Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PLoS Pathog ; 20(2): e1012008, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38354186

RESUMO

Leucine-rich repeat (LRR) proteins are commonly involved in innate immunity of animals and plants, including for pattern recognition of pathogen-derived elicitors. The Anopheles secreted LRR proteins APL1C and LRIM1 are required for malaria ookinete killing in conjunction with the complement-like TEP1 protein. However, the mechanism of parasite immune recognition by the mosquito remains unclear, although it is known that TEP1 lacks inherent binding specificity. Here, we find that APL1C and LRIM1 bind specifically to Plasmodium berghei ookinetes, even after depletion of TEP1 transcript and protein, consistent with a role for the LRR proteins in pathogen recognition. Moreover, APL1C does not bind to ookinetes of the human malaria parasite Plasmodium falciparum, and is not required for killing of this parasite, which correlates LRR binding specificity and immune protection. Most of the live P. berghei ookinetes that migrated into the extracellular space exposed to mosquito hemolymph, and almost all dead ookinetes, are bound by APL1C, thus associating LRR protein binding with parasite killing. We also find that APL1C binds to the surface of P. berghei sporozoites released from oocysts into the mosquito hemocoel and forms a potent barrier limiting salivary gland invasion and mosquito infectivity. Pathogen binding by APL1C provides the first functional explanation for the long-known requirement of APL1C for P. berghei ookinete killing in the mosquito midgut. We propose that secreted mosquito LRR proteins are required for pathogen discrimination and orientation of immune effector activity, potentially as functional counterparts of the immunoglobulin-based receptors used by vertebrates for antigen recognition.


Assuntos
Anopheles , Malária , Animais , Humanos , Proteínas de Repetições Ricas em Leucina , Anopheles/parasitologia , Esporozoítos/metabolismo , Proteínas/metabolismo , Plasmodium berghei/metabolismo
2.
Microbes Infect ; 25(5): 105102, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36708871

RESUMO

To persist in the blood circulation and to be available for mosquitoes, Plasmodium falciparum gametocytes modify the deformability and the permeability of their erythrocyte host via cyclic AMP (cAMP) signaling pathway. Cyclic nucleotide levels are tightly controlled by phosphodiesterases (PDE), however in Plasmodium these proteins are poorly characterized. Here, we characterize the P. falciparum phosphodiesterase delta (PfPDEδ) and we investigate its role in the cAMP signaling-mediated regulation of gametocyte-infected erythrocyte mechanical properties. Our results revealed that PfPDEδ is a dual-function enzyme capable of hydrolyzing both cAMP and cGMP, with a higher affinity for cAMP. We also show that PfPDEδ is the most expressed PDE in mature gametocytes and we propose that it is located in parasitophorous vacuole at the interface between the host cell and the parasite. We conclude that PfPDEδ is the master regulator of both the increase in deformability and the inhibition of channel activity in mature gametocyte stages, and may therefore play a crucial role in the persistence of mature gametocytes in the bloodstream.


Assuntos
Malária Falciparum , Plasmodium falciparum , Animais , Plasmodium falciparum/fisiologia , Diester Fosfórico Hidrolases , Malária Falciparum/parasitologia , Eritrócitos/parasitologia , Transdução de Sinais
3.
Int J Mol Sci ; 23(21)2022 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-36361552

RESUMO

One of the major pathophysiologies of malaria is the development of anemia. Although hemolysis and splenic clearance are well described as causes of malarial anemia, abnormal erythropoiesis has been observed in malaria patients and may contribute significantly to anemia. The interaction between inadequate erythropoiesis and Plasmodium parasite infection, which partly occurs in the bone marrow, has been poorly investigated to date. However, recent findings may provide new insights. This review outlines clinical and experimental studies describing different aspects of ineffective erythropoiesis and dyserythropoiesis observed in malaria patients and in animal or in vitro models. We also highlight the various human and parasite factors leading to erythropoiesis disorders and discuss the impact that Plasmodium parasites may have on the suppression of erythropoiesis.


Assuntos
Anemia , Malária , Plasmodium , Animais , Humanos , Eritropoese/fisiologia , Malária/complicações , Malária/parasitologia , Anemia/etiologia , Medula Óssea
5.
Acta Pharm Sin B ; 12(4): 2089-2102, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35847505

RESUMO

Red blood cells (RBCs) can act as carriers for therapeutic agents and can substantially improve the safety, pharmacokinetics, and pharmacodynamics of many drugs. Maintaining RBCs integrity and lifespan is important for the efficacy of RBCs as drug carrier. We investigated the impact of drug encapsulation by hypotonic dialysis on RBCs physiology and integrity. Several parameters were compared between processed RBCs loaded with l-asparaginase ("eryaspase"), processed RBCs without drug and non-processed RBCs. Processed RBCs were less hydrated and displayed a reduction of intracellular content. We observed a change in the metabolomic but not in the proteomic profile of processed RBCs. Encapsulation process caused moderate morphological changes and was accompanied by an increase of RBCs-derived Extracellular Vesicles release. Despite a decrease in deformability, processed RBCs were not mechanically retained in a spleen-mimicking device and had increased surface-to-volume ratio and osmotic resistance. Processed RBCs half-life was not significantly affected in a mouse model and our previous phase 1 clinical study showed that encapsulation of asparaginase in RBCs prolonged its in vivo half-life compared to free forms. Our study demonstrated that encapsulation by hypotonic dialysis may affect certain characteristics of RBCs but does not significantly affect the in vivo longevity of RBCs or their drug carrier function.

6.
Front Cell Infect Microbiol ; 12: 883759, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35694548

RESUMO

The persistence of erythrocytes infected with Plasmodium falciparum gametocytes in the bloodstream is closely related to the modulation of their mechanical properties. New drugs that increase the stiffness of infected erythrocytes may thus represent a novel approach to block malaria parasite transmission. The phosphodiesterase inhibitor tadalafil has been shown to impair the ability of infected erythrocytes to circulate in an in vitro model for splenic retention. Here, we used a humanized mouse model to address in vivo the effect of tadalafil on the circulation kinetics of mature gametocyte-infected erythrocytes. We show that stiff immature gametocyte-infected erythrocytes are retained in the spleen of humanized mice at rates comparable to that of the in vitro model. Accordingly, tadalafil-induced stiffening of mature gametocyte-infected erythrocytes impairs their circulation in the bloodstream and triggers their retention by the spleen. These in vivo results validate that tadalafil is a novel drug lead potentially capable of blocking malaria parasite transmission by targeting GIE mechanical properties.


Assuntos
Malária Falciparum , Plasmodium falciparum , Animais , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Camundongos , Inibidores de Fosfodiesterase , Baço , Tadalafila/farmacologia
7.
Pharmaceutics ; 14(2)2022 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-35214104

RESUMO

The alkaloid tazopsine 1 was introduced in the late 2000s as a novel antiplasmodial hit compound active against Plasmodium falciparum hepatic stages, with the potential to develop prophylactic drugs based on this novel chemical scaffold. However, the structural determinants of tazopsine 1 bioactivity, together with the exact definition of the pharmacophore, remained elusive, impeding further development. We found that the antitussive drug dextromethorphan (DXM) 3, although lacking the complex pattern of stereospecific functionalization of the natural hit, was harboring significant antiplasmodial activity in vitro despite suboptimal prophylactic activity in a murine model of malaria, precluding its direct repurposing against the disease. The targeted N-alkylation of nor-DXM 15 produced a small library of analogues with greatly improved activity over DXM 3 against P. falciparum asexual stages. Amongst these, N-2'-pyrrolylmethyl-nor-DXM 16i showed a 2- to 36-fold superior inhibitory potency compared to tazopsine 1 and DXM 3 against P. falciparum liver and blood stages, with respectively 760 ± 130 nM and 2.1 ± 0.4 µM IC50 values, as well as liver/blood phase selectivity of 2.8. Furthermore, cpd. 16i showed a 5- to 8-fold increase in activity relative to DXM 3 against P. falciparum stages I-II and V gametocytes, with 18.5 µM and 13.2 µM IC50 values, respectively. Cpd. 16i can thus be considered a promising novel hit compound against malaria in the ent-morphinan series with putative pan cycle activity, paving the way for further therapeutic development (e.g., investigation of its prophylactic activity in vivo).

8.
Microbiol Spectr ; 9(2): e0027421, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34724729

RESUMO

Human malaria infection begins with a one-time asymptomatic liver stage followed by a cyclic symptomatic blood stage. For decades, the research for novel antimalarials focused on the high-throughput screening of molecules that only targeted the asexual blood stages. In a search for new effective compounds presenting a triple action against erythrocytic and liver stages in addition to the ability to block the transmission of the disease via the mosquito vector, 2-amino-thienopyrimidinone derivatives were synthesized and tested for their antimalarial activity. One molecule, named gamhepathiopine (denoted as "M1" herein), was active at submicromolar concentrations against both erythrocytic (50% effective concentration [EC50] = 0.045 µM) and liver (EC50 = 0.45 µM) forms of Plasmodium falciparum. Furthermore, gamhepathiopine efficiently blocked the development of the sporogonic cycle in the mosquito vector by inhibiting the exflagellation step. Moreover, M1 was active against artemisinin-resistant forms (EC50 = 0.227 µM), especially at the quiescent stage. Nevertheless, in mice, M1 showed modest activity due to its rapid metabolization by P450 cytochromes into inactive derivatives, calling for the development of new parent compounds with improved metabolic stability and longer half-lives. These results highlight the thienopyrimidinone scaffold as a novel antiplasmodial chemotype of great interest to search for new drug candidates displaying multistage activity and an original mechanism of action with the potential to be used in combination therapies for malaria elimination in the context of artemisinin resistance. IMPORTANCE This work reports a new chemical structure that (i) displays activity against the human malaria parasite Plasmodium falciparum at 3 stages of the parasitic cycle (blood stage, hepatic stage, and sexual stages), (ii) remains active against parasites that are resistant to the first-line treatment recommended by the World Health Organization (WHO) for the treatment of severe malaria (artemisinins), and (iii) reduces transmission of the parasite to the mosquito vector in a mouse model. This new molecule family could open the way to the conception of novel antimalarial drugs with an original multistage mechanism of action to fight against Plasmodium drug resistance and block interhuman transmission of malaria.


Assuntos
Antimaláricos/farmacologia , Malária Falciparum/tratamento farmacológico , Plasmodium cynomolgi/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Plasmodium yoelii/efeitos dos fármacos , Pirimidinonas/farmacologia , Animais , Antimaláricos/química , Artemisininas/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Cães , Resistência a Medicamentos/fisiologia , Feminino , Células Hep G2 , Humanos , Fígado/parasitologia , Macaca fascicularis , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pirimidinonas/química
9.
mBio ; 12(4): e0163621, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34340541

RESUMO

Clonally variant genes (CVGs) play fundamental roles in the adaptation of Plasmodium falciparum to fluctuating conditions of the human host. However, their expression patterns under the natural conditions of the blood circulation have been characterized in detail for only a few specific gene families. Here, we provide a detailed characterization of the complete P. falciparum transcriptome across the full intraerythrocytic development cycle (IDC) at the onset of a blood infection in malaria-naive human volunteers. We found that the vast majority of transcriptional differences between parasites obtained from the volunteers and the parental parasite line maintained in culture occurred in CVGs. In particular, we observed a major increase in the transcript levels of most genes of the pfmc-2tm and gbp families and of specific genes of other families, such as phist, hyp10, rif, or stevor, in addition to previously reported changes in var and clag3 gene expression. Increased transcript levels of individual pfmc-2tm, rif, and stevor genes involved activation in small subsets of parasites. Large transcriptional differences correlated with changes in the distribution of heterochromatin, confirming their epigenetic nature. Furthermore, the similar expression of several CVGs between parasites collected at different time points along the blood infection suggests that the epigenetic memory for multiple CVG families is lost during transmission stages, resulting in a reset of their transcriptional state. Finally, the CVG expression patterns observed in a volunteer likely infected by a single sporozoite suggest that new epigenetic patterns are established during liver stages. IMPORTANCE The ability of malaria parasites to adapt to changes in the human blood environment, where they produce long-term infection associated with clinical symptoms, is fundamental for their survival. CVGs, regulated at the epigenetic level, play a major role in this adaptive process, as changes in the expression of these genes result in alterations in the antigenic and functional properties of the parasites. However, how these genes are expressed under the natural conditions of the human circulation and how their expression is affected by passage through transmission stages are not well understood. Here, we provide a comprehensive characterization of the expression patterns of these genes at the onset of human blood infections, which reveals major differences with in vitro-cultured parasites. We also show that, during transmission stages, the previous expression patterns for many CVG families are lost, and new patterns are established.


Assuntos
Perfilação da Expressão Gênica , Variação Genética , Interações Hospedeiro-Parasita/genética , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Antígenos de Protozoários/imunologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Transcriptoma
10.
Mol Biochem Parasitol ; 244: 111392, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34171456

RESUMO

Plasmodium falciparum gametocytes modify the mechanical properties of their erythrocyte host to persist for several weeks in the blood circulation and to be available for mosquitoes. These changes are tightly regulated by the plasmodial phosphodiesterase delta that decreases both the stiffness and the permeability of the infected host cell. Here, we address the effect of the phosphodiesterase inhibitor tadalafil on deformability and permeability of gametocyte-infected erythrocytes. We show that this inhibitor drastically increases isosmotic lysis of gametocyte-infected erythrocytes and impairs their ability to circulate in an in vitro model for splenic retention. These findings indicate that tadalafil represents a novel drug lead potentially capable of blocking malaria parasite transmission by impacting gametocyte circulation.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Gametogênese/efeitos dos fármacos , Estágios do Ciclo de Vida/efeitos dos fármacos , Inibidores da Fosfodiesterase 5/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/antagonistas & inibidores , Tadalafila/farmacologia , Fenômenos Biomecânicos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Deformação Eritrocítica/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Eritrócitos/ultraestrutura , Feminino , Expressão Gênica , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/genética , Humanos , Estágios do Ciclo de Vida/genética , Masculino , Plasmodium falciparum/enzimologia , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Reprodução Assexuada/efeitos dos fármacos
11.
Curr Opin Hematol ; 28(3): 158-163, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33631784

RESUMO

PURPOSE OF REVIEW: The current review outlines recent discoveries on the infection of erythroid cells by Plasmodium parasites, focusing on the molecular interactions governing the tropism of parasites for their host cell and the implications of this tropism for parasite biology and erythroid cell maturation. RECENT FINDINGS: Although most studies about the interactions of Plasmodium parasites and their host cell focused on the deadliest human malaria parasite, Plasmodium falciparum, and the erythrocyte, there is increasing evidence that several Plasmodium species, including P. falciparum, also develop within erythroid precursors. These interactions likely modify the remodeling of the host cell by the parasite and affect the maturation of erythroblast and reticulocytes. SUMMARY: A better understanding of the remodeling of immature erythroid cells by Plasmodium parasites will have important implications for the development of antimalarial drugs or vaccines. In addition, deciphering how Plasmodium parasites interfere with erythropoiesis will provide new insights on how these parasites contribute to anemia in malaria patients.


Assuntos
Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Malária/sangue , Malária/parasitologia , Plasmodium/fisiologia , Animais , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Diferenciação Celular , Eritroblastos/metabolismo , Eritroblastos/parasitologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/imunologia , Eritrócitos/metabolismo , Eritropoese , Interações Hospedeiro-Parasita/imunologia , Humanos , Malária/tratamento farmacológico , Malária/imunologia , Vacinas Antimaláricas/imunologia , Reticulócitos/metabolismo , Reticulócitos/parasitologia
12.
Am J Hematol ; 96(4): 480-492, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33476437

RESUMO

Efficient erythropoiesis relies on the expression of the transferrin receptor type 2 (TFR2). In erythroid precursors, TFR2 facilitates the export of the erythropoietin receptor (EPOR) to cell surface, which ensures the survival and proliferation of erythroblasts. Although TFR2 has a crucial role in erythropoiesis regulation, its mechanism of action remains to be clarified. To understand its role better, we aimed at identifying its protein partners by mass-spectrometry after immunoprecipitation in erythroid cells. Here we report the kinase MRCKα (myotonic dystrophy kinase-related CDC42-binding kinase α) as a new partner of both TFR2 and EPOR in erythroblasts. We show that MRCKα is co-expressed with TFR2, and TFR1 during terminal differentiation and regulates the internalization of the two types of transferrin receptors. The knockdown of MRCKα by shRNA in human primary erythroblasts leads to a decreased cell surface expression of both TFR1 and TFR2, an increased cell-surface expression of EPOR, and a delayed differentiation. Additionally, knockout of Mrckα in the murine MEDEP cells also leads to a striking delay in erythropoiesis, showcasing the importance of this kinase in both species. Our data highlight the importance of MRCKα in the regulation of erythropoiesis.


Assuntos
Eritropoese/fisiologia , Miotonina Proteína Quinase/fisiologia , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Endocitose , Eritroblastos/citologia , Eritroblastos/metabolismo , Técnicas de Inativação de Genes , Humanos , Ferro/metabolismo , Camundongos , Miotonina Proteína Quinase/isolamento & purificação , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptores da Eritropoetina/metabolismo , Receptores da Transferrina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo
13.
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
14.
Nat Med ; 26(12): 1929-1940, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33106664

RESUMO

The dry season is a major challenge for Plasmodium falciparum parasites in many malaria endemic regions, where water availability limits mosquito vectors to only part of the year. How P. falciparum bridges two transmission seasons months apart, without being cleared by the human host or compromising host survival, is poorly understood. Here we show that low levels of P. falciparum parasites persist in the blood of asymptomatic Malian individuals during the 5- to 6-month dry season, rarely causing symptoms and minimally affecting the host immune response. Parasites isolated during the dry season are transcriptionally distinct from those of individuals with febrile malaria in the transmission season, coinciding with longer circulation within each replicative cycle of parasitized erythrocytes without adhering to the vascular endothelium. Low parasite levels during the dry season are not due to impaired replication but rather to increased splenic clearance of longer-circulating infected erythrocytes, which likely maintain parasitemias below clinical and immunological radar. We propose that P. falciparum virulence in areas of seasonal malaria transmission is regulated so that the parasite decreases its endothelial binding capacity, allowing increased splenic clearance and enabling several months of subclinical parasite persistence.


Assuntos
Infecções Assintomáticas/epidemiologia , Interações Hospedeiro-Parasita/genética , Malária Falciparum/epidemiologia , Plasmodium falciparum/patogenicidade , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Doenças Endêmicas/prevenção & controle , Eritrócitos/parasitologia , Feminino , Genótipo , Humanos , Lactente , Malária Falciparum/genética , Malária Falciparum/parasitologia , Masculino , Mali/epidemiologia , Pessoa de Meia-Idade , Plasmodium falciparum/genética , Estações do Ano , Adulto Jovem
15.
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
16.
ACS Infect Dis ; 6(7): 1532-1547, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32267151

RESUMO

Three novel tracers designed as fluorescent surrogates of artemisinin-derived antimalarial drugs (i.e., dihydroartemisinin, artemether, arteether, and artemisone) were synthesized from dihydroartemisinin. One of these tracers, corresponding to a dihydroartemisinin/artemether/arteether mimic, showed a combination of excellent physicochemical and biological properties such as hydrolytic stability, high inhibitory potency against blood-stage parasites, similar ring-stage survival assay values than the clinical antimalarials, high cytopermeability and specific labeling of live P. falciparum cells, alkylation of heme, as well as specific covalent labeling of drug-sensitive and drug-resistant P. falciparum proteomes at physiological concentrations, consistent with a multitarget action of the drugs. Our study demonstrates that probes containing the complete structural core of clinical artemisinin derivatives can be stable in biochemical and cellular settings, and recapitulate the complex mechanisms of these frontline, yet threatened, antimalarial drugs.


Assuntos
Antimaláricos , Artemisininas , Antimaláricos/farmacologia , Artemeter , Artemisininas/farmacologia
17.
Front Microbiol ; 10: 2652, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31787966

RESUMO

Plasmodium falciparum sexual parasites, called gametocytes, are the only parasite stages responsible for transmission from humans to Anopheles mosquitoes. During their maturation, P. falciparum gametocytes remodel the structural and mechanical properties of the membrane of their erythrocyte host. This remodeling is induced by the export of several parasite proteins and a dynamic reorganization of the erythrocyte cytoskeleton. Some of these modifications are specific for sexual stages and play a key role for gametocyte maturation, sequestration in internal organs, subsequent release in the bloodstream and ability to persist in circulation. Here we discuss the mechanisms developed by gametocytes to remodel their host cell and the functional relevance of these modifications.

18.
mBio ; 10(4)2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31363031

RESUMO

During its intraerythrocytic development, the malaria parasite Plasmodium falciparum exposes variant surface antigens (VSAs) on infected erythrocytes to establish and maintain an infection. One family of small VSAs is the polymorphic STEVOR proteins, which are marked for export to the host cell surface through their PEXEL signal peptide. Interestingly, some STEVORs have also been reported to localize to the parasite plasma membrane and apical organelles, pointing toward a putative function in host cell egress or invasion. Using deep RNA sequencing analysis, we characterized P. falciparumstevor gene expression across the intraerythrocytic development cycle, including free merozoites, in detail and used the resulting stevor expression profiles for hierarchical clustering. We found that most stevor genes show biphasic expression oscillation, with maximum expression during trophozoite stages and a second peak in late schizonts. We selected four STEVOR variants, confirmed the expected export of these proteins to the host cell membrane, and tracked them to a secondary location, either to the parasite plasma membrane or the secretory organelles of merozoites in late schizont stages. We investigated the function of a particular STEVOR that showed rhoptry localization and demonstrated its role at the parasite-host interface during host cell invasion by specific antisera and targeted gene disruption. Experimentally determined membrane topology of this STEVOR revealed a single transmembrane domain exposing the semiconserved as well as variable protein regions to the cell surface.IMPORTANCE Malaria claims about half a million lives each year. Plasmodium falciparum, the causative agent of the most severe form of the disease, uses proteins that are translocated to the surface of infected erythrocytes for immune evasion. To circumvent the detection of these gene products by the immune system, the parasite evolved a complex strategy that includes gene duplications and elaborate sequence polymorphism. STEVORs are one family of these variant surface antigens and are encoded by about 40 genes. Using deep RNA sequencing of blood-stage parasites, including free merozoites, we first established stevor expression of the cultured isolate and compared it with published transcriptomes. We reveal a biphasic expression of most stevor genes and confirm this for individual STEVORs at the protein level. The membrane topology of a rhoptry-associated variant was experimentally elucidated and linked to host cell invasion, underlining the importance of this multifunctional protein family for parasite proliferation.


Assuntos
Plasmodium falciparum/patogenicidade , Animais , Antígenos de Protozoários/imunologia , Eritrócitos/imunologia , Humanos , Malária/imunologia , Malária/parasitologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
19.
Sci Rep ; 8(1): 17886, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30552367

RESUMO

Plasmodium falciparum gametocytes, the sexual stages responsible for malaria parasite transmission, develop in the human bone marrow parenchyma in proximity to the erythroblastic islands. Yet, mechanisms underlying gametocytes interactions with these islands are unknown. Here, we have investigated whether gametocyte-infected erythrocytes (GIE) adhere to erythroid precursors, and whether a putative adhesion may be mediated by a mechanism similar to the adhesion of erythrocytes infected with P. falciparum asexual stages to uninfected erythrocytes. Cell-cell adhesion assays with human primary erythroblasts or erythroid cell lines revealed that immature GIE do not specifically adhere to erythroid precursors. To determine whether adhesion may be dependent on binding of STEVOR proteins to Glycophorin C on the surface of erythroid cells, we used clonal lines and transgenic parasites that overexpress specific STEVOR proteins known to bind to Glycophorin C in asexual stages. Our results indicate that GIE overexpressing STEVOR do not specifically adhere to erythroblasts, in agreement with our observation that the STEVOR adhesive domain is not exposed at the surface of GIE.


Assuntos
Adesão Celular , Eritroblastos/fisiologia , Eritrócitos/fisiologia , Eritrócitos/parasitologia , Malária Falciparum/patologia , Plasmodium falciparum/crescimento & desenvolvimento , Antígenos de Protozoários/genética , Antígenos de Protozoários/metabolismo , Células Cultivadas , Humanos , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo
20.
Expert Opin Drug Discov ; 13(2): 131-140, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29183148

RESUMO

INTRODUCTION: Efforts on malaria drug discovery are expected to increase in the coming years to achieve malaria eradication. Owing to the increasing number of new potential candidates together with the actual limitations of the primate models, humanized mouse models infected with human Plasmodium spp. (HmHP) now appear as an alternative to the primate model. Areas covered: The authors review the progress obtained in the HmHP in the last two decades, with a special emphasis of their input on the drug discovery pathway. The authors discuss the methodologies and strategies used in these models to obtain an accurate assessment of the compound activity and a reliable prediction of the human efficacious regimen. Expert opinion: Research efforts have led us to an era in which HmHP can successfully be infected with P. falciparum, P vivax and P. ovale. Furthermore, it is now a reality that the complete human cycle of P. falciparum can be obtained in HmHP. The HmHP has shown a real input mainly in the preclinical evaluation of new compounds against the erythrocytic stages of P. falciparum. However, further technical improvements are needed before HmHP may replace the primate model.


Assuntos
Antimaláricos/farmacologia , Descoberta de Drogas/métodos , Malária/tratamento farmacológico , Animais , Modelos Animais de Doenças , Desenho de Fármacos , Humanos , Malária/parasitologia , Camundongos , Camundongos Transgênicos , Plasmodium/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA