Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Parasitol Drugs Drug Resist ; 23: 106-119, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38041930

RESUMO

Plasmodium falciparum is the deadliest causative agent of human malaria. This parasite has historically developed resistance to most drugs, including the current frontline treatments, so new therapeutic targets are needed. Our previous work on guanine quadruplexes (G4s) in the parasite's DNA and RNA has highlighted their influence on parasite biology, and revealed G4 stabilising compounds as promising candidates for repositioning. In particular, quarfloxin, a former anticancer agent, kills blood-stage parasites at all developmental stages, with fast rates of kill and nanomolar potency. Here we explored the molecular mechanism of quarfloxin and its related derivative CX-5461. In vitro, both compounds bound to P. falciparum-encoded G4 sequences. In cellulo, quarfloxin was more potent than CX-5461, and could prevent establishment of blood-stage malaria in vivo in a murine model. CX-5461 showed clear DNA damaging activity, as reported in human cells, while quarfloxin caused weaker signatures of DNA damage. Both compounds caused transcriptional dysregulation in the parasite, but the affected genes were largely different, again suggesting different modes of action. Therefore, CX-5461 may act primarily as a DNA damaging agent in both Plasmodium parasites and mammalian cells, whereas the complete antimalarial mode of action of quarfloxin may be parasite-specific and remains somewhat elusive.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Parasitos , Animais , Humanos , Camundongos , Plasmodium falciparum , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Malária/tratamento farmacológico , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , DNA/metabolismo , DNA/farmacologia , DNA/uso terapêutico , Mamíferos/genética
2.
Front Immunol ; 13: 1005476, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248835

RESUMO

The Malaria Vaccine Technology Roadmap 2013 (World Health Organization) aims to develop safe and effective vaccines by 2030 that will offer at least 75% protective efficacy against clinical malaria and reduce parasite transmission. Here, we demonstrate a highly effective multistage vaccine against both the pre-erythrocytic and sexual stages of Plasmodium falciparum that protects and reduces transmission in a murine model. The vaccine is based on a viral-vectored vaccine platform, comprising a highly-attenuated vaccinia virus strain, LC16m8Δ (m8Δ), a genetically stable variant of a licensed and highly effective Japanese smallpox vaccine LC16m8, and an adeno-associated virus (AAV), a viral vector for human gene therapy. The genes encoding P. falciparum circumsporozoite protein (PfCSP) and the ookinete protein P25 (Pfs25) are expressed as a Pfs25-PfCSP fusion protein, and the heterologous m8Δ-prime/AAV-boost immunization regimen in mice provided both 100% protection against PfCSP-transgenic P. berghei sporozoites and up to 100% transmission blocking efficacy, as determined by a direct membrane feeding assay using parasites from P. falciparum-positive, naturally-infected donors from endemic settings. Remarkably, the persistence of vaccine-induced immune responses were over 7 months and additionally provided complete protection against repeated parasite challenge in a murine model. We propose that application of the m8Δ/AAV malaria multistage vaccine platform has the potential to contribute to the landmark goals of the malaria vaccine technology roadmap, to achieve life-long sterile protection and high-level transmission blocking efficacy.


Assuntos
Antimaláricos , Vacinas Antimaláricas , Malária Falciparum , Animais , Anticorpos Antiprotozoários , Dependovirus/genética , Modelos Animais de Doenças , Humanos , Camundongos , Proteínas de Protozoários/genética
3.
Elife ; 102021 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-34939934

RESUMO

HAP2 is a transmembrane gamete fusogen found in multiple eukaryotic kingdoms and is structurally homologous to viral class II fusogens. Studies in Plasmodium have suggested that HAP2 is an attractive target for vaccines that block transmission of malaria. HAP2 has three extracellular domains, arranged in the order D2, D1, and D3. Here, we report monoclonal antibodies against the D3 fragment of Plasmodium berghei HAP2 and crystal structures of D3 in complex with Fab fragments of two of these antibodies, one of which blocks fertilization of Plasmodium berghei in vitro and transmission of malaria in mosquitoes. We also show how this Fab binds the complete HAP2 ectodomain with electron microscopy. The two antibodies cross-react with HAP2 among multiple plasmodial species. Our characterization of the Plasmodium D3 structure, HAP2 ectodomain architecture, and mechanism of inhibition provide insights for the development of a vaccine to block malaria transmission.


Assuntos
Anticorpos Monoclonais/metabolismo , Células Germinativas/imunologia , Malária/prevenção & controle , Malária/transmissão , Plasmodium berghei/imunologia , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Animais , Sítios de Ligação de Anticorpos , Fenômenos Biofísicos , Culicidae/parasitologia , Células Germinativas/fisiologia , Malária/imunologia , Fusão de Membrana , Ligação Proteica , Proteínas de Protozoários/química
4.
Front Cell Infect Microbiol ; 11: 774537, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869075

RESUMO

Introduction: Naturally acquired immune responses against antigens expressed on the surface of mature gametocytes develop in individuals living in malaria-endemic areas. Evidence suggests that such anti-gametocyte immunity can block the development of the parasite in the mosquito, thus playing a role in interrupting transmission. A better comprehension of naturally acquired immunity to these gametocyte antigens can aid the development of transmission-blocking vaccines and improve our understanding of the human infectious reservoir. Methods: Antigens expressed on the surface of mature gametocytes that had not previously been widely studied for evidence of naturally acquired immunity were identified for protein expression alongside Pfs230-C using either the mammalian HEK293E or the wheat germ cell-free expression systems. Where there was sequence variation in the candidate antigens (3D7 vs a clinical isolate PfKE04), both variants were expressed. ELISA was used to assess antibody responses against these antigens, as well as against crude stage V gametocyte extract (GE) and AMA1 using archived plasma samples from individuals recruited to participate in malaria cohort studies. We analyzed antibody levels (estimated from optical density units using a standardized ELISA) and seroprevalence (defined as antibody levels greater than three standard deviations above the mean levels of a pool of malaria naïve sera). We described the dynamics of antibody responses to these antigens by identifying factors predictive of antibody levels using linear regression models. Results: Of the 25 antigens selected, seven antigens were produced successfully as recombinant proteins, with one variant antigen, giving a total of eight proteins for evaluation. Antibodies to the candidate antigens were detectable in the study population (N = 216), with seroprevalence ranging from 37.0% (95% CI: 30.6%, 43.9%) for PSOP1 to 77.8% (95% CI: 71.6%, 83.1%) for G377 (3D7 variant). Responses to AMA1 and GE were more prevalent than those to the gametocyte proteins at 87.9% (95% CI: 82.8%, 91.9%) and 88.3% (95% CI: 83.1%, 92.4%), respectively. Additionally, both antibody levels and breadth of antibody responses were associated with age and concurrent parasitaemia. Conclusion: Age and concurrent parasitaemia remain important determinants of naturally acquired immunity to gametocyte antigens. Furthermore, we identify novel candidates for transmission-blocking activity evaluation.


Assuntos
Malária Falciparum , Animais , Anticorpos Antiprotozoários , Formação de Anticorpos , Antígenos de Protozoários , Humanos , Plasmodium falciparum , Proteínas de Protozoários/genética , Estudos Soroepidemiológicos
5.
JCI Insight ; 6(23)2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34609964

RESUMO

Controlled human malaria infection (CHMI) provides a highly informative means to investigate host-pathogen interactions and enable in vivo proof-of-concept efficacy testing of new drugs and vaccines. However, unlike Plasmodium falciparum, well-characterized P. vivax parasites that are safe and suitable for use in modern CHMI models are limited. Here, 2 healthy malaria-naive United Kingdom adults with universal donor blood group were safely infected with a clone of P. vivax from Thailand by mosquito-bite CHMI. Parasitemia developed in both volunteers, and prior to treatment, each volunteer donated blood to produce a cryopreserved stabilate of infected RBCs. Following stringent safety screening, the parasite stabilate from one of these donors (PvW1) was thawed and used to inoculate 6 healthy malaria-naive United Kingdom adults by blood-stage CHMI, at 3 different dilutions. Parasitemia developed in all volunteers, who were then successfully drug treated. PvW1 parasite DNA was isolated and sequenced to produce a high-quality genome assembly by using a hybrid assembly method. We analyzed leading vaccine candidate antigens and multigene families, including the vivax interspersed repeat (VIR) genes, of which we identified 1145 in the PvW1 genome. Our genomic analysis will guide future assessment of candidate vaccines and drugs, as well as experimental medicine studies.


Assuntos
Genoma/genética , Malária Falciparum/genética , Animais , Voluntários Saudáveis , Humanos , Masculino , Plasmodium vivax
6.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34135099

RESUMO

Progress towards a protective vaccine against malaria remains slow. To date, only limited protection has been routinely achieved following immunisation with either whole-parasite (sporozoite) or subunit-based vaccines. One major roadblock to vaccine progress, and to pre-erythrocytic parasite biology in general, is the continued reliance on manual salivary gland dissection for sporozoite isolation from infected mosquitoes. Here, we report development of a multi-step method, based on batch processing of homogenised whole mosquitoes, slurry, and density-gradient filtration, which combined with free-flow electrophoresis rapidly produces a pure, infective sporozoite inoculum. Human-infective Plasmodium falciparum and rodent-infective Plasmodium berghei sporozoites produced in this way are two- to threefold more infective than salivary gland dissection sporozoites in in vitro hepatocyte infection assays. In an in vivo rodent malaria model, the same P. berghei sporozoites confer sterile protection from mosquito-bite challenge when immunisation is delivered intravenously or 60-70% protection when delivered intramuscularly. By improving purity, infectivity, and immunogenicity, this method represents a key advancement in capacity to produce research-grade sporozoites, which should impact delivery of a whole-parasite based malaria vaccine at scale in the future.


Assuntos
Culicidae/parasitologia , Malária/prevenção & controle , Plasmodium berghei/patogenicidade , Plasmodium falciparum/patogenicidade , Esporozoítos/patogenicidade , Animais , Modelos Animais de Doenças , Drosophila , Células Hep G2 , Humanos , Imunização , Masculino , Ratos , Esporozoítos/crescimento & desenvolvimento
7.
Sci Rep ; 11(1): 10289, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33986416

RESUMO

There is an urgent need for high throughput, affordable methods of detecting pathogens inside insect vectors to facilitate surveillance. Near-infrared spectroscopy (NIRS) has shown promise to detect arbovirus and malaria in the laboratory but has not been evaluated in field conditions. Here we investigate the ability of NIRS to identify Plasmodium falciparum in Anopheles coluzzii mosquitoes. NIRS models trained on laboratory-reared mosquitoes infected with wild malaria parasites can detect the parasite in comparable mosquitoes with moderate accuracy though fails to detect oocysts or sporozoites in naturally infected field caught mosquitoes. Models trained on field mosquitoes were unable to predict the infection status of other field mosquitoes. Restricting analyses to mosquitoes of uninfectious and highly-infectious status did improve predictions suggesting sensitivity and specificity may be better in mosquitoes with higher numbers of parasites. Detection of infection appears restricted to homogenous groups of mosquitoes diminishing NIRS utility for detecting malaria within mosquitoes.


Assuntos
Anopheles/parasitologia , Mosquitos Vetores/parasitologia , Plasmodium falciparum/isolamento & purificação , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Animais
8.
Sci Rep ; 11(1): 1888, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479319

RESUMO

New antimalarial therapeutics are needed to ensure that malaria cases continue to be driven down, as both emerging parasite resistance to frontline chemotherapies and mosquito resistance to current insecticides threaten control programmes. Plasmodium, the apicomplexan parasite responsible for malaria, causes disease pathology through repeated cycles of invasion and replication within host erythrocytes (the asexual cycle). Antimalarial drugs primarily target this cycle, seeking to reduce parasite burden within the host as fast as possible and to supress recrudescence for as long as possible. Intense phenotypic drug screening efforts have identified a number of promising new antimalarial molecules. Particularly important is the identification of compounds with new modes of action within the parasite to combat existing drug resistance and suitable for formulation of efficacious combination therapies. Here we detail the antimalarial properties of DDD01034957-a novel antimalarial molecule which is fast-acting and potent against drug resistant strains in vitro, shows activity in vivo, and possesses a resistance mechanism linked to the membrane transporter PfABCI3. These data support further medicinal chemistry lead-optimization of DDD01034957 as a novel antimalarial chemical class and provide new insights to further reduce in vivo metabolic clearance.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Malária/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Animais , Antimaláricos/química , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Malária/parasitologia , Camundongos , Estrutura Molecular , Plasmodium/efeitos dos fármacos , Plasmodium/parasitologia , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/parasitologia , Plasmodium falciparum/fisiologia , Especificidade da Espécie
9.
Phytochemistry ; 183: 112632, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33360528

RESUMO

Chemical analysis of the aerial parts obtained from a Tunisian specimen of Daucus carota yielded to the isolation of six undescribed polyoxygenated germacranes and one elemanolide, along with one known metabolite. The stereostructures of the undescribed compounds were determined by extensive spectroscopic analysis including 1D and 2D NMR and HR-ESI-MS analysis. Due to their structural similarity with the Plasmodium transmission-blocking agent daucovirgolide G, the isolated metabolites were evaluated for their inhibitory activity on the development of Plasmodium early sporogonic stages. Three compounds proved to inhibit ookinete formation showing a good transmission blocking efficacy, but the low potency exhibited by these compounds when compared to daucovirgolide G further supports the observation that strict structural requirements do exist for the antimalarial activity of germacranolides.


Assuntos
Antimaláricos , Daucus carota , Antimaláricos/farmacologia , Espectroscopia de Ressonância Magnética , Sesquiterpenos de Germacrano/farmacologia
10.
Nat Cell Biol ; 22(12): 1399-1410, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33230302

RESUMO

Severe infections are a major stress on haematopoiesis, where the consequences for haematopoietic stem cells (HSCs) have only recently started to emerge. HSC function critically depends on the integrity of complex bone marrow (BM) niches; however, what role the BM microenvironment plays in mediating the effects of infection on HSCs remains an open question. Here, using a murine model of malaria and combining single-cell RNA sequencing, mathematical modelling, transplantation assays and intravital microscopy, we show that haematopoiesis is reprogrammed upon infection, whereby the HSC compartment turns over substantially faster than at steady-state and HSC function is drastically affected. Interferon is found to affect both haematopoietic and mesenchymal BM cells and we specifically identify a dramatic loss of osteoblasts and alterations in endothelial cell function. Osteo-active parathyroid hormone treatment abolishes infection-triggered HSC proliferation and-coupled with reactive oxygen species quenching-enables partial rescuing of HSC function.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Malária/fisiopatologia , Nicho de Células-Tronco/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Perfilação da Expressão Gênica/métodos , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Malária/parasitologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Hormônio Paratireóideo/farmacologia , Plasmodium berghei/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Nicho de Células-Tronco/genética
12.
Proc Natl Acad Sci U S A ; 117(13): 7363-7373, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32165544

RESUMO

After being ingested by a female Anopheles mosquito during a bloodmeal on an infected host, and before they can reach the mosquito salivary glands to be transmitted to a new host, Plasmodium parasites must establish an infection of the mosquito midgut in the form of oocysts. To achieve this, they must first survive a series of robust innate immune responses that take place prior to, during, and immediately after ookinete traversal of the midgut epithelium. Understanding how parasites may evade these responses could highlight new ways to block malaria transmission. We show that an ookinete and sporozoite surface protein designated as PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43) is required for parasite evasion of the Anopheles coluzzii complement-like response. Disruption of PIMMS43 in the rodent malaria parasite Plasmodium berghei triggers robust complement activation and ookinete elimination upon mosquito midgut traversal. Silencing components of the complement-like system through RNAi largely restores ookinete-to-oocyst transition but oocysts remain small in size and produce a very small number of sporozoites that additionally are not infectious, indicating that PIMMS43 is also essential for sporogonic development in the oocyst. Antibodies that bind PIMMS43 interfere with parasite immune evasion when ingested with the infectious blood meal and significantly reduce the prevalence and intensity of infection. PIMMS43 genetic structure across African Plasmodium falciparum populations indicates allelic adaptation to sympatric vector populations. These data add to our understanding of mosquito-parasite interactions and identify PIMMS43 as a target of malaria transmission blocking.


Assuntos
Anopheles/imunologia , Mosquitos Vetores/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Animais , Anopheles/metabolismo , Anopheles/parasitologia , Feminino , Interações Hospedeiro-Parasita/imunologia , Humanos , Evasão da Resposta Imune , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Mosquitos Vetores/metabolismo , Mosquitos Vetores/parasitologia , Oocistos/imunologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Esporozoítos/imunologia
13.
Malar J ; 18(1): 381, 2019 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-31783858

RESUMO

BACKGROUND: Malaria disease commences when blood-stage parasites, called merozoites, invade human erythrocytes. Whilst the process of invasion is traditionally seen as being entirely merozoite-driven, emerging data suggests erythrocyte biophysical properties markedly influence invasion. Cholesterol is a major determinant of cell membrane biophysical properties demanding its interrogation as a potential mediator of resistance to merozoite invasion of the erythrocyte. METHODS: Biophysical measurements of erythrocyte deformability by flicker spectroscopy were used to assess changes in erythrocyte bending modulus on forced integration of cholesterol and how these artificial changes affect invasion by human Plasmodium falciparum merozoites. To validate these observations in a natural context, either murine Plasmodium berghei or human Plasmodium falciparum merozoites were tested for their ability to invade erythrocytes from a hypercholesterolaemic mouse model or human clinical erythrocyte samples deriving from patients with a range of serum cholesterol concentrations, respectively. RESULTS: Erythrocyte bending modulus (a measure of deformability) was shown to be markedly affected by artificial modulation of cholesterol content and negatively correlated with merozoite invasion efficiency. In an in vitro infection context, however, erythrocytes taken from hypercholesterolaemic mice or from human clinical samples with varying serum cholesterol levels showed little difference in their susceptibility to merozoite invasion. Explaining this, membrane cholesterol levels in both mouse and human hypercholesterolaemia erythrocytes were subsequently found to be no different from matched normal serum controls. CONCLUSIONS: Based on these observations, serum cholesterol does not appear to impact on erythrocyte susceptibility to merozoite entry. Indeed, no relationship between serum cholesterol and cholesterol content of the erythrocyte is apparent. This work, nonetheless, suggests that native polymorphisms which do affect membrane lipid composition would be expected to affect parasite entry. This supports investigation of erythrocyte biophysical properties in endemic settings, which may yet identify naturally protective lipid-related polymorphisms.


Assuntos
Colesterol/sangue , Dislipidemias/etiologia , Eritrócitos/parasitologia , Malária/fisiopatologia , Plasmodium berghei/fisiologia , Plasmodium falciparum/fisiologia , Animais , Fenômenos Biofísicos , Humanos , Malária Falciparum/fisiopatologia , Masculino , Camundongos
14.
Sci Rep ; 9(1): 18300, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31797966

RESUMO

Inhibiting transmission of Plasmodium is an essential strategy in malaria eradication, and the biological process of gamete fusion during fertilization is a proven target for this approach. Lack of knowledge of the mechanisms underlying fertilization have been a hindrance in the development of transmission-blocking interventions. Here we describe a protein disulphide isomerase essential for malarial transmission (PDI-Trans/PBANKA_0820300) to the mosquito. We show that PDI-Trans activity is male-specific, surface-expressed, essential for fertilization/transmission, and exhibits disulphide isomerase activity which is up-regulated post-gamete activation. We demonstrate that PDI-Trans is a viable anti-malarial drug and vaccine target blocking malarial transmission with the use of PDI inhibitor bacitracin (98.21%/92.48% reduction in intensity/prevalence), and anti-PDI-Trans antibodies (66.22%/33.16% reduction in intensity/prevalence). To our knowledge, these results provide the first evidence that PDI function is essential for malarial transmission, and emphasize the potential of anti-PDI agents to act as anti-malarials, facilitating the future development of novel transmission-blocking interventions.


Assuntos
Antimaláricos , Bacitracina , Vacinas Antimaláricas , Malária , Plasmodium berghei/enzimologia , Isomerases de Dissulfetos de Proteínas/fisiologia , Animais , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Bacitracina/farmacologia , Bacitracina/uso terapêutico , Feminino , Malária/prevenção & controle , Malária/transmissão , Vacinas Antimaláricas/farmacologia , Vacinas Antimaláricas/uso terapêutico , Masculino , Camundongos , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/patogenicidade , Proteínas de Protozoários/fisiologia
15.
Front Immunol ; 10: 2480, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695697

RESUMO

Background: Malaria elimination remains a priority research agenda with the need for interventions that reduce and/or block malaria transmission from humans to mosquitoes. Transmission-blocking vaccines (TBVs) are in development, most of which target the transmission stage (i.e., gametocyte) antigens Pfs230 and Pfs48/45. For these interventions to be implemented, there is a need to understand the naturally acquired immunity to gametocytes. Several studies have measured the prevalence of immune responses to Pfs230 and Pfs48/45 in populations in malaria-endemic areas. Methods: We conducted a systematic review of studies carried out in African populations that measured the prevalence of immune responses to the gametocyte antigens Pfs230 and Pfs48/45. We assessed seroprevalence of antibody responses to the two antigens and investigated the effects of covariates such as age, transmission intensity/endemicity, season, and parasite prevalence on the prevalence of these antibody responses by meta-regression. Results: We identified 12 studies covering 23 sites for inclusion in the analysis. We found that the range of reported seroprevalence to Pfs230 and Pfs48/45 varied widely across studies, from 0 to 64% for Pfs48/45 and from 6 to 72% for Pfs230. We also found a modest association between increased age and increased seroprevalence to Pfs230: adults were associated with higher seroprevalence estimates in comparison to children (ß coefficient 0.21, 95% CI: 0.05-0.38, p = 0.042). Methodological factors were the most significant contributors to heterogeneity between studies which prevented calculation of pooled prevalence estimates. Conclusions: Naturally acquired sexual stage immunity, as detected by antibodies to Pfs230 and Pfs48/45, was present in most studies analyzed. Significant between-study heterogeneity was seen, and methodological factors were a major contributor to this, and prevented further analysis of epidemiological and biological factors. This demonstrates a need for standardized protocols for conducting and reporting seroepidemiological analyses.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Estágios do Ciclo de Vida/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum , Plasmodium falciparum/imunologia , África , Antígenos de Protozoários/uso terapêutico , Feminino , Humanos , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Masculino
16.
Elife ; 72018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29914622

RESUMO

Anti-malarial pre-erythrocytic vaccines (PEV) target transmission by inhibiting human infection but are currently partially protective. It has been posited, but never demonstrated, that co-administering transmission-blocking vaccines (TBV) would enhance malaria control. We hypothesized a mechanism that TBV could reduce parasite density in the mosquito salivary glands, thereby enhancing PEV efficacy. This was tested using a multigenerational population assay, passaging Plasmodium berghei to Anopheles stephensi mosquitoes. A combined efficacy of 90.8% (86.7-94.2%) was observed in the PEV +TBV antibody group, higher than the estimated efficacy of 83.3% (95% CrI 79.1-87.0%) if the two antibodies acted independently. Higher PEV efficacy at lower mosquito parasite loads was observed, comprising the first direct evidence that co-administering anti-sporozoite and anti-transmission interventions act synergistically, enhancing PEV efficacy across a range of TBV doses and transmission intensities. Combining partially effective vaccines of differing anti-parasitic classes is a pragmatic, powerful way to accelerate malaria elimination efforts.


Assuntos
Anticorpos Bloqueadores/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Anticorpos Antiprotozoários/administração & dosagem , Vacinas Antimaláricas/administração & dosagem , Malária/prevenção & controle , Plasmodium berghei/imunologia , Esporozoítos/imunologia , Animais , Anopheles/parasitologia , Sinergismo Farmacológico , Feminino , Humanos , Malária/imunologia , Malária/parasitologia , Camundongos , Mosquitos Vetores/parasitologia , Carga Parasitária , Plasmodium berghei/efeitos dos fármacos , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Glândulas Salivares/parasitologia , Esporozoítos/química , Trofozoítos/química , Trofozoítos/imunologia
17.
Parasit Vectors ; 11(1): 377, 2018 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-29954424

RESUMO

BACKGROUND: The proportion of mosquitoes infected with malaria is an important entomological metric used to assess the intensity of transmission and the impact of vector control interventions. Currently, the prevalence of mosquitoes with salivary gland sporozoites is estimated by dissecting mosquitoes under a microscope or using molecular methods. These techniques are laborious, subjective, and require either expensive equipment or training. This study evaluates the potential of near-infrared spectroscopy (NIRS) to identify laboratory reared mosquitoes infected with rodent malaria. METHODS: Anopheles stephensi mosquitoes were reared in the laboratory and fed on Plasmodium berghei infected blood. After 12 and 21 days post-feeding mosquitoes were killed, scanned and analysed using NIRS and immediately dissected by microscopy to determine the number of oocysts on the midgut wall or sporozoites in the salivary glands. A predictive classification model was used to determine parasite prevalence and intensity status from spectra. RESULTS: The predictive model correctly classifies infectious and uninfectious mosquitoes with an overall accuracy of 72%. The false negative and false positive rates were 30 and 26%, respectively. While NIRS was able to differentiate between uninfectious and highly infectious mosquitoes, differentiating between mid-range infectious groups was less accurate. Multiple scans of the same specimen, with repositioning the mosquito between scans, is shown to improve accuracy. On a smaller dataset NIRS was unable to predict whether mosquitoes harboured oocysts. CONCLUSIONS: To our knowledge, we provide the first evidence that NIRS can differentiate between infectious and uninfectious mosquitoes. Currently, distinguishing between different intensities of infection is challenging. The classification model provides a flexible framework and allows for different error rates to be optimised, enabling the sensitivity and specificity of the technique to be varied according to requirements.


Assuntos
Anopheles/parasitologia , Plasmodium berghei/isolamento & purificação , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Animais , Anopheles/ultraestrutura , Reações Falso-Positivas , Trato Gastrointestinal/citologia , Trato Gastrointestinal/parasitologia , Aprendizado de Máquina , Malária/parasitologia , Malária/transmissão , Microscopia , Mosquitos Vetores/parasitologia , Oocistos/ultraestrutura , Glândulas Salivares/parasitologia , Esporozoítos/ultraestrutura
19.
Sci Rep ; 8(1): 3896, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29497047

RESUMO

With the increasing prevalence of artemisinin-resistant malaria parasites, a highly efficacious and durable vaccine for malaria is urgently required. We have developed an experimental virus-vectored vaccine platform based on an envelope-modified baculovirus dual-expression system (emBDES). Here, we show a conceptually new vaccine platform based on an adenovirus-prime/emBDES-boost heterologous immunization regimen expressing the Plasmodium falciparum circumsporozoite protein (PfCSP). A human adenovirus 5-prime/emBDES-boost heterologous immunization regimen consistently achieved higher sterile protection against transgenic P. berghei sporozoites expressing PfCSP after a mosquito-bite challenge than reverse-ordered or homologous immunization. This high protective efficacy was also achieved with a chimpanzee adenovirus 63-prime/emBDES-boost heterologous immunization regimen against an intravenous sporozoite challenge. Thus, we show that the adenovirus-prime/emBDES-boost heterologous immunization regimen confers sterile protection against sporozoite challenge by two individual routes, providing a promising new malaria vaccine platform for future clinical use.


Assuntos
Vacinas Antimaláricas/imunologia , Esporozoítos/imunologia , Vacinação/métodos , Adenoviridae/imunologia , Infecções por Adenoviridae , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Baculoviridae/imunologia , Modelos Animais de Doenças , Feminino , Imunização/métodos , Imunização Secundária/métodos , Malária/imunologia , Malária Falciparum/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Vacinas de DNA/imunologia
20.
PLoS One ; 13(12): e0209699, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30596727

RESUMO

We recently identified three novel thioredoxin-like genes in the genome of the protozoan parasite Plasmodium that belong to the Phosducin-like family of proteins (PhLP). PhLPs are small cytosolic proteins hypothesized to function in G-protein signaling and protein folding. Although PhLPs are highly conserved in eukaryotes from yeast to mammals, only a few representatives have been experimentally characterized to date. In addition, while PhLPs contain a thioredoxin domain, they lack a CXXC motif, a strong indicator for redox activity, and it is unclear whether members of the PhLP family are enzymatically active. Here, we describe PbPhLP-3 as the first phosducin-like protein of a protozoan organism, Plasmodium berghei. Initial transcription analysis revealed continuous low-level expression of pbphlp-3 throughout the complex Plasmodium life cycle. Attempts to knockout pbphlp-3 in P. berghei did not yield live parasites, suggesting an essential role for the gene in Plasmodium. We cloned, expressed and purified PbPhLP-3 and determined that the recombinant protein is redox active in vitro in a thioredoxin-coupled redox assay. It also has the capacity to reduce the organic compound tert-Butyl hydroperoxide (TBHP) in vitro, albeit at low efficiency. Sequence analysis, structural modeling, and site-directed mutagenesis revealed a conserved cysteine in the thioredoxin domain to be the redox active residue. Lastly, we provide evidence that recombinant human PhLP-3 exhibits redox activity similar to that of PbPhLP-3 and suggest that redox activity may be conserved in PhLP-3 homologs of other species. Our data provide new insight into the function of PhLP-3, which is hypothesized to act as co-chaperones in the folding and regulation of cytoskeletal proteins. We discuss the potential implications of PhLP-3 as a thioredoxin-target protein and possible links between the cellular redox network and the eukaryotic protein folding machinery.


Assuntos
Evolução Biológica , Oxirredução , Plasmodium/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Clonagem Molecular , Evolução Molecular , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Plasmodium/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/isolamento & purificação , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...