP-signalling in regulating GIE deformability. We demonstrated that mature GIE deformability is dependent on reduced cAMP-signalling and on increased phosphodiesterase expression in stage V gametocytes, and that parasite cAMP-dependent kinase activity contributes to the stiffness of immature gametocytes. Importantly, pharmacological agents that raise cAMP levels in transmissible stage V gametocytes render them less deformable and hence less likely to circulate through the spleen. Therefore, phosphodiesterase inhibitors that raise cAMP levels in P. falciparum infected erythrocytes, such as sildenafil, represent new candidate drugs to block transmission of malaria parasites.


Assuntos
Deformação Eritrocítica/fisiologia , Eritrócitos/parasitologia , Malária Falciparum/parasitologia , Plasmodium falciparum/fisiologia , Transdução de Sinais , Animais , Culicidae , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Malária Falciparum/transmissão
6.
PLoS One ; 9(8): e104999, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25127364

RESUMO

Peptide nucleic acids (PNAs) are very attractive antisense and antigene agents, but these molecules are not passively taken into cells. Here, using a functional cell assay and fluorescent-based methods, we investigated cell uptake and antisense activity of a tridecamer PNA that targets the HIV-1 polypurine tract sequence delivered using the arginine-rich (R/W)9 peptide (RRWWRRWRR). At micromolar concentrations, without use of any transfection agents, almost 80% inhibition of the target gene expression was obtained with the conjugate in the presence of the endosomolytic agent chloroquine. We show that chloroquine not only induced escape from endosomes but also enhanced the cellular uptake of the conjugate. Mechanistic studies revealed that (R/W)9-PNA conjugates were internalized via pinocytosis. Replacement of arginines with lysines reduced the uptake of the conjugate by six-fold, resulting in the abolition of intracellular target inhibition. Our results show that the arginines play a crucial role in the conjugate uptake and antisense activity. To determine whether specificity of the interactions of arginines with cell surface proteoglycans result in the internalization, we used flow cytometry to examine uptake of arginine- and lysine-rich conjugates in wild-type CHO-K1 and proteoglycan-deficient A745 cells. The uptake of both conjugates was decreased by four fold in CHO-745 cells; therefore proteoglycans promote internalization of cationic peptides, irrespective of the chemical nature of their positive charges. Our results show that arginine-rich cell-penetrating peptides, especially (R/W)9, are a promising tool for PNA internalization.


Assuntos
Marcação de Genes , HIV-1/genética , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/genética , Ácidos Nucleicos Peptídicos/administração & dosagem , Ácidos Nucleicos Peptídicos/genética , Sequência de Aminoácidos , Animais , Arginina/química , Arginina/metabolismo , Sequência de Bases , Células CHO , Linhagem Celular , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Cricetulus , Endossomos/metabolismo , Glicosaminoglicanos/metabolismo , Infecções por HIV/virologia , Células HeLa , Humanos , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/farmacocinética , Ácidos Nucleicos Peptídicos/química , Ácidos Nucleicos Peptídicos/farmacocinética , Peptídeos , Pinocitose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA