Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 42(16): 3621-3629, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38704253

RESUMO

Recent data indicate increasing disease burden and importance of Plasmodium vivax (Pv) malaria. A robust assay will be essential for blood-stage Pv vaccine development. Results of the in vitro growth inhibition assay (GIA) with transgenic P. knowlesi (Pk) parasites expressing the Pv Duffy-binding protein region II (PvDBPII) correlate with in vivo protection in the first PvDBPII controlled human malaria infection (CHMI) trials, making the PkGIA an ideal selection tool once the precision of the assay is defined. To determine the precision in percentage of inhibition in GIA (%GIA) and in GIA50 (antibody concentration that gave 50 %GIA), ten GIAs with transgenic Pk parasites were conducted with four different anti-PvDBPII human monoclonal antibodies (mAbs) at concentrations of 0.016 to 2 mg/mL, and three GIAs with eighty anti-PvDBPII human polyclonal antibodies (pAbs) at 10 mg/mL. A significant assay-to-assay variation was observed, and the analysis revealed a standard deviation (SD) of 13.1 in the mAb and 5.94 in the pAb dataset for %GIA, with a LogGIA50 SD of 0.299 (for mAbs). Moreover, the ninety-five percent confidence interval (95 %CI) for %GIA or GIA50 in repeat assays was calculated in this investigation. The error range determined in this study will help researchers to compare PkGIA results from different assays and studies appropriately, thus supporting the development of future blood-stage malaria vaccine candidates, specifically second-generation PvDBPII-based formulations.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Vacinas Antimaláricas , Plasmodium knowlesi , Plasmodium vivax , Proteínas de Protozoários , Receptores de Superfície Celular , Vacinas Antimaláricas/imunologia , Plasmodium knowlesi/imunologia , Plasmodium knowlesi/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Plasmodium vivax/imunologia , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Humanos , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/genética , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/sangue , Malária Vivax/prevenção & controle , Malária Vivax/imunologia , Anticorpos Monoclonais/imunologia , Desenvolvimento de Vacinas/métodos , Animais
2.
Trop Biomed ; 38(3): 265-275, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34362869

RESUMO

Malaria caused by Plasmodium knowlesi species has become a public health concern, especially in Malaysia. Plasmodium knowlesi parasite which originates from the macaque species, infects human through the bite of the Anopheles mosquitoes. Research on malaria vaccine has been a continuous effort to eradicate the malaria infection, yet there is no vaccine against P. knowlesi malaria to date. Apical membrane antigen 1 (AMA1) is a unique surface protein of all apicomplexan parasites that plays a crucial role in parasite-host cell invasion and thus has been a long-standing malaria vaccine candidate. The selection of protective epitopes in silico has led to significant advances in the design of the vaccine. The present study aimed to employ bioinformatics tools to predict the potential immunogenic B- and T-cell epitopes in designing malaria vaccine targeting P. knowlesi AMA1 (PkAMA1). B-cell epitopes were predicted using four bioinformatics tools, i.e., BepiPred, ABCpred, BcePred, and IEDB servers whereas T-cell epitopes were predicted using two bioinformatics servers, i.e., NetMHCpan4.1 and NetMHCIIpan-4.0 targeting human major histocompatibility complex (MHC) class I and class II molecules, respectively. The antigenicity of the selected epitopes computed by both B- and T-cell predictors were further analyzed using the VaxiJen server. The results demonstrated that PkAMA1 protein encompasses multi antigenic regions that have the potential for the development of multi-epitope vaccine. Two B- and T-cell epitopes consensus regions, i.e., NSGIRIDLGEDAEVGNSKYRIPAGKCP (codons 28-54) and KTHAASFVIAEDQNTSY RHPAVYDEKNKT (codons 122-150) at domain I (DI) of PkAMA1 were reported. Advancement of bioinformatics in characterization of the target protein may facilitate vaccine development especially in vaccine design which is costly and cumbersome process. Thus, comprehensive B-cell and T-cell epitope prediction of PkAMA1 offers a promising pipeline for the development and design of multi-epitope vaccine against P. knowlesi.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas , Malária , Proteínas de Membrana/imunologia , Plasmodium knowlesi , Proteínas de Protozoários/imunologia , Biologia Computacional , Epitopos de Linfócito T , Humanos , Malária/prevenção & controle , Plasmodium knowlesi/imunologia , Vacinologia
3.
PLoS Pathog ; 17(7): e1008864, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34197567

RESUMO

Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up.


Assuntos
Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium knowlesi/imunologia , Plasmodium vivax/imunologia , Antígenos de Protozoários/imunologia , Células Cultivadas , Humanos , Malária Vivax/prevenção & controle , Proteínas de Protozoários/imunologia
4.
Am J Trop Med Hyg ; 104(2): 680-682, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33319732

RESUMO

Invasion of Plasmodium knowlesi merozoite into human erythrocytes involves molecular interaction between the parasite's Duffy binding protein (PkDBPαII) and the Duffy antigen receptor for chemokines on the erythrocytes. This study investigates the binding activity of human erythrocyte with PkDBPαII of P. knowlesi isolates from high and low parasitemic patients in an erythrocyte binding assay. The binding activity was determined by counting the number and measuring the size of rosettes formed in the assay. The protein PkDBPαII of P. knowlesi isolated from low parasitemia cases produced significantly higher number of rosettes with human erythrocytes than high parasitemia case isolates (65.5 ± 12.9 and 17.2 ± 5.5, respectively). Interestingly, PkDBPαII of isolates from high parasitemia cases formed significantly larger rosettes with human erythrocytes than PkDBPαII of isolates from low parasitemia cases (18,000 ± 13,000 µm2 and 1,315 ± 623 µm2, respectively).


Assuntos
Antígenos de Protozoários/metabolismo , Eritrócitos/metabolismo , Parasitemia/parasitologia , Plasmodium knowlesi/imunologia , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/metabolismo , Antígenos de Protozoários/imunologia , Sistema do Grupo Sanguíneo Duffy , Humanos , Plasmodium knowlesi/química , Ligação Proteica
5.
Malar J ; 19(1): 313, 2020 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-32867784

RESUMO

BACKGROUND: Rhesus macaques are valuable pre-clinical models for malaria vaccine development. The Plasmodium knowlesi/rhesus and Plasmodium falciparum/rhesus models are two established platforms for malaria vaccine testing, and both have previously been used to assess live-attenuated sporozoite vaccines. However, there is evidence that the susceptibility of the rhesus liver to P. knowlesi versus P. falciparum sporozoites likely differs, potentially complicating comparisons between these two platforms. METHODS: To quantify the differing susceptibility of rhesus to P. knowlesi and P. falciparum sporozoites, animals were infected by direct venous inoculation of purified, cryopreserved wild-type P. knowlesi sporozoites (PkSPZ) or P. falciparum sporozoites (PfSPZ). The entire liver was collected 5 days post-infection, and parasite burden in each liver lobe was quantified using an ultrasensitive Plasmodium 18S rRNA RT-PCR biomarker assay. The potential of using 18S rRNA copy number in the rhesus liver to directly measure the efficacy of vaccines targeting P. falciparum sporozoites and liver stages was also theoretically evaluated. RESULTS: Infection of rhesus with a high dose of PkSPZ led to consistently high burden liver stage infections (range 9.5-10.1 log10 copies 18S rRNA/g of liver), with similar amounts of parasite 18S rRNA detected in every liver lobe. Inoculation of rhesus with high doses of PfSPZ led to more variable, lower liver burdens (range 4.9-6.6 log10 copies 18S rRNA/g of liver in infected lobes), with parasite 18S rRNA below the limit of detection in some liver lobes. The low signal and heterogeneity of liver burden in the PfSPZ-infected animals indicates that even this extremely sensitive molecular assay cannot be used to assess reliably vaccine efficacy in the P. falciparum/rhesus platform. CONCLUSIONS: Detection of 18S rRNA in the liver following high dose intravenous PfSPZ confirmed that rhesus are modestly susceptible to wild-type P. falciparum sporozoites. However, comparison of 18S rRNA RT-PCR biomarker signal indicates that the P. falciparum liver burden was 3-5 logs lower than in PkSPZ-infected animals. Quantification of this difference in liver stage burden will help guide and interpret data from pre-clinical studies of live-attenuated sporozoite vaccines in rhesus models.


Assuntos
Macaca mulatta/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Plasmodium knowlesi/imunologia , Esporozoítos/imunologia , Animais , Feminino , Fígado/parasitologia , Macaca mulatta/parasitologia , Masculino , RNA de Protozoário/metabolismo , RNA Ribossômico 18S/metabolismo , Vacinas Atenuadas/imunologia
6.
PLoS Negl Trop Dis ; 14(6): e0008323, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32559186

RESUMO

Malaria is caused by multiple different species of protozoan parasites, and interventions in the pre-elimination phase can lead to drastic changes in the proportion of each species causing malaria. In endemic areas, cross-reactivity may play an important role in the protection and blocking transmission. Thus, successful control of one species could lead to an increase in other parasite species. A few studies have reported cross-reactivity producing cross-immunity, but the extent of cross-reactive, particularly between closely related species, is poorly understood. P. vivax and P. knowlesi are particularly closely related species causing malaria infections in SE Asia, and whilst P. vivax cases are in decline, zoonotic P. knowlesi infections are rising in some areas. In this study, the cross-species reactivity and growth inhibition activity of P. vivax blood-stage antigen-specific antibodies against P. knowlesi parasites were investigated. Bioinformatics analysis, immunofluorescence assay, western blotting, protein microarray, and growth inhibition assay were performed to investigate the cross-reactivity. P. vivax blood-stage antigen-specific antibodies recognized the molecules located on the surface or released from apical organelles of P. knowlesi merozoites. Recombinant P. vivax and P. knowlesi proteins were also recognized by P. knowlesi- and P. vivax-infected patient antibodies, respectively. Immunoglobulin G against P. vivax antigens from both immune animals and human malaria patients inhibited the erythrocyte invasion by P. knowlesi. This study demonstrates that there is extensive cross-reactivity between antibodies against P. vivax to P. knowlesi in the blood stage, and these antibodies can potently inhibit in vitro invasion, highlighting the potential cross-protective immunity in endemic areas.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Reações Cruzadas , Malária/imunologia , Plasmodium knowlesi/imunologia , Plasmodium vivax/imunologia , Animais , Humanos , Camundongos , Análise de Sequência de Proteína
7.
Malar J ; 19(1): 31, 2020 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-31952523

RESUMO

BACKGROUND: Malaria remains a global health problem and accurate surveillance of Plasmodium parasites that are responsible for this disease is required to guide the most effective distribution of control measures. Serological surveillance will be particularly important in areas of low or periodic transmission because patient antibody responses can provide a measure of historical exposure. While methods for detecting host antibody responses to Plasmodium falciparum and Plasmodium vivax are well established, development of serological assays for Plasmodium knowlesi, Plasmodium ovale and Plasmodium malariae have been inhibited by a lack of immunodiagnostic candidates due to the limited availability of genomic information. METHODS: Using the recently completed genome sequences from P. malariae, P. ovale and P. knowlesi, a set of 33 candidate cell surface and secreted blood-stage antigens was selected and expressed in a recombinant form using a mammalian expression system. These proteins were added to an existing panel of antigens from P. falciparum and P. vivax and the immunoreactivity of IgG, IgM and IgA immunoglobulins from individuals diagnosed with infections to each of the five different Plasmodium species was evaluated by ELISA. Logistic regression modelling was used to quantify the ability of the responses to determine prior exposure to the different Plasmodium species. RESULTS: Using sera from European travellers with diagnosed Plasmodium infections, antigens showing species-specific immunoreactivity were identified to select a panel of 22 proteins from five Plasmodium species for serological profiling. The immunoreactivity to the antigens in the panel of sera taken from travellers and individuals living in malaria-endemic regions with diagnosed infections showed moderate power to predict infections by each species, including P. ovale, P. malariae and P. knowlesi. Using a larger set of patient samples and logistic regression modelling it was shown that exposure to P. knowlesi could be accurately detected (AUC = 91%) using an antigen panel consisting of the P. knowlesi orthologues of MSP10, P12 and P38. CONCLUSIONS: Using the recent availability of genome sequences to all human-infective Plasmodium spp. parasites and a method of expressing Plasmodium proteins in a secreted functional form, an antigen panel has been compiled that will be useful to determine exposure to these parasites.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Adulto , Antígenos de Protozoários/genética , Área Sob a Curva , Western Blotting , Reações Cruzadas , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Funções Verossimilhança , Modelos Logísticos , Malária/diagnóstico , Malária/imunologia , Malaui , Malásia , Plasmodium knowlesi/genética , Plasmodium knowlesi/imunologia , Plasmodium malariae/genética , Plasmodium malariae/imunologia , Plasmodium ovale/genética , Plasmodium ovale/imunologia , Proteínas de Protozoários/genética , Curva ROC , Proteínas Recombinantes/imunologia , Suécia , Viagem
8.
Korean J Parasitol ; 57(4): 445-450, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31533414

RESUMO

Human infections due to the monkey malaria parasite Plasmodium knowlesi is increasingly being reported from most Southeast Asian countries specifically Malaysia. The parasite causes severe and fatal malaria thus there is a need for urgent measures for its control. In this study, the level of polymorphisms, haplotypes and natural selection of full-length pkmsp8 in 37 clinical samples from Malaysian Borneo along with 6 lab-adapted strains were investigated. Low levels of polymorphism were observed across the full-length gene, the double epidermal growth factor (EGF) domains were mostly conserved, and non-synonymous substitutions were absent. Evidence of strong negative selection pressure in the non-EGF regions were found indicating functional constrains acting at different domains. Phylogenetic haplotype network analysis identified shared haplotypes and indicated geographical clustering of samples originating from Peninsular Malaysia and Malaysian Borneo. This is the first study to genetically characterize the full-length msp8 gene from clinical isolates of P. knowlesi from Malaysia; however, further functional characterization would be useful for future rational vaccine design.


Assuntos
Antígenos de Protozoários/genética , Malária/parasitologia , Plasmodium knowlesi/genética , Proteínas de Protozoários/genética , Antígenos de Protozoários/imunologia , Haplótipos , Malária/prevenção & controle , Malásia , Plasmodium knowlesi/imunologia , Polimorfismo Genético , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/genética , Vacinas Protozoárias/normas
9.
Elife ; 82019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31205002

RESUMO

Tackling relapsing Plasmodium vivax and zoonotic Plasmodium knowlesi infections is critical to reducing malaria incidence and mortality worldwide. Understanding the biology of these important and related parasites was previously constrained by the lack of robust molecular and genetic approaches. Here, we establish CRISPR-Cas9 genome editing in a culture-adapted P. knowlesi strain and define parameters for optimal homology-driven repair. We establish a scalable protocol for the production of repair templates by PCR and demonstrate the flexibility of the system by tagging proteins with distinct cellular localisations. Using iterative rounds of genome-editing we generate a transgenic line expressing P. vivax Duffy binding protein (PvDBP), a lead vaccine candidate. We demonstrate that PvDBP plays no role in reticulocyte restriction but can alter the macaque/human host cell tropism of P. knowlesi. Critically, antibodies raised against the P. vivax antigen potently inhibit proliferation of this strain, providing an invaluable tool to support vaccine development.


Assuntos
Edição de Genes/métodos , Malária Vivax/genética , Parasitos/genética , Plasmodium knowlesi/genética , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/metabolismo , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , Humanos , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária Vivax/imunologia , Parasitos/imunologia , Parasitos/fisiologia , Plasmodium knowlesi/imunologia , Plasmodium knowlesi/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo
10.
Nat Microbiol ; 4(9): 1497-1507, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31133755

RESUMO

The most widespread form of malaria is caused by Plasmodium vivax. To replicate, this parasite must invade immature red blood cells through a process requiring interaction of the P. vivax Duffy binding protein (PvDBP) with its human receptor, the Duffy antigen receptor for chemokines. Naturally acquired antibodies that inhibit this interaction associate with clinical immunity, suggesting PvDBP as a leading candidate for inclusion in a vaccine to prevent malaria due to P. vivax. Here, we isolated a panel of monoclonal antibodies from human volunteers immunized in a clinical vaccine trial of PvDBP. We screened their ability to prevent PvDBP from binding to the Duffy antigen receptor for chemokines, and their capacity to block red blood cell invasion by a transgenic Plasmodium knowlesi parasite genetically modified to express PvDBP and to prevent reticulocyte invasion by multiple clinical isolates of P. vivax. This identified a broadly neutralizing human monoclonal antibody that inhibited invasion of all tested strains of P. vivax. Finally, we determined the structure of a complex of this antibody bound to PvDBP, indicating the molecular basis for inhibition. These findings will guide future vaccine design strategies and open up possibilities for testing the prophylactic use of such an antibody.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Receptores de Superfície Celular/imunologia , Anticorpos Antiprotozoários/química , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Antígenos de Protozoários/metabolismo , Cristalografia por Raios X , Sistema do Grupo Sanguíneo Duffy/metabolismo , Epitopos de Linfócito B , Eritrócitos/parasitologia , Variação Genética , Humanos , Fragmentos Fab das Imunoglobulinas , Vacinas Antimaláricas/administração & dosagem , Malária Vivax/parasitologia , Plasmodium knowlesi/genética , Plasmodium knowlesi/crescimento & desenvolvimento , Plasmodium knowlesi/imunologia , Plasmodium vivax/genética , Plasmodium vivax/crescimento & desenvolvimento , Ligação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Reticulócitos/parasitologia
11.
Lancet Planet Health ; 3(4): e179-e186, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31029229

RESUMO

BACKGROUND: Land use changes disrupt ecosystems, altering the transmission of vector-borne diseases. These changes have been associated with increasing incidence of zoonotic malaria caused by Plasmodium knowlesi; however, the population-level distributions of infection and exposure remain unknown. We aimed to measure prevalence of serological exposure to P knowlesi and assess associated risk factors. METHODS: We did an environmentally stratified, population-based, cross-sectional survey across households in the Kudat, Kota Marudu, Pitas, and Ranau districts in northern Sabah, Malaysia, encompassing a range of ecologies. Using blood samples, the transmission intensity of P knowlesi and other malaria species was measured by specific antibody prevalence and infection detected using molecular methods. Proportions and configurations of land types were extracted from maps derived from satellite images; a data-mining approach was used to select variables. A Bayesian hierarchical model for P knowlesi seropositivity was developed, incorporating questionnaire data about individual and household-level risk factors with selected landscape factors. FINDINGS: Between Sept 17, 2015, and Dec 12, 2015, 10 100 individuals with a median age of 25 years (range 3 months to 105 years) were sampled from 2849 households in 180 villages. 5·1% (95% CI 4·8-5·4) were seropositive for P knowlesi, and marked historical decreases were observed in the transmission of Plasmodium falciparum and Plasmodium vivax. Nine Plasmodium spp infections were detected. Age, male sex, contact with macaques, forest use, and raised house construction were positively associated with P knowlesi exposure, whereas residing at higher geographical elevations and use of insecticide were protective. Agricultural and forest variables, such as proportions and fragmentation of land cover types, predicted exposure at different spatial scales from households. INTERPRETATION: Although few infections were detected, P knowlesi exposure was observed in all demographic groups and was associated with occupational factors. Results suggest that agricultural expansion and forest fragmentation affect P knowlesi exposure, supporting linkages between land use change and P knowlesi transmission. FUNDING: UK Medical Research Council, Natural Environment Research Council, Economic and Social Research Council, and Biotechnology and Biosciences Research Council.


Assuntos
Malária/transmissão , Plasmodium knowlesi/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Infecções Assintomáticas/epidemiologia , Criança , Pré-Escolar , Estudos Transversais , Feminino , Humanos , Lactente , Malária/epidemiologia , Malária/parasitologia , Malásia/epidemiologia , Masculino , Pessoa de Meia-Idade , Prevalência , Fatores de Risco , Estudos Soroepidemiológicos , Adulto Jovem , Zoonoses/epidemiologia , Zoonoses/parasitologia , Zoonoses/transmissão
12.
PLoS One ; 14(1): e0210252, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30673723

RESUMO

The development of a sterilizing vaccine against malaria remains one of the highest priorities for global health research. While sporozoite vaccines targeting the pre-erythrocytic stage show great promise, it has not been possible to maintain efficacy long-term, likely due to an inability of these vaccines to maintain effector memory T cell responses in the liver. Vaccines based on human cytomegalovirus (HCMV) might overcome this limitation since vectors based on rhesus CMV (RhCMV), the homologous virus in rhesus macaques (RM), elicit and indefinitely maintain high frequency, non-exhausted effector memory T cells in extralymphoid tissues, including the liver. Moreover, RhCMV strain 68-1 elicits CD8+ T cells broadly recognizing unconventional epitopes exclusively restricted by MHC-II and MHC-E. To evaluate the potential of these unique immune responses to protect against malaria, we expressed four Plasmodium knowlesi (Pk) antigens (CSP, AMA1, SSP2/TRAP, MSP1c) in RhCMV 68-1 or in Rh189-deleted 68-1, which additionally elicits canonical MHC-Ia-restricted CD8+ T cells. Upon inoculation of RM with either of these Pk Ag expressing RhCMV vaccines, we obtained T cell responses to each of the four Pk antigens. Upon challenge with Pk sporozoites we observed a delayed appearance of blood stage parasites in vaccinated RM consistent with a 75-80% reduction of parasite release from the liver. Moreover, the Rh189-deleted RhCMV/Pk vectors elicited sterile protection in one RM. Once in the blood, parasite growth was not affected. In contrast to T cell responses induced by Pk infection, RhCMV vectors maintained sustained T cell responses to all four malaria antigens in the liver post-challenge. The delayed appearance of blood stage parasites is thus likely due to a T cell-mediated inhibition of liver stage parasite development. As such, this vaccine approach can be used to efficiently test new T cell antigens, improve current vaccines targeting the liver stage and complement vaccines targeting erythrocytic antigens.


Assuntos
Antígenos de Protozoários/imunologia , Citomegalovirus/genética , Vacinas Antimaláricas/imunologia , Malária/imunologia , Parasitemia/imunologia , Plasmodium knowlesi/imunologia , Esporozoítos/imunologia , Animais , Anopheles/imunologia , Anopheles/parasitologia , Feminino , Vetores Genéticos/administração & dosagem , Memória Imunológica , Fígado/imunologia , Fígado/parasitologia , Macaca mulatta , Malária/sangue , Malária/parasitologia , Malária/prevenção & controle , Masculino , Parasitemia/sangue , Parasitemia/parasitologia , Parasitemia/prevenção & controle , Plasmodium knowlesi/genética , Proteínas de Protozoários/imunologia , Linfócitos T/imunologia , Linfócitos T/parasitologia
13.
Parasit Vectors ; 11(1): 527, 2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-30257710

RESUMO

BACKGROUND: The merozoite of the zoonotic Plasmodium knowlesi invades human erythrocytes via the binding of its Duffy binding protein (PkDBPαII) to the Duffy antigen on the eythrocytes. The Duffy antigen has two immunologically distinct forms, Fya and Fyb. In this study, the erythrocyte-binding assay was used to quantitatively determine and compare the binding level of PkDBPαII to Fya+/b+ and Fya+/b- human erythrocytes. RESULTS: In the erythrocyte-binding assay, binding level was determined by scoring the number of rosettes that were formed by erythrocytes surrounding transfected mammalian COS-7 cells which expressed PkDBPαII. The assay result revealed a significant difference in the binding level. The number of rosettes scored for Fya+/b+ was 1.64-fold higher than that of Fya+/b- (155.50 ± 34.32 and 94.75 ± 23.16 rosettes, respectively; t(6) = -2.935, P = 0.026). CONCLUSIONS: The erythrocyte-binding assay provided a simple approach to quantitatively determine the binding level of PkDBPαII to the erythrocyte Duffy antigen. Using this assay, PkDBPαII was found to display higher binding to Fya+/b+ erythrocytes than to Fya+/b- erythrocytes.


Assuntos
Antígenos de Protozoários/metabolismo , Sistema do Grupo Sanguíneo Duffy/genética , Sistema do Grupo Sanguíneo Duffy/metabolismo , Eritrócitos/metabolismo , Plasmodium knowlesi/imunologia , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/metabolismo , Alelos , Animais , Antígenos de Protozoários/genética , Células COS , Chlorocebus aethiops , Humanos , Fenótipo , Ligação Proteica , Proteínas de Protozoários/genética , Receptores de Superfície Celular/genética
14.
Malar J ; 17(1): 272, 2018 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-30049277

RESUMO

BACKGROUND: The rapid process of malaria erythrocyte invasion involves ligand-receptor interactions. Inducing antibodies against specific ligands or receptors that abrogate the invasion process is a key challenge for blood stage vaccine development. However, few candidates were reported and remain to be validated for the discovery of new vaccine candidates in Plasmodium knowlesi. METHODS: In order to investigate the efficacy of pre-clinical vaccine candidates in P. knowlesi-infected human cases, this study describes an in vitro invasion inhibition assay, using a P. knowlesi strain adapted to in vitro growth in human erythrocytes, PkA1-H.1. Recombinant proteins of P. knowlesi Duffy binding protein alpha (PkDBPα) and apical membrane antigen 1 (PkAMA1) were produced in Escherichia coli system and rabbit antibodies were generated from immune animals. RESULTS: PkDBPα and PkAMA1 recombinant proteins were expressed as insoluble and produced as a functional refolded form for this study. Antibodies against PkDBPα and PkAMA1 specifically recognized recombinant proteins and native parasite proteins in schizont-stage parasites on the merozoite organelles. Single and combination of anti-PkDBPα and anti-PkAMA1 antibodies elicited strong growth inhibitory effects on the parasite in concentration-dependent manner. Meanwhile, IgG prevalence of PkDBPα and PkAMA1 were observed in 13.0 and 46.7% in human clinical patients, respectively. CONCLUSION: These data provide support for the validation of in vitro growth inhibition assay using antibodies of DBPα and AMA1 in human-adapted P. knowlesi parasite PkA1-H.1 strain.


Assuntos
Antígenos de Protozoários/imunologia , Eritrócitos/parasitologia , Proteínas de Membrana/imunologia , Plasmodium knowlesi/imunologia , Proteínas de Protozoários/imunologia , Receptores de Superfície Celular/imunologia , Adulto , Idoso , Escherichia coli/genética , Humanos , Microrganismos Geneticamente Modificados/genética , Pessoa de Meia-Idade , Adulto Jovem
15.
PLoS Negl Trop Dis ; 12(6): e0006432, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29902171

RESUMO

BACKGROUND: Primarily impacting poor, rural populations, the zoonotic malaria Plasmodium knowlesi is now the main cause of human malaria within Malaysian Borneo. While data is increasingly available on symptomatic cases, little is known about community-level patterns of exposure and infection. Understanding the true burden of disease and associated risk factors within endemic communities is critical for informing evidence-based control measures. METHODOLOGY/PRINCIPAL FINDINGS: We conducted comprehensive surveys in three areas where P. knowlesi transmission is reported: Limbuak, Pulau Banggi and Matunggung, Kudat, Sabah, Malaysia and Bacungan, Palawan, the Philippines. Infection prevalence was low with parasites detected by PCR in only 0.2% (4/2503) of the population. P. knowlesi PkSERA3 ag1 antibody responses were detected in 7.1% (95% CI: 6.2-8.2%) of the population, compared with 16.1% (14.6-17.7%) and 12.6% (11.2-14.1%) for P. falciparum and P. vivax. Sero-prevalence was low in individuals <10 years old for P. falciparum and P. vivax consistent with decreased transmission of non-zoonotic malaria species. Results indicated marked heterogeneity in transmission intensity between sites and P. knowlesi exposure was associated with agricultural work (OR 1.63; 95% CI 1.07-2.48) and higher levels of forest cover (OR 2.40; 95% CI 1.29-4.46) and clearing (OR 2.14; 95% CI 1.35-3.40) around houses. Spatial patterns of P. knowlesi exposure differed from exposure to non-zoonotic malaria and P. knowlesi exposed individuals were younger on average than individuals exposed to non-zoonotic malaria. CONCLUSIONS/SIGNIFICANCE: This is the first study to describe serological exposure to P. knowlesi and associated risk factors within endemic communities. Results indicate community-level patterns of infection and exposure differ markedly from demographics of reported cases, with higher levels of exposure among women and children. Further work is needed to understand these variations in risk across a wider population and spatial scale.


Assuntos
Malária/epidemiologia , Plasmodium knowlesi/isolamento & purificação , Estudos Soroepidemiológicos , Adolescente , Adulto , Animais , Anticorpos Antiprotozoários/sangue , Infecções Assintomáticas/epidemiologia , Criança , Fazendeiros , Feminino , Florestas , Humanos , Malária/imunologia , Malária/parasitologia , Malária/transmissão , Malásia/epidemiologia , Masculino , Pessoa de Meia-Idade , Filipinas/epidemiologia , Plasmodium knowlesi/genética , Plasmodium knowlesi/imunologia , Reação em Cadeia da Polimerase , Saúde Pública , Fatores de Risco , Adulto Jovem , Zoonoses
16.
PLoS Negl Trop Dis ; 12(6): e0006457, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29902183

RESUMO

BACKGROUND: Plasmodium knowlesi is the most common cause of malaria in Malaysian Borneo, with reporting limited to clinical cases presenting to health facilities and scarce data on the true extent of transmission. Serological estimations of transmission have been used with other malaria species to garner information about epidemiological patterns. However, there are a distinct lack of suitable serosurveillance tools for this neglected disease. METHODOLOGY/PRINCIPAL FINDINGS: Using in silico tools, we designed and expressed four novel P. knowlesi protein products to address the distinct lack of suitable serosurveillance tools: PkSERA3 antigens 1 and 2, PkSSP2/TRAP and PkTSERA2 antigen 1. Antibody prevalence to these antigens was determined by ELISA for three time-points post-treatment from a hospital-based clinical treatment trial in Sabah, East Malaysia (n = 97 individuals; 241 total samples for all time points). Higher responses were observed for the PkSERA3 antigen 2 (67%, 65/97) across all time-points (day 0: 36.9% 34/92; day 7: 63.8% 46/72; day 28: 58.4% 45/77) with significant differences between the clinical cases and controls (n = 55, mean plus 3 SD) (day 0 p<0.0001; day 7 p<0.0001; day 28 p<0.0001). Using boosted regression trees, we developed models to classify P. knowlesi exposure (cross-validated AUC 88.9%; IQR 86.1-91.3%) and identified the most predictive antibody responses. CONCLUSIONS/SIGNIFICANCE: The PkSERA3 antigen 2 had the highest relative variable importance in all models. Further validation of these antigens is underway to determine the specificity of these tools in the context of multi-species infections at the population level.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Malária/diagnóstico , Plasmodium knowlesi/isolamento & purificação , Antígenos de Protozoários/sangue , Antígenos de Protozoários/genética , Biomarcadores/sangue , Simulação por Computador , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Malária/epidemiologia , Malária/imunologia , Malária/transmissão , Malásia/epidemiologia , Plasmodium knowlesi/imunologia , Prevalência , Proteínas Recombinantes/imunologia
17.
Am J Trop Med Hyg ; 99(1): 17-23, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29761762

RESUMO

Important strides have been made within the past decade toward malaria elimination in many regions, and with this progress, the feasibility of eradication is once again under discussion. If the ambitious goal of eradication is to be achieved by 2040, all species of Plasmodium infecting humans will need to be targeted with evidence-based and concerted interventions. In this perspective, the potential barriers to achieving global malaria elimination are discussed with respect to the related diversities in host, parasite, and vector populations. We argue that control strategies need to be reorientated from a sequential attack on each species, dominated by Plasmodium falciparum to one that targets all species in parallel. A set of research themes is proposed to mitigate the potential setbacks on the pathway to a malaria-free world.


Assuntos
Erradicação de Doenças/métodos , Malária Falciparum/prevenção & controle , Malária Vivax/prevenção & controle , Malária/prevenção & controle , Animais , Anopheles/parasitologia , Antimaláricos/uso terapêutico , Cloroquina/uso terapêutico , Erradicação de Doenças/economia , Interações Hospedeiro-Parasita , Humanos , Malária/tratamento farmacológico , Malária/imunologia , Malária/parasitologia , Malária Falciparum/tratamento farmacológico , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Vivax/tratamento farmacológico , Malária Vivax/imunologia , Malária Vivax/parasitologia , Mosquitos Vetores/parasitologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Plasmodium knowlesi/efeitos dos fármacos , Plasmodium knowlesi/imunologia , Plasmodium knowlesi/patogenicidade , Plasmodium malariae/efeitos dos fármacos , Plasmodium malariae/imunologia , Plasmodium malariae/patogenicidade , Plasmodium ovale/efeitos dos fármacos , Plasmodium ovale/imunologia , Plasmodium ovale/patogenicidade , Plasmodium vivax/efeitos dos fármacos , Plasmodium vivax/imunologia , Plasmodium vivax/patogenicidade , Primaquina/uso terapêutico
18.
Parasitology ; 145(1): 56-70, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27938428

RESUMO

The primate malaria Plasmodium knowlesi has a long-standing history as an experimental malaria model. Studies using this model parasite in combination with its various natural and experimental non-human primate hosts have led to important advances in vaccine development and in our understanding of malaria invasion, immunology and parasite-host interactions. The adaptation to long-term in vitro continuous blood stage culture in rhesus monkey, Macaca fascicularis and human red blood cells, as well as the development of various transfection methodologies has resulted in a highly versatile experimental malaria model, further increasing the potential of what was already a very powerful model. The growing evidence that P. knowlesi is an important human zoonosis in South-East Asia has added relevance to former and future studies of this parasite species.


Assuntos
Modelos Animais de Doenças , Haplorrinos , Interações Hospedeiro-Parasita , Malária/parasitologia , Plasmodium knowlesi/fisiologia , Adaptação Biológica , Animais , Eritrócitos/parasitologia , Humanos , Macaca fascicularis , Macaca mulatta , Malária/imunologia , Malária/prevenção & controle , Malária/veterinária , Vacinas Antimaláricas/análise , Vacinas Antimaláricas/farmacologia , Doenças dos Macacos/imunologia , Doenças dos Macacos/parasitologia , Doenças dos Macacos/prevenção & controle , Plasmodium knowlesi/imunologia , Zoonoses/imunologia , Zoonoses/parasitologia , Zoonoses/prevenção & controle
19.
Parasitology ; 145(1): 71-84, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28720171

RESUMO

Plasmodium knowlesi has risen in importance as a zoonotic parasite that has been causing regular episodes of malaria throughout South East Asia. The P. knowlesi genome sequence generated in 2008 highlighted and confirmed many similarities and differences in Plasmodium species, including a global view of several multigene families, such as the large SICAvar multigene family encoding the variant antigens known as the schizont-infected cell agglutination proteins. However, repetitive DNA sequences are the bane of any genome project, and this and other Plasmodium genome projects have not been immune to the gaps, rearrangements and other pitfalls created by these genomic features. Today, long-read PacBio and chromatin conformation technologies are overcoming such obstacles. Here, based on the use of these technologies, we present a highly refined de novo P. knowlesi genome sequence of the Pk1(A+) clone. This sequence and annotation, referred to as the 'MaHPIC Pk genome sequence', includes manual annotation of the SICAvar gene family with 136 full-length members categorized as type I or II. This sequence provides a framework that will permit a better understanding of the SICAvar repertoire, selective pressures acting on this gene family and mechanisms of antigenic variation in this species and other pathogens.


Assuntos
Variação Antigênica/genética , Genoma de Protozoário/imunologia , Plasmodium knowlesi/genética , Plasmodium knowlesi/imunologia , Sequência de Bases , Genes de Protozoários/imunologia , Família Multigênica/imunologia
20.
Malar J ; 16(1): 16, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-28056979

RESUMO

BACKGROUND: The spread of artemisinin-resistant Plasmodium falciparum is a global health concern. Myanmar stands at the frontier of artemisinin-resistant P. falciparum. Myanmar also has the highest reported malaria burden in Southeast Asia; it is integral in the World Health Organization's plan to eliminate malaria in Southeast Asia, yet few epidemiological data exist for the general population in Myanmar. METHODS: This cross-sectional, probability household survey was conducted in Phyu township, Bago Region (central Myanmar), during the wet season of 2013. Interviewers collected clinical and behavioural data, recorded tympanic temperature and obtained dried blood spots for malaria PCR and serology. Plasmodium falciparum positive samples were tested for genetic mutations in the K13 region that may confer artemisinin resistance. Estimated type-specific malaria PCR prevalence and seroprevalence were calculated, with regression analysis to identify risk factors for seropositivity to P. falciparum. Data were weighted to account for unequal selection probabilities. RESULTS: 1638 participants were sampled (500 households). Weighted PCR prevalence was low (n = 41, 2.5%) and most cases were afebrile (93%). Plasmodium falciparum was the most common species (n = 19. 1.1%) and five (26%) P. falciparum samples harboured K13 mutations. Plasmodium knowlesi was detected in 1.0% (n = 16) and Plasmodium vivax was detected in 0.4% (n = 7). Seroprevalence was 9.4% for P. falciparum and 3.1% for P. vivax. Seroconversion to P. falciparum was 0.003/year in the whole population, but 16-fold higher in men over 23 years old (LR test p = 0.016). DISCUSSION: This is the first population-based seroprevalence study from central Myanmar. Low overall prevalence was discovered. However, these data suggest endemic transmission continues, probably associated with behavioural risk factors amongst working-age men. Genetic mutations associated with P. falciparum artemisinin resistance, the presence of P. knowlesi and discrete demographic risk groups present opportunities and challenges for malaria control. Responses targeted to working-age men, capable of detecting sub-clinical infections, and considering all species will facilitate malaria elimination in this setting.


Assuntos
Doenças Assintomáticas/epidemiologia , Malária/epidemiologia , Plasmodium falciparum/isolamento & purificação , Plasmodium knowlesi/isolamento & purificação , Plasmodium vivax/isolamento & purificação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Estudos Transversais , Características da Família , Feminino , Humanos , Lactente , Malária/parasitologia , Masculino , Pessoa de Meia-Idade , Mianmar/epidemiologia , Plasmodium/classificação , Plasmodium/genética , Plasmodium/isolamento & purificação , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Plasmodium knowlesi/genética , Plasmodium knowlesi/imunologia , Plasmodium vivax/genética , Plasmodium vivax/imunologia , Reação em Cadeia da Polimerase , Estudos Soroepidemiológicos , Inquéritos e Questionários , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA