Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
NPJ Vaccines ; 9(1): 10, 2024 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-38184681

RESUMO

The receptor-binding domain, region II, of the Plasmodium vivax Duffy binding protein (PvDBPII) binds the Duffy antigen on the reticulocyte surface to mediate invasion. A heterologous vaccine challenge trial recently showed that a delayed dosing regimen with recombinant PvDBPII SalI variant formulated with adjuvant Matrix-MTM reduced the in vivo parasite multiplication rate (PMR) in immunized volunteers challenged with the Thai P. vivax isolate PvW1. Here, we describe extensive analysis of the polyfunctional antibody responses elicited by PvDBPII immunization and identify immune correlates for PMR reduction. A classification algorithm identified antibody features that significantly contribute to PMR reduction. These included antibody titre, receptor-binding inhibitory titre, dissociation constant of the PvDBPII-antibody interaction, complement C1q and Fc gamma receptor binding and specific IgG subclasses. These data suggest that multiple immune mechanisms elicited by PvDBPII immunization are likely to be associated with protection and the immune correlates identified could guide the development of an effective vaccine for P. vivax malaria. Importantly, all the polyfunctional antibody features that correlated with protection cross-reacted with both PvDBPII SalI and PvW1 variants, suggesting that immunization with PvDBPII should protect against diverse P. vivax isolates.

2.
Cell Host Microbe ; 31(12): 2080-2092.e5, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38056460

RESUMO

Vivax malaria has long been thought to be absent from sub-Saharan Africa owing to the high proportion of individuals lacking the Duffy antigen receptor for chemokines (DARC) in their erythrocytes. The interaction between P. vivax Duffy-binding protein (PvDBP) and DARC is assumed to be the main pathway used by merozoites to invade reticulocytes. However, the increasing number of reports of vivax malaria cases in genotypically Duffy-negative (DN) individuals has raised questions regarding the P. vivax invasion pathway(s). Here, we show that a subset of DN erythroblasts transiently express DARC during terminal erythroid differentiation and that P. vivax merozoites, irrespective of their origin, can invade DARC+ DN erythroblasts. These findings reveal that a large number of DN individuals may represent a silent reservoir of deep P. vivax infections at the sites of active erythropoiesis with low or no parasitemia, and it may represent an underestimated biological problem with potential clinical consequences in sub-Saharan Africa.


Assuntos
Malária Vivax , Humanos , Antígenos de Protozoários , Proteínas de Protozoários/metabolismo , Plasmodium vivax/metabolismo , Eritrócitos , Sistema do Grupo Sanguíneo Duffy/genética , Sistema do Grupo Sanguíneo Duffy/metabolismo
3.
Lancet Reg Health West Pac ; 37: 100792, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37693871

RESUMO

Background: Assessing the status of malaria transmission in endemic areas becomes increasingly challenging as countries approach elimination. Serology can provide robust estimates of malaria transmission intensities, and multiplex serological assays allow for simultaneous assessment of markers of recent and historical malaria exposure. Methods: Here, we evaluated different statistical and machine learning methods for analyzing multiplex malaria-specific antibody response data to classify recent and historical exposure to Plasmodium falciparum and Plasmodium vivax. To assess these methods, we utilized samples from a health-facility based survey (n = 9132) in the Philippines, where we quantified antibody responses against 8 P. falciparum and 6 P. vivax-specific antigens from 3 sites with varying transmission intensity. Findings: Measurements of antibody responses and seroprevalence were consistent with the 3 sites' known endemicity status. Among the models tested, a machine learning (ML) approach (Random Forest model) using 4 serological markers (PfGLURP R2, Etramp5.Ag1, GEXP18, and PfMSP119) gave better predictions for P. falciparum recent infection in Palawan (AUC: 0.9591, CI 0.9497-0.9684) than individual antigen seropositivity. Although the ML approach did not improve P. vivax infection predictions, ML classifications confirmed the absence of recent exposure to P. falciparum and P. vivax in both Occidental Mindoro and Bataan. For predicting historical P. falciparum and P. vivax transmission, seroprevalence and seroconversion rates based on cumulative exposure markers AMA1 and MSP119 showed reliable trends in the 3 sites. Interpretation: Our study emphasizes the utility of serological markers in predicting recent and historical exposure in a sub-national elimination setting, and also highlights the potential use of machine learning models using multiplex antibody responses to improve assessment of the malaria transmission status of countries aiming for elimination. This work also provides baseline antibody data for monitoring risk in malaria-endemic areas in the Philippines. Funding: Newton Fund, Philippine Council for Health Research and Development, UK Medical Research Council.

4.
Infect Immun ; 91(10): e0026823, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37754682

RESUMO

In this study, we investigated how different categories of prenatal malaria exposure (PME) influence levels of maternal antibodies in cord blood samples and the subsequent risk of malaria in early childhood in a birth cohort study (N = 661) nested within the COSMIC clinical trial (NCT01941264) in Burkina Faso. Plasmodium falciparum infections during pregnancy and infants' clinical malaria episodes detected during the first year of life were recorded. The levels of maternal IgG and IgG1-4 to 15 P. falciparum antigens were measured in cord blood by quantitative suspension array technology. Results showed a significant variation in the magnitude of maternal antibody levels in cord blood, depending on the PME category, with past placental malaria (PM) more frequently associated with significant increases of IgG and/or subclass levels across three groups of antigens defined as pre-erythrocytic, erythrocytic, and markers of PM, as compared to those from the cord of non-exposed control infants. High levels of antibodies to certain erythrocytic antigens (i.e., IgG to EBA140 and EBA175, IgG1 to EBA175 and MSP142, and IgG3 to EBA140 and MSP5) were independent predictors of protection from clinical malaria during the first year of life. By contrast, high levels of IgG, IgG1, and IgG2 to the VAR2CSA DBL1-2 and IgG4 to DBL3-4 were significantly associated with an increased risk of clinical malaria. These findings indicate that PME categories have different effects on the levels of maternal-derived antibodies to malaria antigens in children at birth, and this might drive heterogeneity to clinical malaria susceptibility in early childhood.


Assuntos
Malária Falciparum , Malária , Criança , Lactente , Recém-Nascido , Humanos , Pré-Escolar , Feminino , Gravidez , Plasmodium falciparum , Estudos de Coortes , Burkina Faso/epidemiologia , Exposição Materna , Placenta , Anticorpos Antiprotozoários , Malária/epidemiologia , Imunoglobulina G , Antígenos de Protozoários
5.
Sci Rep ; 13(1): 13904, 2023 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-37626150

RESUMO

The invasion of reticulocytes by Plasmodium vivax merozoites is dependent on the interaction of the Plasmodium vivax Duffy Binding Protein (PvDBP) with the Duffy antigen receptor for chemokines (DARC). The N-terminal cysteine-rich region II of PvDBP (PvDBPII), which binds DARC, is a leading P. vivax malaria vaccine candidate. Here, we have evaluated the immunogenicity of recombinant PvDBPII formulated with the adjuvants Matrix-M and GLA-SE in mice. Analysis of the antibody responses revealed comparable ELISA recognition titres as well as similar recognition of native PvDBP in P. vivax schizonts by immunofluorescence assay. Moreover, antibodies elicited by the two adjuvant formulations had similar functional properties such as avidity, isotype profile and inhibition of PvDBPII-DARC binding. Furthermore, the anti-PvDBPII antibodies were able to block the interaction of DARC with the homologous PvDBPII SalI allele as well as the heterologous PvDBPII PvW1 allele from a Thai clinical isolate that is used for controlled human malaria infections (CHMI). The cross-reactivity of these antibodies with PvW1 suggest that immunization with the PvDBPII SalI strain should neutralize reticulocyte invasion by the challenge P. vivax strain PvW1.


Assuntos
Malária Vivax , Vacinas , Humanos , Animais , Camundongos , Plasmodium vivax , Proteínas de Transporte , Adjuvantes Imunológicos , Adjuvantes Farmacêuticos , Anticorpos , Malária Vivax/prevenção & controle
6.
Nat Commun ; 14(1): 4004, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37414792

RESUMO

Pregnant women attending first antenatal care (ANC) visits represent a promising malaria surveillance target in Sub-Saharan Africa. We assessed the spatio-temporal relationship between malaria trends at ANC (n = 6471) and in children in the community (n = 3933) and at health facilities (n = 15,467) in southern Mozambique (2016-2019). ANC P. falciparum rates detected by quantitative polymerase chain reaction mirrored rates in children, regardless of gravidity and HIV status (Pearson correlation coefficient [PCC] > 0.8, χ²<1.1), with a 2-3 months lag. Only at rapid diagnostic test detection limits at moderate-to-high transmission, did multigravidae show lower rates than children (PCC = 0.61, 95%CI[-0.12-0.94]). Seroprevalence against the pregnancy-specific antigen VAR2CSA reflected declining malaria trends (PCC = 0.74, 95%CI[0.24-0.77]). 60% (9/15) of hotspots detected from health facility data (n = 6662) using a novel hotspot detector, EpiFRIenDs, were also identified with ANC data (n = 3616). Taken together, we show that ANC-based malaria surveillance offers contemporary information on temporal trends and geographic distribution of malaria burden in the community.


Assuntos
Malária , Cuidado Pré-Natal , Criança , Gravidez , Feminino , Humanos , Estudos Soroepidemiológicos , Malária/diagnóstico , Malária/epidemiologia , Instalações de Saúde , Moçambique/epidemiologia
7.
Sci Transl Med ; 15(704): eadf1782, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37437014

RESUMO

There are no licensed vaccines against Plasmodium vivax. We conducted two phase 1/2a clinical trials to assess two vaccines targeting P. vivax Duffy-binding protein region II (PvDBPII). Recombinant viral vaccines using chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) vectors as well as a protein and adjuvant formulation (PvDBPII/Matrix-M) were tested in both a standard and a delayed dosing regimen. Volunteers underwent controlled human malaria infection (CHMI) after their last vaccination, alongside unvaccinated controls. Efficacy was assessed by comparisons of parasite multiplication rates in the blood. PvDBPII/Matrix-M, given in a delayed dosing regimen, elicited the highest antibody responses and reduced the mean parasite multiplication rate after CHMI by 51% (n = 6) compared with unvaccinated controls (n = 13), whereas no other vaccine or regimen affected parasite growth. Both viral-vectored and protein vaccines were well tolerated and elicited expected, short-lived adverse events. Together, these results support further clinical evaluation of the PvDBPII/Matrix-M P. vivax vaccine.


Assuntos
Malária , Parasitos , Humanos , Animais , Plasmodium vivax , Vacinação
8.
Trends Parasitol ; 39(3): 200-211, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36642689

RESUMO

During its life cycle, the human malaria parasite Plasmodium falciparum is subjected to elevated levels of oxidative stress that cause damage to membrane lipids, a process referred to as lipid peroxidation. Control and repair of lipid peroxidation is critical for survival of P. falciparum. Here, we present an introduction into lipid peroxidation and review the current knowledge about the control and repair of the damage caused by lipid peroxidation in P. falciparum blood stages. We also review the recent identification of host peroxiredoxin 6 (PRDX6), as a key lipid-peroxidation-repair enzyme in P. falciparum blood stages. Such critical host factors provide novel targets for development of drugs against malaria.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Parasitos , Animais , Humanos , Peroxidação de Lipídeos , Malária Falciparum/parasitologia , Plasmodium falciparum , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico
9.
Front Immunol ; 14: 1193079, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38299155

RESUMO

We have previously reported primary endpoints of a clinical trial testing two vaccine platforms for the delivery of Plasmodium vivax malaria DBPRII: viral vectors (ChAd63, MVA), and protein/adjuvant (PvDBPII with 50µg Matrix-M™ adjuvant). Delayed boosting was necessitated due to trial halts during the pandemic and provides an opportunity to investigate the impact of dosing regimens. Here, using flow cytometry - including agnostic definition of B cell populations with the clustering tool CITRUS - we report enhanced induction of DBPRII-specific plasma cell and memory B cell responses in protein/adjuvant versus viral vector vaccinees. Within protein/adjuvant groups, delayed boosting further improved B cell immunogenicity compared to a monthly boosting regimen. Consistent with this, delayed boosting also drove more durable anti-DBPRII serum IgG. In an independent vaccine clinical trial with the P. falciparum malaria RH5.1 protein/adjuvant (50µg Matrix-M™) vaccine candidate, we similarly observed enhanced circulating B cell responses in vaccinees receiving a delayed final booster. Notably, a higher frequency of vaccine-specific (putatively long-lived) plasma cells was detected in the bone marrow of these delayed boosting vaccinees by ELISPOT and correlated strongly with serum IgG. Finally, following controlled human malaria infection with P. vivax parasites in the DBPRII trial, in vivo growth inhibition was observed to correlate with DBPRII-specific B cell and serum IgG responses. In contrast, the CD4+ and CD8+ T cell responses were impacted by vaccine platform but not dosing regimen and did not correlate with in vivo growth inhibition in a challenge model. Taken together, our DBPRII and RH5 data suggest an opportunity for protein/adjuvant dosing regimen optimisation in the context of rational vaccine development against pathogens where protection is antibody-mediated.


Assuntos
Malária Vivax , Vacinas , Humanos , Plasmodium falciparum , Medula Óssea , Antígenos de Protozoários , Adjuvantes Imunológicos , Malária Vivax/prevenção & controle , Imunoglobulina G
10.
medRxiv ; 2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35664997

RESUMO

Background: There are no licensed vaccines against Plasmodium vivax , the most common cause of malaria outside of Africa. Methods: We conducted two Phase I/IIa clinical trials to assess the safety, immunogenicity and efficacy of two vaccines targeting region II of P. vivax Duffy-binding protein (PvDBPII). Recombinant viral vaccines (using ChAd63 and MVA vectors) were administered at 0, 2 months or in a delayed dosing regimen (0, 17, 19 months), whilst a protein/adjuvant formulation (PvDBPII/Matrix-M™) was administered monthly (0, 1, 2 months) or in a delayed dosing regimen (0, 1, 14 months). Delayed regimens were due to trial halts during the COVID-19 pandemic. Volunteers underwent heterologous controlled human malaria infection (CHMI) with blood-stage P. vivax parasites at 2-4 weeks following their last vaccination, alongside unvaccinated controls. Efficacy was assessed by comparison of parasite multiplication rate (PMR) in blood post-CHMI, modelled from parasitemia measured by quantitative polymerase-chain-reaction (qPCR). Results: Thirty-two volunteers were enrolled and vaccinated (n=16 for each vaccine). No safety concerns were identified. PvDBPII/Matrix-M™, given in the delayed dosing regimen, elicited the highest antibody responses and reduced the mean PMR following CHMI by 51% (range 36-66%; n=6) compared to unvaccinated controls (n=13). No other vaccine or regimen impacted parasite growth. In vivo growth inhibition of blood-stage P. vivax correlated with functional antibody readouts of vaccine immunogenicity. Conclusions: Vaccination of malaria-naïve adults with a delayed booster regimen of PvDBPII/ Matrix-M™ significantly reduces the growth of blood-stage P. vivax . Funded by the European Commission and Wellcome Trust; VAC069, VAC071 and VAC079 ClinicalTrials.gov numbers NCT03797989 , NCT04009096 and NCT04201431 .

11.
Cell Rep ; 39(11): 110923, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35705035

RESUMO

The uptake and digestion of host hemoglobin by malaria parasites during blood-stage growth leads to significant oxidative damage of membrane lipids. Repair of lipid peroxidation damage is crucial for parasite survival. Here, we demonstrate that Plasmodium falciparum imports a host antioxidant enzyme, peroxiredoxin 6 (PRDX6), during hemoglobin uptake from the red blood cell cytosol. PRDX6 is a lipid-peroxidation repair enzyme with phospholipase A2 (PLA2) activity. Inhibition of PRDX6 with a PLA2 inhibitor, Darapladib, increases lipid-peroxidation damage in the parasite and disrupts transport of hemoglobin-containing vesicles to the food vacuole, causing parasite death. Furthermore, inhibition of PRDX6 synergistically reduces the survival of artemisinin-resistant parasites following co-treatment of parasite cultures with artemisinin and Darapladib. Thus, PRDX6 is a host-derived drug target for development of antimalarial drugs that could help overcome artemisinin resistance.


Assuntos
Antimaláricos , Artemisininas , Malária Falciparum , Malária , Peroxirredoxina VI , Animais , Antimaláricos/farmacologia , Artemisininas/metabolismo , Artemisininas/farmacologia , Benzaldeídos/farmacologia , Resistência a Medicamentos , Hemoglobinas/metabolismo , Humanos , Lipídeos , Malária/tratamento farmacológico , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Camundongos , Oximas/farmacologia , Peroxirredoxina VI/imunologia , Peroxirredoxina VI/metabolismo , Plasmodium falciparum
12.
Cell Rep Med ; 3(6): 100662, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35732155

RESUMO

Serological markers are a promising tool for surveillance and targeted interventions for Plasmodium vivax malaria. P. vivax is closely related to the zoonotic parasite P. knowlesi, which also infects humans. P. vivax and P. knowlesi are co-endemic across much of South East Asia, making it important to design serological markers that minimize cross-reactivity in this region. To determine the degree of IgG cross-reactivity against a panel of P. vivax serological markers, we assayed samples from human patients with P. knowlesi malaria. IgG antibody reactivity is high against P. vivax proteins with high sequence identity with their P. knowlesi ortholog. IgG reactivity peaks at 7 days post-P. knowlesi infection and is short-lived, with minimal responses 1 year post-infection. We designed a panel of eight P. vivax proteins with low levels of cross-reactivity with P. knowlesi. This panel can accurately classify recent P. vivax infections while reducing misclassification of recent P. knowlesi infections.


Assuntos
Malária Vivax , Malária , Plasmodium knowlesi , Humanos , Imunoglobulina G , Malária/diagnóstico , Malária Vivax/diagnóstico , Plasmodium vivax
13.
PLoS Negl Trop Dis ; 16(6): e0010138, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35727821

RESUMO

BACKGROUND: Soil-transmitted helminths (STH), Schistosoma spp. and Plasmodium falciparum are parasites of major public health importance and co-endemic in many sub-Saharan African countries. Management of these infections requires detection and treatment of infected people and evaluation of large-scale measures implemented. Diagnostic tools are available but their low sensitivity, especially for low intensity helminth infections, leaves room for improvement. Antibody serology could be a useful approach thanks to its potential to detect both current infection and past exposure. METHODOLOGY: We evaluated total IgE responses and specific-IgG levels to 9 antigens from STH, 2 from Schistosoma spp., and 16 from P. falciparum, as potential markers of current infection in a population of children and adults from Southern Mozambique (N = 715). Antibody responses were measured by quantitative suspension array Luminex technology and their performance was evaluated by ROC curve analysis using microscopic and molecular detection of infections as reference. PRINCIPAL FINDINGS: IgG against the combination of EXP1, AMA1 and MSP2 (P. falciparum) in children and NIE (Strongyloides stercoralis) in adults and children had the highest accuracies (AUC = 0.942 and AUC = 0.872, respectively) as markers of current infection. IgG against the combination of MEA and Sm25 (Schistosoma spp.) were also reliable markers of current infection (AUC = 0.779). In addition, IgG seropositivity against 20 out of the 27 antigens in the panel differentiated the seropositive endemic population from the non-endemic population, suggesting a possible role as markers of exposure although sensitivity could not be assessed. CONCLUSIONS: We provided evidence for the utility of antibody serology to detect current infection with parasites causing tropical diseases in endemic populations. In addition, most of the markers have potential good specificity as markers of exposure. We also showed the feasibility of measuring antibody serology with a platform that allows the integration of control and elimination programs for different pathogens.


Assuntos
Helmintos , Malária Falciparum , Adulto , Animais , Criança , Humanos , Imunoglobulina G , Malária Falciparum/diagnóstico , Malária Falciparum/epidemiologia , Moçambique/epidemiologia , Plasmodium falciparum , Schistosoma
14.
Parasitol Int ; 90: 102592, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35489701

RESUMO

The unique biology of Plasmodium vivax, with its ability to form latent hypnozoites in the liver stage and the early appearance of gametocytes during blood stage infection, makes it difficult to target for elimination with standard malaria control tools. Here, we use modelling studies to demonstrate that vaccines that target different stages of P. vivax could greatly assist efforts to eliminate P. vivax. Combination of vaccines that target different P. vivax life cycle stages may be required to achieve high efficacy. Our simulations demonstrate that repeated rounds of mass vaccination with multi-stage vaccines can help achieve pre-elimination levels of P. vivax in both low and high transmission settings. We review the status of global efforts to develop vaccines for P. vivax malaria. We describe the status of the leading P. vivax vaccine candidates and share some thoughts on the prospects for availability of an effective vaccine for P. vivax malaria.


Assuntos
Vacinas Antimaláricas , Malária Vivax , Malária , Animais , Estágios do Ciclo de Vida , Malária Vivax/prevenção & controle , Plasmodium vivax
15.
BMC Med ; 20(1): 89, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-35260169

RESUMO

BACKGROUND: Plasmodium vivax (P. vivax) is the dominant Plasmodium spp. causing the disease malaria in low-transmission regions outside of Africa. These regions often feature high proportions of asymptomatic patients with sub-microscopic parasitaemia and relapses. Naturally acquired antibody responses are induced after Plasmodium infection, providing partial protection against high parasitaemia and clinical episodes. However, previous work has failed to address the presence and maintenance of such antibody responses to P. vivax particularly in low-transmission regions. METHODS: We followed 34 patients in western Thailand after symptomatic P. vivax infections to monitor antibody kinetics over 9 months, during which no recurrent infections occurred. We assessed total IgG, IgG subclass and IgM levels to up to 52 P. vivax proteins every 2-4 weeks using a multiplexed Luminex® assay and identified protein-specific variation in antibody longevity. Mathematical modelling was used to generate the estimated half-life of antibodies, long-, and short-lived antibody-secreting cells. RESULTS: Generally, an increase in antibody level was observed within 1-week post symptomatic infection, followed by an exponential decay of different rates. We observed mostly IgG1 dominance and IgG3 sub-dominance in this population. IgM responses followed similar kinetic patterns to IgG, with some proteins unexpectedly inducing long-lived IgM responses. We also monitored antibody responses against 27 IgG-immunogenic antigens in 30 asymptomatic individuals from a similar region. Our results demonstrate that most antigens induced robust and long-lived total IgG responses following asymptomatic infections in the absence of (detected) boosting infections. CONCLUSIONS: Our work provides new insights into the development and maintenance of naturally acquired immunity to P. vivax and will guide the potential use of serology to indicate immune status and/or identify populations at risk.


Assuntos
Malária Vivax , Malária , Anticorpos Antiprotozoários , Antígenos de Protozoários , Humanos , Cinética , Malária Vivax/epidemiologia , Plasmodium vivax , Proteínas de Protozoários , Tailândia/epidemiologia
16.
C R Biol ; 345(2): 91-133, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36847467

RESUMO

Vivax malaria is an infectious disease caused by Plasmodium vivax, a parasitic protozoan transmitted by female Anopheline mosquitoes. Historically, vivax malaria has often been regarded as a benign self-limiting infection due to the observation of low parasitemia in Duffy-positive patients in endemic transmission areas and the virtual absence of infections in Duffy-negative individuals in Sub Saharan Africa. However, the latest estimates show that the burden of the disease is not decreasing in many countries and cases of vivax infections in Duffy-negative individuals are increasingly reported throughout Africa. This raised questions about the accuracy of diagnostics and the evolution of interactions between humans and parasites. For a long time, our knowledge on P. vivax biology has been hampered due to the limited access to biological material and the lack of robust in vitro culture methods. Consequently, little is currently known about P. vivax blood stage invasion mechanisms. The introduction of omics technologies with novel and accessible techniques such as third generation sequencing and RNA sequencing at single cell level, two-dimensional electrophoresis, liquid chromatography, and mass spectrometry, has progressively improved our understanding of P. vivax genetics, transcripts, and proteins. This review aims to provide broad insights into P. vivax invasion mechanisms generated by genomics, transcriptomics, and proteomics and to illustrate the importance of integrated multi-omics studies.


Le paludisme à Plasmodium vivax est une maladie infectieuse causée par un parasite protozoaire Plasmodium vivax, transmis par les moustiques Anophèle femelles. Historiquement, le paludisme à P. vivax a souvent été considéré comme une infection bénigne en raison de l'observation d'une faible parasitémie chez les patients Duffy-positifs dans les zones d'endémie et de la quasi-absence d'infections chez les individus Duffy-négatifs vivant majoritairement en Afrique subsaharienne. Cependant, les dernières estimations montrent que le poids de la maladie ne diminue pas dans de nombreux pays et que des cas d'infections à P. vivax chez des individus Duffy-négatifs sont de plus en plus souvent observés en Afrique. Cela soulève des interrogations sur la précision des diagnostics et l'évolution des interactions hôte-parasite. Pendant longtemps, nos connaissances sur la biologie de P. vivax ont été entravées par un accès limité au matériel biologique et un manque de méthodes robustes pour la culture in vitro. Par conséquent, nous n'avons encore que peu d'informations concernant les mécanismes d'invasion des stades sanguins de P. vivax. L'introduction des technologies dites « omiques ¼, avec le développement de techniques innovantes et abordables telles que le séquençage d'ADN de troisième génération, le séquençage ARN à l'échelle de la cellule « single-cell ¼, l'électrophorèse bidimensionnelle, la chromatographie liquide et la spectrométrie de masse, a progressivement amélioré notre compréhension des gènes, des transcrits et des protéines de P. vivax. Cette revue a non seulement pour but de fournir un aperçu général des mécanismes d'invasion de P. vivax acquis grâce aux techniques génomiques, transcriptomiques et protéomiques mais également d'illustrer l'importance de la complémentarité de ces approches.


Assuntos
Malária Vivax , Plasmodium vivax , Animais , Humanos , Feminino , Plasmodium vivax/genética , Plasmodium vivax/metabolismo , Malária Vivax/genética , Malária Vivax/epidemiologia , Malária Vivax/parasitologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , África
17.
Microbiol Spectr ; 9(3): e0110921, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34878303

RESUMO

Coinfection with Plasmodium falciparum and helminths may impact the immune response to these parasites because they induce different immune profiles. We studied the effects of coinfections on the antibody profile in a cohort of 715 Mozambican children and adults using the Luminex technology with a panel of 16 antigens from P. falciparum and 11 antigens from helminths (Ascaris lumbricoides, hookworm, Trichuris trichiura, Strongyloides stercoralis, and Schistosoma spp.) and measured antigen-specific IgG and total IgE responses. We compared the antibody profile between groups defined by P. falciparum and helminth previous exposure (based on serology) and/or current infection (determined by microscopy and/or qPCR). In multivariable regression models adjusted by demographic, socioeconomic, water, and sanitation variables, individuals exposed/infected with P. falciparum and helminths had significantly higher total IgE and antigen-specific IgG levels, magnitude (sum of all levels) and breadth of response to both types of parasites compared to individuals exposed/infected with only one type of parasite (P ≤ 0.05). There was a positive association between exposure/infection with P. falciparum and exposure/infection with helminths or the number of helminth species, and vice versa (P ≤ 0.001). In addition, children coexposed/coinfected tended (P = 0.062) to have higher P. falciparum parasitemia than those single exposed/infected. Our results suggest that an increase in the antibody responses in coexposed/coinfected individuals may reflect higher exposure and be due to a more permissive immune environment to infection in the host. IMPORTANCE Coinfection with Plasmodium falciparum and helminths may impact the immune response to these parasites because they induce different immune profiles. We compared the antibody profile between groups of Mozambican individuals defined by P. falciparum and helminth previous exposure and/or current infection. Our results show a significant increase in antibody responses in individuals coexposed/coinfected with P. falciparum and helminths in comparison with individuals exposed/infected with only one of these parasites, and suggest that this increase is due to a more permissive immune environment to infection in the host. Importantly, this study takes previous exposure into account, which is particularly relevant in endemic areas where continuous infections imprint and shape the immune system. Deciphering the implications of coinfections deserves attention because accounting for the real interactions that occur in nature could improve the design of integrated disease control strategies.


Assuntos
Anticorpos Anti-Helmínticos/sangue , Anticorpos Antiprotozoários/sangue , Coinfecção/imunologia , Helmintos/imunologia , Plasmodium falciparum/imunologia , Adolescente , Adulto , Animais , Anticorpos Anti-Helmínticos/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Helmintos/imunologia , Antígenos de Protozoários/imunologia , Criança , Pré-Escolar , Feminino , Helmintíase/imunologia , Helmintíase/patologia , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Malária Falciparum/imunologia , Malária Falciparum/patologia , Masculino , Moçambique , Carga Parasitária , Solo/parasitologia , Adulto Jovem
18.
Front Microbiol ; 12: 643501, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276583

RESUMO

Thailand is aiming for malaria elimination by the year 2030. However, the high proportion of asymptomatic infections and the presence of the hidden hypnozoite stage of Plasmodium vivax are impeding these efforts. We hypothesized that a validated surveillance tool utilizing serological markers of recent exposure to P. vivax infection could help to identify areas of ongoing transmission. The objective of this exploratory study was to assess the ability of P. vivax serological exposure markers to detect residual transmission "hot-spots" in Western Thailand. Total IgG levels were measured against a panel of 23 candidate P. vivax serological exposure markers using a multiplexed bead-based assay. A total of 4,255 plasma samples from a cross-sectional survey conducted in 2012 of endemic areas in the Kanchanaburi and Ratchaburi provinces were assayed. We compared IgG levels with multiple epidemiological factors that are associated with an increased risk of P. vivax infection in Thailand, including age, gender, and spatial location, as well as Plasmodium infection status itself. IgG levels to all proteins were significantly higher in the presence of a P. vivax infection (n = 144) (T-test, p < 0.0001). Overall seropositivity rates varied from 2.5% (PVX_097625, merozoite surface protein 8) to 16.8% (PVX_082670, merozoite surface protein 7), with 43% of individuals seropositive to at least 1 protein. Higher IgG levels were associated with older age (>18 years, p < 0.05) and males (17/23 proteins, p < 0.05), supporting the paradigm that men have a higher risk of infection than females in this setting. We used a Random Forests algorithm to predict which individuals had exposure to P. vivax parasites in the last 9-months, based on their IgG antibody levels to a panel of eight previously validated P. vivax proteins. Spatial clustering was observed at the village and regional level, with a moderate correlation between PCR prevalence and sero-prevalence as predicted by the algorithm. Our data provides proof-of-concept for application of such surrogate markers as evidence of recent exposure in low transmission areas. These data can be used to better identify geographical areas with asymptomatic infection burdens that can be targeted in elimination campaigns.

19.
Open Forum Infect Dis ; 8(6): ofab228, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34159216

RESUMO

To achieve malaria elimination, new tools are required to explicitly target Plasmodium vivax. Recently, a novel panel of P. vivax proteins were identified and validated as serological markers for detecting recent exposure to P. vivax within the last 9 months. In order to improve the sensitivity and specificity of these markers, immunoglobulin M (IgM) in addition to immunoglobulin G (IgG) antibody responses were compared with a down-selected panel of 20 P. vivax proteins. IgM was tested using archival plasma samples from observational cohort studies conducted in malaria-endemic regions of Thailand and Brazil. IgM responses to these proteins generally had poorer classification performance than IgG.

20.
Front Microbiol ; 12: 684005, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34108954

RESUMO

We review the role of signaling pathways in regulation of the key processes of merozoite egress and red blood cell invasion by Plasmodium falciparum and, in particular, the importance of the second messengers, cAMP and Ca2+, and cyclic nucleotide dependent kinases. cAMP-dependent protein kinase (PKA) is comprised of cAMP-binding regulatory, and catalytic subunits. The less well conserved cAMP-binding pockets should make cAMP analogs attractive drug leads, but this approach is compromised by the poor membrane permeability of cyclic nucleotides. We discuss how the conserved nature of ATP-binding pockets makes ATP analogs inherently prone to off-target effects and how ATP analogs and genetic manipulation can be useful research tools to examine this. We suggest that targeting PKA interaction partners as well as substrates, or developing inhibitors based on PKA interaction sites or phosphorylation sites in PKA substrates, may provide viable alternative approaches for the development of anti-malarial drugs. Proximity of PKA to a substrate is necessary for substrate phosphorylation, but the P. falciparum genome encodes few recognizable A-kinase anchor proteins (AKAPs), suggesting the importance of PKA-regulatory subunit myristylation and membrane association in determining substrate preference. We also discuss how Pf14-3-3 assembles a phosphorylation-dependent signaling complex that includes PKA and calcium dependent protein kinase 1 (CDPK1) and how this complex may be critical for merozoite invasion, and a target to block parasite growth. We compare altered phosphorylation levels in intracellular and egressed merozoites to identify potential PKA substrates. Finally, as host PKA may have a critical role in supporting intracellular parasite development, we discuss its role at other stages of the life cycle, as well as in other apicomplexan infections. Throughout our review we propose possible new directions for the therapeutic exploitation of cAMP-PKA-signaling in malaria and other diseases caused by apicomplexan parasites.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...