Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
J Immunol Methods ; 532: 113728, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39059746

RESUMO

Immunoassay relies on antibodies, but traditional antibodies such as monoclonal antibody (mAb) require animal immunization and complex procedures. Single-chain variable fragment (scFv) becomes a potential alternative to mAb with advantages of the low cost, rapid and easy prepared. In the present study, we prepared scFvs against dihydroartemisinin (DHA) based on E. coli and HEK293T cell expression system, named MBP-scFv and scFv-Fc, respectively. Their properties were compared with the parent mAb. The calculated affinity constants of mAb, MBP-scFv and scFv-Fc were 2.1 × 108 L mol-1, 2.2 × 107 L mol-1 and 1.6 × 108 L mol-1, respectively. The half inhibitory concentration (IC50) of mAb, MBP-scFv and scFv-Fc were 1.16 ng mL-1, 2.15 ng mL-1 and 6.57 ng mL-1, respectively. Both the scFv showed less sensitive than the mAb based on the IC50. The cross-reactivities of MBP-scFv for artemisinin and artesunate exhibited similarities to the mAb, yet the cross-reactivities of scFv-Fc for these compounds exceeded those of the mAb significantly. The stability of the scFvs was ascertained to be maintained for over 5 days at room temperature, and for more than a month at both 4 °C and - 20 °C. After that, the indirect competitive enzyme-linked immunosorbent assays (icELISAs) based on the scFv from E. coli were used to detect the DHA content in eight drug samples, and the result was consistent with ultra-performance liquid chromatography simultaneously. Although scFv can be used for quantitative determination of drugs, but it still cannot completely replace mAb in immunoassay without evolution and modification.


Assuntos
Anticorpos Monoclonais , Artemisininas , Anticorpos de Cadeia Única , Artemisininas/imunologia , Artemisininas/farmacologia , Anticorpos de Cadeia Única/imunologia , Humanos , Anticorpos Monoclonais/imunologia , Células HEK293 , Afinidade de Anticorpos , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/imunologia , Reações Cruzadas/imunologia , Antimaláricos/imunologia , Ensaio de Imunoadsorção Enzimática , Especificidade de Anticorpos
2.
Comput Biol Chem ; 92: 107495, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33940529

RESUMO

The development of the anti-malaria vaccine holds a promising future in malaria control. One of the anti-malaria vaccine strategies known as the transmission-blocking vaccine (TBV) is to inhibit the parasite transmission between humans and mosquitoes by targeting the parasite gametocyte. Previously, we found that P48/45 included in the 6-Cysteine protein family shared by Plasmodium sp. We also detected vaccine properties possessed by all human-infecting Plasmodium and could be used as a cross-species anti-malaria vaccine. In this study, we investigated the efficacy of P48/45 through the ancestral and consensus reconstruction approach. P48/45 phylogenetic and time tree analysis was done by RAXML and BEAST2. GRASP server and Ugene software were used to reconstruct ancestral and consensus sequences, respectively. The protein structural prediction was made by using a psipred and Rosetta program. Each protein characteristic of P48/45 was analyzed by assessing hydrophobicity and Post-Translational Modification sites. Meanwhile, the Epitope sequence for B-cell, T-cell, and HLA was determined using an immunoinformatics approach. Lastly, molecular docking simulation was done to determine native binding interactions of P48/45-P230. The result showed a distinct protein characteristic of ancestral and consensus sequences. The immunogenicity analysis revealed the number of epitopes in the ancestral sequence is greater than the consensus sequence. The study also found a conserved epitope located in the binding site and consists of specific Post-Translational Modification sites. Hence, our research provides detailed insight into ancestral and consensus P48/45 efficacy for the cross-species anti-malaria vaccine.


Assuntos
Antimaláricos/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Antimaláricos/química , Antimaláricos/farmacologia , Sequência Consenso , Humanos , Vacinas Antimaláricas/química , Vacinas Antimaláricas/farmacologia , Filogenia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/genética , Proteínas de Protozoários/efeitos dos fármacos , Proteínas de Protozoários/genética , Software
3.
J Exp Med ; 218(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33830176

RESUMO

Antimalarial antibody responses are essential for mediating the clearance of Plasmodium parasite-infected RBCs from infected hosts. However, the rapid appearance of large numbers of plasmablasts in Plasmodium-infected hosts can suppress the development and function of durable humoral immunity. Here, we identify that the formation of plasmablast populations in Plasmodium-infected mice is mechanistically linked to both hemolysis-induced exposure of phosphatidylserine on damaged RBCs and inflammatory cues. We also show that virus and Trypanosoma infections known to trigger hemolytic anemia and high-grade inflammation also induce exuberant plasmablast responses. The induction of hemolysis or administration of RBC membrane ghosts increases plasmablast differentiation. The phosphatidylserine receptor Axl is critical for optimal plasmablast formation, and blocking phosphatidylserine limits plasmablast expansions and reduces Plasmodium parasite burden in vivo. Our findings support that strategies aimed at modulating polyclonal B cell activation and phosphatidylserine exposure may improve immune responses against Plasmodium parasites and potentially other infectious diseases that are associated with anemia.


Assuntos
Diferenciação Celular/imunologia , Hemólise/imunologia , Fosfatidilserinas/imunologia , Plasmócitos/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antimaláricos/imunologia , Linfócitos B/imunologia , Linfócitos B/parasitologia , Células Cultivadas , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Imunidade Humoral/imunologia , Malária/imunologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/parasitologia , Plasmodium yoelii/imunologia
4.
PLoS Pathog ; 17(2): e1009288, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33529242

RESUMO

Immunity against malaria depends on germinal center (GC)-derived antibody responses that are orchestrated by T follicular helper (TFH) cells. Emerging data show that the regulatory cytokine IL-10 plays an essential role in promoting GC B cell responses during both experimental malaria and virus infections. Here we investigated the cellular source and temporal role of IL-10, and whether IL-10 additionally signals to CD4 T-cells to support anti-Plasmodium humoral immunity. Distinct from reports of virus infection, we found that IL-10 was expressed by conventional, Foxp3-negative effector CD4 T cells and functioned in a B cell-intrinsic manner only during the first 96 hours of Plasmodium infection to support humoral immunity. The critical functions of IL-10 manifested only before the orchestration of GC responses and were primarily localized outside of B cell follicles. Mechanistically, our studies showed that the rapid and transient provision of IL-10 promoted B cell expression of anti-apoptotic factors, MHC class II, CD83, and cell-cell adhesion proteins that are essential for B cell survival and interaction with CD4 T cells. Together, our data reveal temporal features and mechanisms by which IL-10 critically supports humoral immunity during blood-stage Plasmodium infection, information that may be useful for developing new strategies designed to lessen the burden of malaria.


Assuntos
Formação de Anticorpos/imunologia , Antimaláricos/imunologia , Linfócitos T CD4-Positivos/imunologia , Interleucina-10/metabolismo , Ativação Linfocitária/imunologia , Malária/imunologia , Plasmodium yoelii/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Malária/metabolismo , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
5.
Curr Rheumatol Rev ; 17(2): 141-152, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33121412

RESUMO

The new coronavirus infection (Covid-19) is a pandemic that has affected the whole world and progresses with high morbidity and mortality. It has a high contagion rate and a course capable of rapid lung involvement with severe acute respiratory distress syndrome (ARDS) and pulmonary insufficiency. A severe clinical picture develops as a result of a "perfect cytokine storm" which results from possible immunological mechanisms triggered by the viral infection. Immune system dysregulation and possible autoinflammatory and autoimmune mechanisms are responsible for a higher amount of cytokines release from immune cells. Although no clear treatment of Covid-19 infection has emerged yet, it is argued that some disease-modifying anti-rheumatic drugs (DMARDs) may be effective in addition to anti-viral treatments. These drugs (anti-malarial drugs, colchicum dispert, biologics) have been well known to rheumatologists for years because they are used in the treatment of many inflammatory rheumatologic diseases. Another important issue is whether DMARDs, which can cause severe immunosuppression, pose a risk for Covid-19 infection and whether they have been discontinued beforehand. Although there are insufficient data on this subject, considering the risk of disease reactivation, patients may continue their DMARDs treatment under the supervision of a rheumatologist. In this article, the possible immunological mechanisms in the pathogenesis of Covid-19 infection and the efficacy and safety of various DMARDs used in the treatment are discussed from a rheumatologist's perspective in the light of recent literature data.


Assuntos
Antirreumáticos/administração & dosagem , Tratamento Farmacológico da COVID-19 , COVID-19/epidemiologia , Síndrome da Liberação de Citocina/tratamento farmacológico , Síndrome da Liberação de Citocina/epidemiologia , Reumatologistas/tendências , Corticosteroides/administração & dosagem , Corticosteroides/imunologia , Antimaláricos/administração & dosagem , Antimaláricos/imunologia , Antirreumáticos/imunologia , COVID-19/imunologia , Síndrome da Liberação de Citocina/imunologia , Humanos , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/imunologia
6.
JCI Insight ; 5(22)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33048842

RESUMO

Plasma antimalarial Ab can mediate antiparasite immunity but has not previously been characterized at the molecular level. Here, we develop an innovative strategy to characterize humoral responses by integrating profiles of plasma immunoglobulins (IGs) or Abs with those expressed on B cells as part of the B cell receptor. We applied this strategy to define plasma IG and to determine variable (V) gene usage after vaccination with the Plasmodium falciparum zygote antigen Pfs25. Using proteomic tools coupled with bulk immunosequencing data, we determined human antigen-binding fragment [F(ab')2] peptide sequences from plasma IG of adults who received 4 doses of Pfs25-EPA/Alhydrogel. Specifically, Pfs25 antigen-specific F(ab')2 peptides (Pfs25-IG) were aligned to cDNA sequences of IG heavy (IGH) chain complementarity determining region 3 from a data set generated by total peripheral B cell immunosequencing of the entire vaccinated population. IGHV4 was the most commonly identified IGHV subgroup of Pfs25-IG, a pattern that was corroborated by V heavy/V light chain sequencing of Pfs25-specific single B cells from 5 vaccinees and by matching plasma Pfs25-IG peptides and V-(D)-J sequences of Pfs25-specific single B cells from the same donor. Among 13 recombinant human mAbs generated from IG sequences of Pfs25-specific single B cells, a single IGHV4 mAb displayed strong neutralizing activity, reducing the number of P. falciparum oocysts in infected mosquitoes by more than 80% at 100 µg/mL. Our approach characterizes the human plasma Ab repertoire in response to the Pfs25-EPA/Alhydrogel vaccine and will be useful for studying circulating Abs in response to other vaccines as well as those induced during infections or autoimmune disorders.


Assuntos
Anticorpos Antiprotozoários/sangue , Antimaláricos/imunologia , Linfócitos B/imunologia , Imunoglobulinas/sangue , Malária Falciparum/sangue , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos , Adolescente , Adulto , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Antimaláricos/administração & dosagem , Ensaios Clínicos como Assunto , Feminino , Humanos , Imunoglobulinas/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Masculino , Pessoa de Meia-Idade , Vacinação , Adulto Jovem
7.
Immunity ; 53(4): 733-744.e8, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32946741

RESUMO

Discovering potent human monoclonal antibodies (mAbs) targeting the Plasmodium falciparum circumsporozoite protein (PfCSP) on sporozoites (SPZ) and elucidating their mechanisms of neutralization will facilitate translation for passive prophylaxis and aid next-generation vaccine development. Here, we isolated a neutralizing human mAb, L9 that preferentially bound NVDP minor repeats of PfCSP with high affinity while cross-reacting with NANP major repeats. L9 was more potent than six published neutralizing human PfCSP mAbs at mediating protection against mosquito bite challenge in mice. Isothermal titration calorimetry and multiphoton microscopy showed that L9 and the other most protective mAbs bound PfCSP with two binding events and mediated protection by killing SPZ in the liver and by preventing their egress from sinusoids and traversal of hepatocytes. This study defines the subdominant PfCSP minor repeats as neutralizing epitopes, identifies an in vitro biophysical correlate of SPZ neutralization, and demonstrates that the liver is an important site for antibodies to prevent malaria.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/imunologia , Antimaláricos/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Adolescente , Adulto , Animais , Linhagem Celular , Linhagem Celular Tumoral , Epitopos/imunologia , Feminino , Células HEK293 , Hepatócitos/imunologia , Hepatócitos/parasitologia , Humanos , Fígado/imunologia , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Vacinas Antimaláricas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Adulto Jovem
9.
Parasite Immunol ; 42(4): e12702, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32020650

RESUMO

AIMS: Schistosomiasis and malaria are endemic in sub-Saharan Africa where Schistosoma haematobium (Sh) and Plasmodium falciparum (Pf) coinfections are thus frequent. We explored the effect of Sh infection on antibody responses directed to Pf merozoite antigens and on malaria susceptibility in Beninese children. METHODS AND RESULTS: A total of 268 children were followed during a malaria transmission season. Detection of Pf infection was performed by microscopy and rapid diagnostic tests. Sh infection was determined in urine by microscopy. Antimalarial antibody, cytokine and HLA-G concentrations were quantified by ELISA. The expression of HLA-G receptors by immune cells was assessed by flow cytometry. Children infected by Sh had higher concentrations of IgG1 directed to MSP3 and GLURPR0 , IgG2 directed to GLURPR0 and IgG3 directed to MSP3, GLURPR0 and GLURPR2 and have lower Pf densities than those uninfected by Sh. No difference in cytokine and HLA-G concentrations was observed between Sh egg carriers and non-carriers. CONCLUSION: Schistosoma haematobium modulates host immune responses directed to Pf antigens. The absence of immune downregulation usually observed during helminth infections is surprising in our study. We hypothesize that the stage of Sh development could partly explain the immune pathways leading to increased antibody levels that favour better control of Pf parasitemia.


Assuntos
Anticorpos Antiprotozoários/sangue , Antimaláricos/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Schistosoma haematobium/imunologia , Esquistossomose Urinária/imunologia , Animais , Antígenos de Protozoários/imunologia , Antimaláricos/uso terapêutico , Benin , Criança , Pré-Escolar , Coinfecção/parasitologia , Citocinas/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Malária Falciparum/complicações , Masculino , Esquistossomose Urinária/complicações , Esquistossomose Urinária/tratamento farmacológico
10.
Virulence ; 11(1): 88-103, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31900030

RESUMO

Individuals growing up in malaria endemic areas gradually develop protection against clinical malaria and passive transfer experiments in humans have demonstrated that this protection is mediated in part by protective antibodies. However, neither the target antigens, specific effector mechanisms, nor the role of continual parasite exposure have been elucidated, which complicates vaccine development. Progress has been made in defining the innate signaling pathways activated by parasite components, including DNA, RNA, hemozoin, and phospholipids, which initiate the immune response and will be the focus of this review. The challenge that remains within the field is to understand the role of these early responses in the development of protective adaptive responses that clear iRBC and block merozoite invasion so that optimal vaccines and therapeutics may be produced.


Assuntos
Eritrócitos/parasitologia , Imunidade/imunologia , Estágios do Ciclo de Vida/imunologia , Merozoítos/imunologia , Parasitos/imunologia , Imunidade Adaptativa/imunologia , Animais , Antígenos de Protozoários/imunologia , Antimaláricos/imunologia , Células Dendríticas , Hemeproteínas , Humanos , Células Matadoras Naturais , Malária/imunologia , Malária/prevenção & controle , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Fosfolipídeos/imunologia , Pigmentos Biológicos/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/imunologia
11.
Vaccine ; 37(32): 4468-4476, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31262583

RESUMO

In both preclinical animal studies and human clinical trials, adult females tend to develop greater adaptive immune responses than males following receipt of either viral or bacterial vaccines. While there is currently no approved malaria vaccine, several anti-sporozoite vaccines, including RTS,S/AS01 and attenuated sporozoite vaccines, are in development, but the impact of sex and age on their efficacy remains undefined. To examine sex differences in the efficacy of anti-sporozoite stage malaria vaccination, adult (10 weeks of age) or juvenile (11 days of age) male and female C3H mice were twice vaccinated with irradiated transgenic Plasmodium berghei sporozoites expressing the P. falciparum circumsporozoite (CSP) protein and 45 days post boost vaccination, mice were challenged with transgenic P. berghei via mosquito bite or intradermal challenge. Immunization with irradiated sporozoites resulted in greater protection against challenge in adult females, which was associated with greater anti-CSP antibody production and avidity, as well as greater hepatic, but not splenic, CD8+ T cell IFNƴ production in adult females than adult males. No sex differences in adaptive immune responses or protection were observed in mice vaccinated prior to puberty, suggesting a role for sex steroid hormones. Depletion of testosterone in males increased, whereas rescue of testosterone decreased, anti-CSP antibody production, the number of antigen-specific CD8+ T cells isolated from the liver, and protection following parasite challenge. Conversely, depletion of sex steroids in female mice did not alter vaccine-induced responses or protection following challenge. These data suggest that elevated testosterone concentrations in males reduce adaptive immunity and contribute to sex differences in malaria vaccine efficacy.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Esporozoítos/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antimaláricos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Imunização/métodos , Fígado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Plasmodium berghei/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Baço/imunologia , Vacinação/métodos
12.
Int J Parasitol Drugs Drug Resist ; 8(3): 451-458, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30396012

RESUMO

Both vaccine and therapeutic approaches to malaria are based on conventional paradigms; whole organism or single antigen epitope-based vaccines administered with or without an adjuvant, and chemotherapeutics (anti-malaria drugs) that are toxic to the parasite. Two major problems that limit the effectiveness of these approaches are i) high levels of antigenic variation within parasite populations rendering vaccination efficacy against all variants difficult, and ii) the capacity of the parasite to quickly evolve resistance to drugs. We describe a new approach to both protection from and treatment of malaria parasites that involves the direct stimulation of the host innate immune response through the administration of a Toll-Like Receptor-2 (TLR2) agonist. The activity of PEG-Pam2Cys against the hepatocytic stages, erythrocytic stages and gametocytes of the rodent malaria parasite Plasmodium yoelii was investigated in laboratory mice. We show that administration of PEG-Pam2Cys, a soluble form of the TLR2 agonist S-[2,3-bis(palmitoyloxy)propyl] cysteine (Pam2Cys), significantly and dramatically reduces the numbers of malaria parasites that grow in the livers of mice following subsequent challenge with sporozoites. We also show that treatment can also clear parasites from the liver when administered subsequent to the establishment of infection. Finally, PEG-Pam2Cys can reduce the numbers of mosquitoes that are infected, and the intensity of their infection, following blood feeding on gametocytaemic mice. These results suggest that this compound could represent a novel liver stage anti-malarial that can be used both for the clearance of parasites following exposure and for the prevention of the establishment of infection.


Assuntos
Antimaláricos/uso terapêutico , Imunoterapia/métodos , Lipopeptídeos/uso terapêutico , Malária/tratamento farmacológico , Malária/prevenção & controle , Plasmodium yoelii/efeitos dos fármacos , Esporozoítos/efeitos dos fármacos , Animais , Anticorpos Antiprotozoários/sangue , Antimaláricos/administração & dosagem , Antimaláricos/imunologia , Terapia Combinada/métodos , Culicidae/efeitos dos fármacos , Culicidae/parasitologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Imunidade Inata/efeitos dos fármacos , Lipopeptídeos/administração & dosagem , Lipopeptídeos/imunologia , Fígado/efeitos dos fármacos , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Camundongos , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/imunologia , Receptor 2 Toll-Like/agonistas
13.
Trends Parasitol ; 34(10): 843-860, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30122551

RESUMO

Despite the recent successes of artemisinin-based antimalarial drugs, many still die from severe malaria, and eradication efforts are hindered by the limited drugs currently available to target transmissible gametocyte parasites and liver-resident dormant Plasmodium vivax hypnozoites. Host-targeted therapy is a new direction for infectious disease drug development and aims to interfere with host molecules, pathways, or networks that are required for infection or that contribute to disease. Recent advances in our understanding of host pathways involved in parasite development and pathogenic mechanisms in severe malaria could facilitate the development of host-targeted interventions against Plasmodium infection and malaria disease. This review discusses new opportunities for host-targeted therapeutics for malaria and the potential to harness drug polypharmacology to simultaneously target multiple host pathways using a single drug intervention.


Assuntos
Antimaláricos/imunologia , Sistemas de Liberação de Medicamentos , Antimaláricos/farmacologia , Interações Hospedeiro-Parasita/efeitos dos fármacos , Humanos , Fatores Imunológicos/farmacologia
14.
Lancet Infect Dis ; 18(9): 969-982, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30061051

RESUMO

BACKGROUND: Pfs25H-EPA is a protein-protein conjugate transmission-blocking vaccine against Plasmodium falciparum that is safe and induces functional antibodies in malaria-naive individuals. In this field trial, we assessed Pfs25H-EPA/Alhydrogel for safety and functional immunogenicity in Malian adults. METHODS: This double-blind, randomised, comparator-controlled, dose-escalation trial in Bancoumana, Mali, was done in two staggered phases, an initial pilot safety assessment and a subsequent main phase. Healthy village residents aged 18-45 years were eligible if they had normal laboratory results (including HIV, hepatitis B, hepatitis C tests) and had not received a previous malaria vaccine or recent immunosuppressive drugs, vaccines, or blood products. Participants in the pilot safety cohort and the main cohort were assigned (1:1) by block randomisation to a study vaccine group. Participants in the pilot safety cohort received two doses of Pfs25H-EPA/Alhydrogel 16 µg or Euvax B (comparator vaccine), and participants in the main cohort received Pfs25H-EPA/Alhydrogel 47 µg or comparator vaccine (Euvax B for the first, second, and third vaccinations and Menactra for the fourth vaccination). Participants and investigators were masked to group assignment, and randomisation codes in sealed envelopes held by a site pharmacist. Vials with study drug for injection were covered by opaque tape and labelled with a study identification number. Group assignments were unmasked at final study visit. The primary outcomes were safety and tolerability for all vaccinees. The secondary outcome measure was immunogenicity 14 days after vaccination in the per-protocol population, as confirmed by the presence of antibodies against Pfs25H measured by ELISA IgG and antibody functionality assessed by standard membrane feeding assays and by direct skin feeding assays. This trial is registered with ClinicalTrials.gov, number NCT01867463. FINDINGS: Between May 15, and Jun 16, 2013, 230 individuals were screened for eligibility. 20 individuals were enrolled in the pilot safety cohort; ten participants were assigned to receive Pfs25H-EPA/Alhydrogel 16 µg, and ten participants were assigned to receive comparator vaccine. 100 individuals were enrolled in the main cohort; 50 participants were assigned to receive Pfs25H-EPA/Alhydrogel 47 µg, and 50 participants were assigned to receive comparator vaccine. Compared with comparator vaccinees, Pfs25H vaccinees had more solicited adverse events (137 events vs 86 events; p=0·022) and treatment-related adverse events (191 events vs 126 events, p=0·034), but the number of other adverse events did not differ between study vaccine groups (792 vs 683). Pfs25H antibody titres increased with each dose, with a peak geometric mean of 422·3 ELISA units (95% CI 290-615) after the fourth dose, but decreased relatively rapidly thereafter, with a half-life of 42 days for anti-Pfs25H and 59 days for anti-EPA (median ratio of titres at day 600 to peak, 0·19 for anti-Pfs25H vs 0·29 for anti-EPA; p=0·009). Serum transmission-reducing activity was greater for Pfs25H than for comparator vaccine after the fourth vaccine dose (p<0·001) but not after the third dose (p=0·09). Repeated direct skin feeds were well tolerated, but the number of participants who infected at least one mosquito did not differ between Pfs25H and comparator vaccinees after the fourth dose (p=1, conditional exact). INTERPRETATION: Pfs25H-EPA/Alhydrogel was well tolerated and induced significant serum activity by standard membrane feeding assays but transmission blocking activity was not confirmed by weekly direct skin feed. This activity required four doses, and titres decreased rapidly after the fourth dose. Alternative antigens or combinations should be assessed to improve activity. FUNDING: Division of Intramural Research, National Institute of Allergy and Infectious Diseases.


Assuntos
Antimaláricos/imunologia , Antimaláricos/toxicidade , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/toxicidade , Malária Falciparum/tratamento farmacológico , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/toxicidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Antimaláricos/uso terapêutico , Método Duplo-Cego , Feminino , Humanos , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/epidemiologia , Masculino , Mali/epidemiologia , Pessoa de Meia-Idade , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/uso terapêutico
15.
Infect Immun ; 86(7)2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29735521

RESUMO

Long-lasting and sterile homologous protection against malaria can be achieved by the exposure of malaria-naive volunteers under chemoprophylaxis to Plasmodium falciparum-infected mosquitoes (chemoprophylaxis and sporozoite [CPS] immunization). While CPS-induced antibodies neutralize sporozoite infectivity in vitro and in vivo, antibody-mediated effector mechanisms are still poorly understood. Here, we investigated whether complement contributes to CPS-induced preerythrocytic immunity. Sera collected before and after CPS immunization in the presence of active or inactive complement were assessed for the recognition of homologous NF54 and heterologous NF135.C10 sporozoites, complement fixation, sporozoite lysis, and possible subsequent effects on in vitro sporozoite infectivity in human hepatocytes. CPS immunization induced sporozoite-specific IgM (P < 0.0001) and IgG (P = 0.001) antibodies with complement-fixing capacities (P < 0.0001). Sporozoite lysis (P = 0.017), traversal (P < 0.0001), and hepatocyte invasion inhibition (P < 0.0001) by CPS-induced antibodies were strongly enhanced in the presence of active complement. Complement-mediated invasion inhibition in the presence of CPS-induced antibodies negatively correlated with cumulative parasitemia during CPS immunizations (P = 0.013). While IgG antibodies similarly recognized homologous and heterologous sporozoites, IgM binding to heterologous sporozoites was reduced (P = 0.023). Although CPS-induced antibodies did not differ in their abilities to fix complement, lyse sporozoites, or inhibit the traversal of homologous and heterologous sporozoites, heterologous sporozoite invasion was more strongly inhibited in the presence of active complement (P = 0.008). These findings demonstrate that CPS-induced antibodies have complement-fixing activity, thereby significantly further enhancing the functional inhibition of homologous and heterologous sporozoite infectivity in vitro The combined data highlight the importance of complement as an additional immune effector mechanism in preerythrocytic immunity after whole-parasite immunization against Plasmodium falciparum malaria.


Assuntos
Formação de Anticorpos/fisiologia , Antimaláricos/imunologia , Antimaláricos/uso terapêutico , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/tratamento farmacológico , Malária Falciparum/imunologia , Esporozoítos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Humanos , Imunização , Esporozoítos/imunologia , Vacinação
16.
Front Immunol ; 9: 2926, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30631323

RESUMO

Plasmodium spp., the causative agent of malaria, caused 212 million infections in 2016 with 445,000 deaths, mostly in children. Adults acquire enough immunity to prevent clinical symptoms but never develop sterile immunity. The only vaccine for malaria, RTS,S, shows promising protection of a limited duration against clinical malaria in infants but no significant protection against severe disease. There is now abundant evidence that T cell functions are inhibited during malaria, which may explain why vaccine are not efficacious. Studies have now clearly shown that T cell immunity against malaria is subdued by multiple the immune regulatory receptors, in particular, by programmed cell-death-1 (PD-1). Given there is an urgent need for an efficacious malarial treatment, compounded with growing drug resistance, a better understanding of malarial immunity is essential. This review will examine molecular signals that affect T cell-mediated immunity against malaria.


Assuntos
Antimaláricos/imunologia , Imunidade Celular/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Adulto , Criança , Humanos , Lactente , Malária/imunologia , Malária/parasitologia , Plasmodium/imunologia , Plasmodium/fisiologia , Linfócitos T/parasitologia
17.
Acta Trop ; 159: 111-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27001144

RESUMO

Antibodies that impede the invasion of Plasmodium falciparum (P. falciparum) merozoites into erythrocytes play a critical role in anti-malarial immunity. The Growth Inhibition Assay (GIA) is an in vitro measure of the functional capacity of such antibodies to limit erythrocyte invasion and/or parasite growth. Up to now, it is unclear whether growth-inhibitory activity correlates with protection from clinical disease and there are inconsistent results from studies performed with GIA. Studies that have focused on the relationship between IgGs and their in vitro parasite Growth Inhibition Activity (GIAc) in infants aged less than two years old are rare. Here, we used clinical and parasitological data to precisely define symptomatic or asymptomatic infection with P. falciparum in groups of infants followed-up actively for 18 months post-natally. We quantified the levels of IgG1 and IgG3 directed to a panel of candidate P. falciparum vaccine antigens (AMA-1, MSP1, 2, 3 and GLURP) using ELISA and the functional activity of IgG was quantified using GIA. Data were then correlated with individuals' infection status. At 18 months of age, infants harbouring infections at the time of blood sampling had an average 19% less GIAc than those not infected (p=0.004, multivariate linear regression). GIAc decreased from 12 to 18 months of age (p=0.003, Wilcoxon matched pairs test). Antibody levels quantified at 18 months in infants were strongly correlated with their exposure to malarial infection, however GIAc was not correlated with malaria infectious status (asymptomatic and symptomatic groups). In conclusion, both infection status at blood draw and age influence parasite growth inhibition mediated by IgG in the GIA. Both factors must be taken into account when correlations between GIAc and anti-malarial protection or vaccine efficacy have to be made.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Antimaláricos/imunologia , Merozoítos/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Fatores Etários , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/sangue , Benin , Ensaio de Imunoadsorção Enzimática , Eritrócitos/parasitologia , Feminino , Humanos , Imunoglobulina G/sangue , Lactente , Recém-Nascido , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Masculino , Metalotioneína 3 , Proteínas do Tecido Nervoso/sangue
18.
Anal Bioanal Chem ; 408(22): 6003-8, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26873200

RESUMO

Artesunate is a frontline antimalarial drug for treating Plasmodium falciparum malaria. To produce specific antibodies to artesunate, the carboxyl group of artesunate was directly conjugated to carrier protein as the immunogen. A specific monoclonal antibody (mAb) 3D82G6 against artesunate was obtained by high-throughput screening of positive hybridoma clones. This monoclonal antibody had 4.0, 0.5, and 0.9 % cross reactivities with artemisinin, dihydroartemisinin, and artemether, respectively. A dipstick immunoassay was developed, and the indicator range for artesunate was 1000-2000 ng mL(-1). No interference was observed with artemisinin, dihydroartemisinin, artemether, and other commonly used antimalarial drugs for up to 20,000 ng mL(-1). The dipsticks were used for determination of artesunate contents in commercial drugs, and the results were agreeable with those determined by high-performance liquid chromatography. This dipstick, with its specificity and sensitivity for artesunate and simplicity to use, makes it a potential point-of-care device for rapid quality evaluation of artesunate-containing antimalarial drugs. Graphical Abstract Specific monoclonal antibody-based lateral flow dipstick for artesunate.


Assuntos
Anticorpos Imobilizados/química , Anticorpos Monoclonais/química , Antimaláricos/análise , Artemisininas/análise , Imunoensaio/métodos , Sistemas Automatizados de Assistência Junto ao Leito , Animais , Anticorpos Imobilizados/imunologia , Anticorpos Monoclonais/imunologia , Antimaláricos/imunologia , Artemisininas/imunologia , Artesunato , Coloide de Ouro/química , Humanos , Hibridomas , Limite de Detecção , Malária Falciparum/tratamento farmacológico , Camundongos , Fitas Reagentes/análise
19.
Elife ; 42015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25714922

RESUMO

Protection against malaria in humans can be achieved by repeated exposure to infected mosquito bites during prophylactic chloroquine treatment (chemoprophylaxis and sporozoites (CPS)). We established a new mouse model of CPS immunization to investigate the stage and strain-specificity of malaria immunity. Immunization with Plasmodium chabaudi by mosquito bite under chloroquine cover does not generate pre-erythrocytic immunity, which is acquired only after immunization with high sporozoite doses. Instead, CPS immunization by bite elicits long-lived protection against blood-stage parasites. Blood-stage immunity is effective against a virulent, genetically distinct strain of P. chabaudi. Importantly, if exposure to blood-stage parasitemia is extended, blood-stage parasites induce cross-stage immunity targeting pre-erythrocytic stages. We therefore show that CPS immunization can induce robust, long-lived heterologous blood-stage immunity, in addition to protection against pre-erythrocytic parasites following high dose sporozoite immunization. Cross-stage immunity elicited by blood-stage parasites may further enhance efficacy of this immunization regimen.


Assuntos
Cloroquina/imunologia , Eritrócitos/imunologia , Malária/imunologia , Plasmodium chabaudi/imunologia , Esporozoítos/imunologia , Animais , Antimaláricos/imunologia , Antimaláricos/farmacologia , Quimioprevenção/métodos , Cloroquina/farmacologia , Culicidae/imunologia , Culicidae/parasitologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunização/métodos , Insetos Vetores/imunologia , Insetos Vetores/parasitologia , Malária/parasitologia , Malária/prevenção & controle , Camundongos Endogâmicos C57BL , Parasitemia/tratamento farmacológico , Parasitemia/imunologia , Parasitemia/parasitologia , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium chabaudi/fisiologia , Fatores de Tempo
20.
Mediators Inflamm ; 2014: 728939, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25132735

RESUMO

Although the roles of CD8+ T cells and a major preerythrocytic antigen, the circumsporozoite (CS) protein, in contributing protective antimalaria immunity induced by radiation-attenuated sporozoites, have been shown by a number of studies, the extent to which these players contribute to antimalaria immunity is still unknown. To address this question, we have generated C57BL/6 (B6) transgenic (Tg) mice, expressing K(d) molecules under the MHC-I promoter, called MHC-I-K(d)-Tg mice. In this study, we first determined that a single immunizing dose of IrPySpz induced a significant level of antimalaria protective immunity in MHC-I-K(d)-Tg mice but not in B6 mice. Then, by depleting various T-cell subsets in vivo, we determined that CD8+ T cells are the main mediator of the protective immunity induced by IrPySpz. Furthermore, when we immunized (MHC-I-K(d)-Tg × CS-Tg) F1 mice with IrPySpz after crossing MHC-I-K(d)-Tg mice with PyCS-transgenic mice (CS-Tg), which are unable to mount PyCS-specific immunity, we found that IrPySpz immunization failed to induce protective antimalaria immunity in (MHC-I-K(d)-Tg × CS-Tg) F1 mice, thus indicating the absence of PyCS antigen-dependent immunity in these mice. These results indicate that protective antimalaria immunity induced by IrPySpz in MHC-I-K(d)-Tg mice is mediated by CS protein-specific, K(d)-restricted CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Esporozoítos/imunologia , Esporozoítos/metabolismo , Animais , Anopheles , Antimaláricos/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasmodium yoelii/imunologia , Plasmodium yoelii/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...