Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.166
Filtrar
1.
Life Sci Alliance ; 5(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34857648

RESUMO

Artemisinin-based combination therapies (ACT) are the frontline treatments against malaria worldwide. Recently the use of traditional infusions from Artemisia annua (from which artemisinin is obtained) or Artemisia afra (lacking artemisinin) has been controversially advocated. Such unregulated plant-based remedies are strongly discouraged as they might constitute sub-optimal therapies and promote drug resistance. Here, we conducted the first comparative study of the anti-malarial effects of both plant infusions in vitro against the asexual erythrocytic stages of Plasmodium falciparum and the pre-erythrocytic (i.e., liver) stages of various Plasmodium species. Low concentrations of either infusion accounted for significant inhibitory activities across every parasite species and stage studied. We show that these antiplasmodial effects were essentially artemisinin-independent and were additionally monitored by observations of the parasite apicoplast and mitochondrion. In particular, the infusions significantly incapacitated sporozoites, and for Plasmodium vivax and P. cynomolgi, disrupted the hypnozoites. This provides the first indication that compounds other than 8-aminoquinolines could be effective antimalarials against relapsing parasites. These observations advocate for further screening to uncover urgently needed novel antimalarial lead compounds.


Assuntos
Antimaláricos/farmacologia , Artemisia/química , Artemisininas/farmacologia , Extratos Vegetais/farmacologia , Plasmodium/efeitos dos fármacos , Antimaláricos/química , Artemisininas/química , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/parasitologia , Humanos , Estágios do Ciclo de Vida/efeitos dos fármacos , Malária/tratamento farmacológico , Malária/parasitologia , Testes de Sensibilidade Parasitária , Extratos Vegetais/química , Plasmodium/crescimento & desenvolvimento
2.
PLoS Pathog ; 17(11): e1009770, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34784388

RESUMO

PfSPZ Vaccine against malaria is composed of Plasmodium falciparum (Pf) sporozoites (SPZ) manufactured using aseptically reared Anopheles stephensi mosquitoes. Immune response genes of Anopheles mosquitoes such as Leucin-Rich protein (LRIM1), inhibit Plasmodium SPZ development (sporogony) in mosquitoes by supporting melanization and phagocytosis of ookinetes. With the aim of increasing PfSPZ infection intensities, we generated an A. stephensi LRIM1 knockout line, Δaslrim1, by embryonic genome editing using CRISPR-Cas9. Δaslrim1 mosquitoes had a significantly increased midgut bacterial load and an altered microbiome composition, including elimination of commensal acetic acid bacteria. The alterations in the microbiome caused increased mosquito mortality and unexpectedly, significantly reduced sporogony. The survival rate of Δaslrim1 mosquitoes and their ability to support PfSPZ development, were partially restored by antibiotic treatment of the mosquitoes, and fully restored to baseline when Δaslrim1 mosquitoes were produced aseptically. Deletion of LRIM1 also affected reproductive capacity: oviposition, fecundity and male fertility were significantly compromised. Attenuation in fecundity was not associated with the altered microbiome. This work demonstrates that LRIM1's regulation of the microbiome has a major impact on vector competence and longevity of A. stephensi. Additionally, LRIM1 deletion identified an unexpected role for this gene in fecundity and reduction of sperm transfer by males.


Assuntos
Anopheles/fisiologia , Sistemas CRISPR-Cas , Proteínas de Insetos/metabolismo , Malária/parasitologia , Mosquitos Vetores/crescimento & desenvolvimento , Plasmodium/crescimento & desenvolvimento , Reprodução , Animais , Bactérias/crescimento & desenvolvimento , Sistema Digestório/microbiologia , Feminino , Proteínas de Insetos/antagonistas & inibidores , Proteínas de Insetos/genética , Masculino , Mosquitos Vetores/genética , Mosquitos Vetores/parasitologia
3.
PLoS Biol ; 19(10): e3001434, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34673764

RESUMO

Productive transmission of malaria parasites hinges upon the execution of key transcriptional and posttranscriptional regulatory events. While much is now known about how specific transcription factors activate or repress sexual commitment programs, far less is known about the production of a preferred mRNA homeostasis following commitment and through the host-to-vector transmission event. Here, we show that in Plasmodium parasites, the NOT1 scaffold protein of the CAF1/CCR4/Not complex is duplicated, and one paralogue is dedicated for essential transmission functions. Moreover, this NOT1-G paralogue is central to the sex-specific functions previously associated with its interacting partners, as deletion of not1-g in Plasmodium yoelii leads to a comparable or complete arrest phenotype for both male and female parasites. We show that, consistent with its role in other eukaryotes, PyNOT1-G localizes to cytosolic puncta throughout much of the Plasmodium life cycle. PyNOT1-G is essential to both the complete maturation of male gametes and to the continued development of the fertilized zygote originating from female parasites. Comparative transcriptomics of wild-type and pynot1-g- parasites shows that loss of PyNOT1-G leads to transcript dysregulation preceding and during gametocytogenesis and shows that PyNOT1-G acts to preserve mRNAs that are critical to sexual and early mosquito stage development. Finally, we demonstrate that the tristetraprolin (TTP)-binding domain, which acts as the typical organization platform for RNA decay (TTP) and RNA preservation (ELAV/HuR) factors is dispensable for PyNOT1-G's essential blood stage functions but impacts host-to-vector transmission. Together, we conclude that a NOT1-G paralogue in Plasmodium fulfills the complex transmission requirements of both male and female parasites.


Assuntos
Estágios do Ciclo de Vida , Parasitos/crescimento & desenvolvimento , Parasitos/metabolismo , Plasmodium/crescimento & desenvolvimento , Plasmodium/metabolismo , Proteínas de Protozoários/metabolismo , Homologia de Sequência de Aminoácidos , Animais , Citosol/metabolismo , Feminino , Duplicação Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/fisiologia , Masculino , Camundongos , Modelos Biológicos , Domínios Proteicos , Mapas de Interação de Proteínas , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Deleção de Sequência , Maturidade Sexual/fisiologia , Transcriptoma/genética , Zigoto/crescimento & desenvolvimento
4.
Front Immunol ; 12: 680020, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484178

RESUMO

Lipid-derived signaling molecules known as eicosanoids have integral roles in mediating immune and inflammatory processes across metazoans. This includes the function of prostaglandins and their cognate G protein-coupled receptors (GPCRs) to employ their immunological actions. In insects, prostaglandins have been implicated in the regulation of both cellular and humoral immune responses, yet in arthropods of medical importance, studies have been limited. Here, we describe a prostaglandin E2 receptor (AgPGE2R) in the mosquito Anopheles gambiae and demonstrate that its expression is most abundant in oenocytoid immune cell populations. Through the administration of prostaglandin E2 (PGE2) and AgPGE2R-silencing, we demonstrate that prostaglandin E2 signaling regulates a subset of prophenoloxidases (PPOs) and antimicrobial peptides (AMPs) that are strongly expressed in populations of oenocytoids. We demonstrate that PGE2 signaling via the AgPGE2R significantly limits both bacterial replication and Plasmodium oocyst survival. Additional experiments establish that PGE2 treatment increases phenoloxidase (PO) activity through the increased expression of PPO1 and PPO3, genes essential to anti-Plasmodium immune responses that promote oocyst killing. We also provide evidence that the mechanisms of PGE2 signaling are concentration-dependent, where high concentrations of PGE2 promote oenocytoid lysis, negating the protective effects of lower concentrations of PGE2 on anti-Plasmodium immunity. Taken together, our results provide new insights into the role of PGE2 signaling on immune cell function and its contributions to mosquito innate immunity that promote pathogen killing.


Assuntos
Anopheles/imunologia , Anopheles/microbiologia , Anopheles/parasitologia , Dinoprostona/metabolismo , Oocistos/imunologia , Plasmodium/imunologia , Transdução de Sinais , Animais , Anopheles/classificação , Hemócitos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Viabilidade Microbiana , Mosquitos Vetores/imunologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/parasitologia , Filogenia , Plasmodium/crescimento & desenvolvimento , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo
5.
PLoS Negl Trop Dis ; 15(8): e0009668, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34437538

RESUMO

Protozoan parasites are responsible for severe disease and suffering in humans worldwide. Apart from disease transmission via insect vectors and contaminated soil, food, or water, transmission may occur congenitally or by way of blood transfusion and organ transplantation. Several recent outbreaks associated with fresh produce and potable water emphasize the need for vigilance and monitoring of protozoan parasites that cause severe disease in humans globally. Apart from the tropical parasite Plasmodium spp., other protozoa causing debilitating and fatal diseases such as Trypanosoma spp. and Naegleria fowleri need to be studied in more detail. Climate change and socioeconomic issues such as migration continue to be major drivers for the spread of these neglected tropical diseases beyond endemic zones. Due to the complex life cycles of protozoa involving multiple hosts, vectors, and stringent growth conditions, studying these parasites has been challenging. While in vivo models may provide insights into host-parasite interaction, the ethical aspects of laboratory animal use and the challenge of ready availability of parasite life stages underline the need for in vitro models as valid alternatives for culturing and maintaining protozoan parasites. To our knowledge, this review is the first of its kind to highlight available in vitro models for protozoa causing highly infectious diseases. In recent years, several research efforts using new technologies such as 3D organoid and spheroid systems for protozoan parasites have been introduced that provide valuable tools to advance complex culturing models and offer new opportunities toward the advancement of parasite in vitro studies. In vitro models aid scientists and healthcare providers in gaining insights into parasite infection biology, ultimately enabling the use of novel strategies for preventing and treating these diseases.


Assuntos
Plasmodium/crescimento & desenvolvimento , Trypanosoma/crescimento & desenvolvimento , Animais , Interações Hospedeiro-Parasita , Humanos , Estágios do Ciclo de Vida , Doenças Parasitárias/parasitologia , Parasitologia/métodos
6.
Commun Biol ; 4(1): 760, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145386

RESUMO

PP1 is a conserved eukaryotic serine/threonine phosphatase that regulates many aspects of mitosis and meiosis, often working in concert with other phosphatases, such as CDC14 and CDC25. The proliferative stages of the malaria parasite life cycle include sexual development within the mosquito vector, with male gamete formation characterized by an atypical rapid mitosis, consisting of three rounds of DNA synthesis, successive spindle formation with clustered kinetochores, and a meiotic stage during zygote to ookinete development following fertilization. It is unclear how PP1 is involved in these unusual processes. Using real-time live-cell and ultrastructural imaging, conditional gene knockdown, RNA-seq and proteomic approaches, we show that Plasmodium PP1 is implicated in both mitotic exit and, potentially, establishing cell polarity during zygote development in the mosquito midgut, suggesting that small molecule inhibitors of PP1 should be explored for blocking parasite transmission.


Assuntos
Estágios do Ciclo de Vida/genética , Meiose/genética , Mitose/genética , Plasmodium/crescimento & desenvolvimento , Proteína Fosfatase 1/genética , Proteínas de Protozoários/genética , Proliferação de Células/genética , Malária/prevenção & controle , Malária/transmissão , Mosquitos Vetores/parasitologia , Plasmodium/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas de Protozoários/metabolismo
7.
Front Immunol ; 12: 683404, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34168652

RESUMO

Antibody immunity against malaria is effective but non-sterile. In addition to antibody-mediated inhibition, neutralisation or opsonisation of malaria parasites, antibody-mediated complement activation is also important in defense against infection. Antibodies form immune complexes with parasite-derived antigens that can activate the classical complement pathway. The complement system provides efficient surveillance for infection, and its activation leads to parasite lysis or parasite opsonisation for phagocytosis. The induction of complement-fixing antibodies contributes significantly to the development of protective immunity against clinical malaria. These complement-fixing antibodies can form immune complexes that are recognised by complement receptors on innate cells of the immune system. The efficient clearance of immune complexes is accompanied by complement receptor internalisation, abrogating the detrimental consequences of excess complement activation. Here, we review the mechanisms of activation of complement by alternative, classical, and lectin pathways in human malaria at different stages of the Plasmodium life cycle with special emphasis on how complement-fixing antibodies contribute to protective immunity. We briefly touch upon the action of anaphylatoxins, the assembly of membrane attack complex, and the possible reasons underlying the resistance of infected erythrocytes towards antibody-mediated complement lysis, relevant to their prolonged survival in the blood of the human host. We make suggestions for further research on effector functions of antibody-mediated complement activation that would guide future researchers in deploying complement-fixing antibodies in preventive or therapeutic strategies against malaria.


Assuntos
Anticorpos Antiprotozoários/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Interações Hospedeiro-Parasita/imunologia , Malária/imunologia , Plasmodium/imunologia , Complexo Antígeno-Anticorpo/imunologia , Via Clássica do Complemento/imunologia , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Estágios do Ciclo de Vida , Malária/parasitologia , Plasmodium/crescimento & desenvolvimento
8.
Infect Genet Evol ; 92: 104908, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33975022

RESUMO

Plasmodium parasites, the cause of malaria, have a complex life cycle, infecting alternatively vertebrate hosts and female Anopheles mosquitoes and undergoing intra- and extra-cellular development in several organs of these hosts. Most of the ~5000 protein-coding genes present in Plasmodium genomes are only expressed at specific life stages, and different genes might therefore be subject to different selective pressures depending on the biological activity of the parasite and its microenvironment at this point in development. Here, we estimate the selective constraints on the protein-coding sequences of all annotated genes of rodent and primate Plasmodium parasites and, using data from scRNA-seq experiments spanning many developmental stages, analyze their variation with regard to when these genes are expressed in the parasite life cycle. Our study reveals extensive variation in selective constraints throughout the parasites' development and highlights stages that are evolving more rapidly than others. These findings provide novel insights into the biology of these parasites and could provide important information to develop better treatment strategies or vaccines against these medically-important organisms.


Assuntos
Doenças dos Símios Antropoides/parasitologia , Malária/veterinária , Doenças dos Macacos/parasitologia , Plasmodium/genética , Doenças dos Roedores/parasitologia , Seleção Genética , Animais , Estágios do Ciclo de Vida , Malária/parasitologia , Plasmodium/crescimento & desenvolvimento
9.
Gene ; 792: 145723, 2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-34019936

RESUMO

Avian malaria is a common and widespread disease of birds caused by a diverse group of pathogens of the genera Plasmodium. We investigated the transcriptomal profiles of one of the most common species, Plasmodium relictum, lineage SGS1, at multiple timepoints during the blood stages of the infection under experimental settings. The parasite showed well separated overall transcriptome profiles between day 8 and 20 after the infection, shown by well separated PCA profiles. Moreover, gene expression becomes more heterogenous within the experimental group late in the infection, either due to adaptations to individual differences between the experimental hosts, or due to desynchronisation of the life-cycle of the parasite. Overall, this study shows how the avian malaria system can be used to study gene expression of the avian Plasmodium parasite under controlled experimental settings, thus allowing for future comparative analysis of gene responses of parasite with different life-history traits and host effects.


Assuntos
Estágios do Ciclo de Vida/genética , Malária Aviária/parasitologia , Plasmodium/genética , Proteínas de Protozoários/genética , Transcriptoma , Animais , Aves/parasitologia , Eritrócitos/parasitologia , Regulação da Expressão Gênica , Ontologia Genética , Anotação de Sequência Molecular , Filogenia , Plasmodium/classificação , Plasmodium/crescimento & desenvolvimento , Plasmodium/metabolismo , Análise de Componente Principal , Proteínas de Protozoários/classificação , Proteínas de Protozoários/metabolismo
10.
mSphere ; 6(2)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33789941

RESUMO

Mosquitoes may feed multiple times during their life span in addition to those times needed to acquire and transmit malaria. To determine the impact of subsequent blood feeding on parasite development in Anopheles gambiae, we examined Plasmodium parasite infection with or without an additional noninfected blood meal. We found that an additional blood meal significantly reduced Plasmodium berghei immature oocyst numbers, yet had no effect on the human parasite Plasmodium falciparum These observations were reproduced when mosquitoes were fed an artificial protein meal, suggesting that parasite losses are independent of blood ingestion. We found that feeding with either a blood or protein meal compromises midgut basal lamina integrity as a result of the physical distention of the midgut, enabling the recognition and lysis of immature P. berghei oocysts by mosquito complement. Moreover, we demonstrate that additional feeding promotes P. falciparum oocyst growth, suggesting that human malaria parasites exploit host resources provided with blood feeding to accelerate their growth. This is in contrast to experiments with P. berghei, where the size of surviving oocysts is independent of an additional blood meal. Together, these data demonstrate distinct differences in Plasmodium species in evading immune detection and utilizing host resources at the oocyst stage, representing an additional, yet unexplored component of vectorial capacity that has important implications for the transmission of malaria.IMPORTANCE Mosquitoes must blood feed multiple times to acquire and transmit malaria. However, the impact of an additional mosquito blood meal following malaria parasite infection has not been closely examined. Here, we demonstrate that additional feeding affects mosquito vector competence; namely, additional feeding significantly limits Plasmodium berghei infection, yet has no effect on infection of the human parasite P. falciparum Our experiments support that these killing responses are mediated by the physical distension of the midgut and by temporary damage to the midgut basal lamina that exposes immature P. berghei oocysts to mosquito complement, while human malaria parasites are able to evade these killing mechanisms. In addition, we provide evidence that additional feeding promotes P. falciparum oocyst growth. This is in contrast to P. berghei, where oocyst size is independent of an additional blood meal. This suggests that human malaria parasites are able to exploit host resources provided by an additional feeding to accelerate their growth. In summary, our data highlight distinct differences in malaria parasite species in evading immune recognition and adapting to mosquito blood feeding. These observations have important, yet previously unexplored, implications for the impact of multiple blood meals on the transmission of malaria.


Assuntos
Anopheles/parasitologia , Comportamento Alimentar , Interações Hospedeiro-Parasita , Plasmodium/crescimento & desenvolvimento , Plasmodium/imunologia , Animais , Anopheles/fisiologia , Sangue , Feminino , Evasão da Resposta Imune , Malária/parasitologia , Malária/transmissão , Refeições , Camundongos , Mosquitos Vetores/parasitologia , Oocistos/crescimento & desenvolvimento , Oocistos/imunologia , Plasmodium/classificação , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia
11.
Eur J Med Chem ; 219: 113439, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-33887681

RESUMO

The synthesis and antimicrobial activity of new spiro-ß-lactams is reported. The design of the new molecules was based on the structural modulation of two previously identified lead spiro-penicillanates with dual activity against HIV and Plasmodium. The spiro-ß-lactams synthesized were assayed for their in vitro activity against HIV-1, providing relevant structure-activity relationship information. Among the tested compounds, two spirocyclopentenyl-ß-lactams were identified as having remarkable nanomolar activity against HIV-1. Additionally, the same molecules showed promising antiplasmodial activity, inhibiting both the hepatic and blood stages of Plasmodium infection.


Assuntos
Fármacos Anti-HIV/farmacologia , Antimaláricos/farmacologia , HIV-1/efeitos dos fármacos , Plasmodium/efeitos dos fármacos , beta-Lactamas/química , Fármacos Anti-HIV/síntese química , Antimaláricos/síntese química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , HIV-1/isolamento & purificação , Humanos , Estágios do Ciclo de Vida/efeitos dos fármacos , Conformação Molecular , Plasmodium/crescimento & desenvolvimento , Compostos de Espiro/química , Estereoisomerismo , Relação Estrutura-Atividade , beta-Lactamas/síntese química , beta-Lactamas/farmacologia
12.
Mol Microbiol ; 115(5): 870-881, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33191548

RESUMO

Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.


Assuntos
Hepatócitos/parasitologia , Malária/parasitologia , Plasmodium/fisiologia , Esporozoítos/fisiologia , Animais , Humanos , Fígado/parasitologia , Plasmodium/genética , Plasmodium/crescimento & desenvolvimento , Esporozoítos/genética , Esporozoítos/crescimento & desenvolvimento
13.
Mol Microbiol ; 115(5): 829-838, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33112460

RESUMO

Malaria-causing parasites are transmitted from humans to mosquitoes when developmentally arrested gametocytes are taken up by a female Anopheles during a blood meal. The changes in environment from human to mosquito activate gametogenesis, including a drop in temperature, a rise in pH, and a mosquito-derived molecule, xanthurenic acid. Signaling receptors have not been identified in malaria parasites but mounting evidence indicates that cGMP homeostasis is key to sensing extracellular cues in gametocytes. Low levels of cGMP maintained by phosphodiesterases prevent precocious activation of gametocytes in the human blood. Upon ingestion, initiation of gametogenesis depends on the activation of a hybrid guanylyl cyclase/P4-ATPase. Elevated cGMP levels lead to the rapid mobilization of intracellular calcium that relies upon the activation of both cGMP-dependent protein kinase and phosphoinositide phospholipase C. Once calcium is released, a cascade of phosphorylation events mediated by calcium-dependent protein kinases and phosphatases regulates the cellular processes required for gamete formation. cGMP signaling also triggers timely egress from the host cell at other life cycle stages of malaria parasites and in Toxoplasma gondii, a related apicomplexan parasite. This suggests that cGMP signaling is a versatile platform transducing external cues into calcium signals at important decision points in the life cycle of apicomplexan parasites.


Assuntos
Anopheles/parasitologia , GMP Cíclico/metabolismo , Malária/parasitologia , Plasmodium/metabolismo , Animais , Anopheles/fisiologia , Cálcio/metabolismo , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Humanos , Estágios do Ciclo de Vida , Malária/transmissão , Plasmodium/genética , Plasmodium/crescimento & desenvolvimento , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
14.
Biol Cell ; 113(2): 79-94, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33125724

RESUMO

In eco-evolutionary studies of parasite-host interactions, virulence is defined as a reduction in host fitness as a result of infection relative to an uninfected host. Pathogen virulence may either promote parasite transmission, when correlated with higher parasite replication rate, or decrease the transmission rate if the pathogen quickly kills the host. This evolutionary mechanism, referred to as 'trade-off' theory, proposes that pathogen virulence evolves towards a level that most benefits the transmission. It has been generally predicted that pathogens evolve towards low virulence in their insect vectors, mainly due to the high dependence of parasite transmission on their vector survival. Therefore, the degree of virulence which malaria parasites impose on mosquito vectors may depend on several external and internal factors. Here, we review briefly (i) the role of mosquito in parasite development, with a particular focus on mosquito midgut as the battleground between Plasmodium and the mosquito host. We aim to point out (ii) the histology of the mosquito midgut epithelium and its role in host defence against parasite's countermeasures in the three main battle sites, namely (a) the lumen (microbiota and biochemical environment), (b) the peritrophic membrane (physical barrier) and (c) the tubular epithelium including the basal membrane (physical and biochemical barrier). Lastly, (iii) we describe the impact which malaria parasite and its virulence factors have on mosquito fitness.


Assuntos
Mosquitos Vetores/parasitologia , Plasmodium/fisiologia , Animais , Sistema Digestório/parasitologia , Aptidão Genética , Interações Hospedeiro-Parasita , Humanos , Malária/parasitologia , Malária/transmissão , Plasmodium/crescimento & desenvolvimento , Plasmodium/patogenicidade
15.
Cell Rep ; 32(12): 108170, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32966787

RESUMO

The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.


Assuntos
Anemia/etiologia , Anemia/prevenção & controle , Bisfosfoglicerato Mutase/deficiência , Malária Cerebral/enzimologia , Malária Cerebral/prevenção & controle , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Bisfosfoglicerato Mutase/química , Bisfosfoglicerato Mutase/genética , Bisfosfoglicerato Mutase/metabolismo , Estabilidade Enzimática , Eritrócitos/enzimologia , Eritrócitos/parasitologia , Eritropoese , Matriz Extracelular/metabolismo , Feminino , Células HEK293 , Humanos , Malária Cerebral/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação/genética , Parasitos/crescimento & desenvolvimento , Plasmodium/crescimento & desenvolvimento , Policitemia
16.
Sci Rep ; 10(1): 14025, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32820190

RESUMO

Emergence of resistant Plasmodium species makes drug efficacy testing a crucial part of malaria control. Here we describe a novel assay for sensitive, fast and simple drug screening via the magneto-optical detection of hemozoin, a natural biomarker formed during the hemoglobin metabolism of Plasmodium species. By quantifying hemozoin production over the intraerythrocytic cycle, we reveal that hemozoin formation is already initiated by ~ 6-12 h old ring-stage parasites. We demonstrate that the new assay is capable of drug efficacy testing with incubation times as short as 6-10 h, using synchronized P. falciparum 3D7 cultures incubated with chloroquine, piperaquine and dihydroartemisinin. The determined 50% inhibitory concentrations agree well with values established by standard assays requiring significantly longer testing time. Accordingly, we conclude that magneto-optical hemozoin detection provides a practical approach for the quick assessment of drug effect with short incubation times, which may also facilitate stage-specific assessment of drug inhibitory effects.


Assuntos
Antimaláricos/farmacologia , Hemeproteínas/análise , Avaliação Pré-Clínica de Medicamentos , Resistência a Medicamentos , Humanos , Plasmodium/efeitos dos fármacos , Plasmodium/crescimento & desenvolvimento
17.
Parasitol Res ; 119(10): 3503-3515, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32772176

RESUMO

Malaria, babesiosis, trypanosomosis, and leishmaniasis are some of the most life-threatening parasites, but the range of drugs to treat them is limited. An effective, safe, and low-cost drug with a large activity spectrum is urgently needed. For this purpose, an aryl amino alcohol derivative called Alsinol was resynthesized, screened in silico, and tested against Plasmodium, Babesia, Trypanosoma, and Leishmania. In silico Alsinol follows the Lipinski and Ghose rules. In vitro it had schizontocidal activity against Plasmodium falciparum and was able to inhibit gametocytogenesis; it was particularly active against late gametocytes. In malaria-infected mice, it showed a dose-dependent activity similar to chloroquine. It demonstrated a similar level of activity to reference compounds against Babesia divergens, and against promastigotes, and amastigotes stages of Leishmania in vitro. It inhibited the in vitro growth of two African animal strains of Trypanosoma but was ineffective in vivo in our experimental conditions. It showed moderate toxicity in J774A1 and Vero cell models. The study demonstrated that Alsinol has a large spectrum of activity and is potentially affordable to produce. Nevertheless, challenges remain in the process of scaling up synthesis, creating a suitable clinical formulation, and determining the safety margin in preclinical models.


Assuntos
Amino Álcoois/farmacologia , Antiprotozoários/farmacologia , Amino Álcoois/síntese química , Amino Álcoois/química , Animais , Antiprotozoários/síntese química , Antiprotozoários/química , Babesia/efeitos dos fármacos , Babesia/crescimento & desenvolvimento , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Modelos Animais de Doenças , Leishmania/efeitos dos fármacos , Leishmania/crescimento & desenvolvimento , Estágios do Ciclo de Vida/efeitos dos fármacos , Camundongos , Plasmodium/efeitos dos fármacos , Plasmodium/crescimento & desenvolvimento , Infecções por Protozoários/tratamento farmacológico , Infecções por Protozoários/parasitologia , Resultado do Tratamento , Trypanosoma/efeitos dos fármacos , Trypanosoma/crescimento & desenvolvimento , Células Vero
18.
Molecules ; 25(15)2020 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-32731386

RESUMO

Malaria remains one of the most prevalent infectious diseases worldwide, primarily affecting some of the most vulnerable populations around the globe. Despite achievements in the treatment of this devastating disease, there is still an urgent need for the discovery of new drugs that tackle infection by Plasmodium parasites. However, de novo drug development is a costly and time-consuming process. An alternative strategy is to evaluate the anti-plasmodial activity of compounds that are already approved for other purposes, an approach known as drug repurposing. Here, we will review efforts to assess the anti-plasmodial activity of existing drugs, with an emphasis on the obligatory and clinically silent liver stage of infection. We will also review the current knowledge on the classes of compounds that might be therapeutically relevant against Plasmodium in the context of other communicable diseases that are prevalent in regions where malaria is endemic. Repositioning existing compounds may constitute a faster solution to the current gap of prophylactic and therapeutic drugs that act on Plasmodium parasites, overall contributing to the global effort of malaria eradication.


Assuntos
Antimaláricos/uso terapêutico , Reposicionamento de Medicamentos , Fígado/parasitologia , Malária/tratamento farmacológico , Plasmodium/crescimento & desenvolvimento , Animais , Humanos , Malária/metabolismo , Malária/patologia
19.
Cell Chem Biol ; 27(7): 817-826.e5, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32619453

RESUMO

Advances in infectious disease control strategies through genetic manipulation of insect microbiomes have heightened interest in microbially produced small molecules within mosquitoes. Herein, 33 mosquito-associated bacterial genomes were mined and over 700 putative biosynthetic gene clusters (BGCs) were identified, 135 of which belong to known classes of BGCs. After an in-depth analysis of the 135 BGCs, iron-binding siderophores were chosen for further investigation due to their high abundance and well-characterized bioactivities. Through various metabolomic strategies, eight siderophore scaffolds were identified in six strains of mosquito-associated bacteria. Among these, serratiochelin A and pyochelin were found to reduce female Anopheles gambiae overall fecundity likely by lowering their blood-feeding rate. Serratiochelin A and pyochelin were further found to inhibit the Plasmodium parasite asexual blood and liver stages in vitro. Our work supplies a bioinformatic resource for future mosquito-microbiome studies and highlights an understudied source of bioactive small molecules.


Assuntos
Anopheles/microbiologia , Antimaláricos/farmacologia , Bactérias/genética , Reprodução/efeitos dos fármacos , Sideróforos/farmacologia , Animais , Anopheles/crescimento & desenvolvimento , Anopheles/parasitologia , Bactérias/classificação , Genoma Bacteriano , Humanos , Intestinos/microbiologia , Estágios do Ciclo de Vida/efeitos dos fármacos , Microbiota/genética , Família Multigênica , Fenóis/farmacologia , Filogenia , Plasmodium/efeitos dos fármacos , Plasmodium/crescimento & desenvolvimento , Tiazóis/farmacologia
20.
Interdiscip Sci ; 12(2): 217-225, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32394271

RESUMO

Malaria is one of the epidemics that can cause human death. Accurate and rapid diagnosis of malaria is important for treatment. Due to the limited number of data and human factors, the prediction performance and reliability of traditional classification methods are often affected. In this study, we propose an efficient and novel classification network named Attentive Dense Circular Net (ADCN) which based on Convolutional Neural Networks (CNN). The ADCN is inspired by the ideas of residual and dense networks and combines with the attention mechanism. We train and evaluate our proposed model on a publicly available red blood cells (RBC) dataset and compare ADCN with several well-established CNN models. Compared to other best performing CNN model in our experiments, ADCN shows superiority in all performance criteria such as accuracy (97.47% vs 94.61%), sensitivity (97.86% vs 95.20%) and specificity (97.07% vs 92.87%). Finally, we discuss the obtained results and analyze the difficulties of RBCs classification.


Assuntos
Eritrócitos/parasitologia , Malária/diagnóstico , Modelos Biológicos , Redes Neurais de Computação , Plasmodium/crescimento & desenvolvimento , Aprendizado Profundo , Eritrócitos/classificação , Humanos , Malária/sangue , Malária/parasitologia , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...