Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.949
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 178(1): 216-228.e21, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31204103

RESUMO

The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) is the leading target for next-generation vaccines against the disease-causing blood-stage of malaria. However, little is known about how human antibodies confer functional immunity against this antigen. We isolated a panel of human monoclonal antibodies (mAbs) against PfRH5 from peripheral blood B cells from vaccinees in the first clinical trial of a PfRH5-based vaccine. We identified a subset of mAbs with neutralizing activity that bind to three distinct sites and another subset of mAbs that are non-functional, or even antagonistic to neutralizing antibodies. We also identify the epitope of a novel group of non-neutralizing antibodies that significantly reduce the speed of red blood cell invasion by the merozoite, thereby potentiating the effect of all neutralizing PfRH5 antibodies as well as synergizing with antibodies targeting other malaria invasion proteins. Our results provide a roadmap for structure-guided vaccine development to maximize antibody efficacy against blood-stage malaria.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/imunologia , Eritrócitos/parasitologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Adolescente , Adulto , Animais , Sítios de Ligação , Proteínas de Transporte/imunologia , Reações Cruzadas/imunologia , Epitopos/imunologia , Feminino , Células HEK293 , Voluntários Saudáveis , Humanos , Malária Falciparum/parasitologia , Masculino , Merozoítos/fisiologia , Pessoa de Meia-Idade , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/imunologia , Coelhos , Ratos , Ratos Sprague-Dawley , Adulto Jovem
2.
Immunity ; 54(12): 2859-2876.e7, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34788599

RESUMO

Repeat antigens, such as the Plasmodium falciparum circumsporozoite protein (PfCSP), use both sequence degeneracy and structural diversity to evade the immune response. A few PfCSP-directed antibodies have been identified that are effective at preventing malaria infection, including CIS43, but how these repeat-targeting antibodies might be improved has been unclear. Here, we engineered a humanized mouse model in which B cells expressed inferred human germline CIS43 (iGL-CIS43) B cell receptors and used both vaccination and bioinformatic analysis to obtain variant CIS43 antibodies with improved protective capacity. One such antibody, iGL-CIS43.D3, was significantly more potent than the current best-in-class PfCSP-directed antibody. We found that vaccination with a junctional epitope peptide was more effective than full-length PfCSP at recruiting iGL-CIS43 B cells to germinal centers. Structure-function analysis revealed multiple somatic hypermutations that combinatorically improved protection. This mouse model can thus be used to understand vaccine immunogens and to develop highly potent anti-malarial antibodies.


Assuntos
Subpopulações de Linfócitos B/imunologia , Epitopos/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Transferência Adotiva , Animais , Anticorpos Antiprotozoários/metabolismo , Modelos Animais de Doenças , Epitopos/genética , Engenharia Genética , Humanos , Evasão da Resposta Imune , Imunogenicidade da Vacina , Camundongos , Camundongos SCID , Proteínas de Protozoários/genética , Relação Estrutura-Atividade , Vacinação
3.
Nature ; 625(7995): 578-584, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38123677

RESUMO

The symptoms of malaria occur during the blood stage of infection, when parasites invade and replicate within human erythrocytes. The PfPCRCR complex1, containing PfRH5 (refs. 2,3), PfCyRPA, PfRIPR, PfCSS and PfPTRAMP, is essential for erythrocyte invasion by the deadliest human malaria parasite, Plasmodium falciparum. Invasion can be prevented by antibodies3-6 or nanobodies1 against each of these conserved proteins, making them the leading blood-stage malaria vaccine candidates. However, little is known about how PfPCRCR functions during invasion. Here we present the structure of the PfRCR complex7,8, containing PfRH5, PfCyRPA and PfRIPR, determined by cryogenic-electron microscopy. We test the hypothesis that PfRH5 opens to insert into the membrane9, instead showing that a rigid, disulfide-locked PfRH5 can mediate efficient erythrocyte invasion. We show, through modelling and an erythrocyte-binding assay, that PfCyRPA-binding antibodies5 neutralize invasion through a steric mechanism. We determine the structure of PfRIPR, showing that it consists of an ordered, multidomain core flexibly linked to an elongated tail. We also show that the elongated tail of PfRIPR, which is the target of growth-neutralizing antibodies6, binds to the PfCSS-PfPTRAMP complex on the parasite membrane. A modular PfRIPR is therefore linked to the merozoite membrane through an elongated tail, and its structured core presents PfCyRPA and PfRH5 to interact with erythrocyte receptors. This provides fresh insight into the molecular mechanism of erythrocyte invasion and opens the way to new approaches in rational vaccine design.


Assuntos
Eritrócitos , Malária Falciparum , Complexos Multiproteicos , Parasitos , Plasmodium falciparum , Proteínas de Protozoários , Animais , Humanos , Anticorpos Neutralizantes/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Microscopia Crioeletrônica , Dissulfetos/química , Dissulfetos/metabolismo , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Malária Falciparum/patologia , Merozoítos/metabolismo , Complexos Multiproteicos/química , Complexos Multiproteicos/imunologia , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Parasitos/metabolismo , Parasitos/patogenicidade , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/ultraestrutura
4.
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
5.
Immunity ; 47(5): 835-847.e4, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29150238

RESUMO

Immune response (Ir) genes, originally proposed by Baruj Benacerraf to explain differential antigen-specific responses in animal models, have become synonymous with the major histocompatibility complex (MHC). We discovered a non-MHC-linked Ir gene in a T cell receptor (TCR) locus that was required for CD8+ T cell responses to the Plasmodium berghei GAP5040-48 epitope in mice expressing the MHC class I allele H-2Db. GAP5040-48-specific CD8+ T cell responses emerged from a very large pool of naive Vß8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vß8.1+ TCR-H-2Db-GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1ß residues present exclusively in the Vß8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vß8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope.


Assuntos
Antígeno de Histocompatibilidade H-2D/genética , Plasmodium berghei/imunologia , Proteínas de Protozoários/imunologia , Receptores de Antígenos de Linfócitos T/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Regiões Determinantes de Complementaridade , Epitopos , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/imunologia
6.
Immunity ; 47(6): 1197-1209.e10, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29195810

RESUMO

Antibodies against the NANP repeat of circumsporozoite protein (CSP), the major surface antigen of Plasmodium falciparum (Pf) sporozoites, can protect from malaria in animal models but protective humoral immunity is difficult to induce in humans. Here we cloned and characterized rare affinity-matured human NANP-reactive memory B cell antibodies elicited by natural Pf exposure that potently inhibited parasite transmission and development in vivo. We unveiled the molecular details of antibody binding to two distinct protective epitopes within the NANP repeat. NANP repeat recognition was largely mediated by germline encoded and immunoglobulin (Ig) heavy-chain complementarity determining region 3 (HCDR3) residues, whereas affinity maturation contributed predominantly to stabilizing the antigen-binding site conformation. Combined, our findings illustrate the power of exploring human anti-CSP antibody responses to develop tools for malaria control in the mammalian and the mosquito vector and provide a molecular basis for the structure-based design of next-generation CSP malaria vaccines.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Imunidade Humoral , Cadeias Pesadas de Imunoglobulinas/imunologia , Malária Falciparum/prevenção & controle , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/química , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Linfócitos B/imunologia , Linfócitos B/parasitologia , Cristalografia por Raios X , Epitopos/química , Epitopos/imunologia , Feminino , Expressão Gênica , Humanos , Cadeias Pesadas de Imunoglobulinas/biossíntese , Cadeias Pesadas de Imunoglobulinas/química , Memória Imunológica , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Masculino , Camundongos , Modelos Moleculares , Plasmodium berghei/imunologia , Plasmodium falciparum/imunologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esporozoítos/química , Esporozoítos/imunologia
7.
Nature ; 582(7810): 104-108, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32427965

RESUMO

Malaria caused by Plasmodium falciparum remains the leading single-agent cause of mortality in children1, yet the promise of an effective vaccine has not been fulfilled. Here, using our previously described differential screening method to analyse the proteome of blood-stage P. falciparum parasites2, we identify P. falciparum glutamic-acid-rich protein (PfGARP) as a parasite antigen that is recognized by antibodies in the plasma of children who are relatively resistant-but not those who are susceptible-to malaria caused by P. falciparum. PfGARP is a parasite antigen of 80 kDa that is expressed on the exofacial surface of erythrocytes infected by early-to-late-trophozoite-stage parasites. We demonstrate that antibodies against PfGARP kill trophozoite-infected erythrocytes in culture by inducing programmed cell death in the parasites, and that vaccinating non-human primates with PfGARP partially protects against a challenge with P. falciparum. Furthermore, our longitudinal cohort studies showed that, compared to individuals who had naturally occurring anti-PfGARP antibodies, Tanzanian children without anti-PfGARP antibodies had a 2.5-fold-higher risk of severe malaria and Kenyan adolescents and adults without these antibodies had a twofold-higher parasite density. By killing trophozoite-infected erythrocytes, PfGARP could synergize with other vaccines that target parasite invasion of hepatocytes or the invasion of and egress from erythrocytes.


Assuntos
Apoptose/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Parasitos/imunologia , Plasmodium falciparum/citologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adolescente , Adulto , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Aotidae/imunologia , Aotidae/parasitologia , Caspases/metabolismo , Criança , Estudos de Coortes , DNA de Protozoário/química , DNA de Protozoário/metabolismo , Ativação Enzimática , Eritrócitos/parasitologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Quênia , Vacinas Antimaláricas/imunologia , Malária Falciparum/parasitologia , Masculino , Camundongos , Parasitos/citologia , Parasitos/crescimento & desenvolvimento , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/química , Tanzânia , Trofozoítos/citologia , Trofozoítos/crescimento & desenvolvimento , Trofozoítos/imunologia , Vacúolos/imunologia
8.
Nature ; 587(7833): 309-312, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32650338

RESUMO

The Plasmodium species that cause malaria are obligate intracellular parasites, and disease symptoms occur when these parasites replicate in human blood. Despite the risk of immune detection, the parasite delivers proteins that bind to host receptors on the cell surfaces of infected erythrocytes. In the causative parasite of the most deadly form of malaria in humans, Plasmodium falciparum, RIFINs form the largest family of surface proteins displayed by erythrocytes1. Some RIFINs can bind to inhibitory immune receptors, and these RIFINs act as targets for unusual antibodies that contain a LAIR1 ectodomain2-4 or as ligands for LILRB15. RIFINs stimulate the activation of and signalling by LILRB15, which could potentially lead to the dampening of human immune responses. Here, to understand how RIFINs activate LILRB1-mediated signalling, we determine the structure of a RIFIN bound to LILRB1. We show that this RIFIN mimics the natural activating ligand of LILRB1, MHC class I, in its LILRB1-binding mode. A single mutation in the RIFIN disrupts the complex, blocks LILRB1 binding of all tested RIFINs and abolishes signalling in a reporter assay. In a supported lipid bilayer system, which mimics the activation of natural killer (NK) cells by antibody-dependent cell-mediated cytotoxicity, both RIFIN and MHC are recruited to the immunological synapse of NK cells and reduce the activation of NK cells, as measured by the mobilization of perforin. Therefore, LILRB1-binding RIFINs mimic the binding mode of the natural ligand of LILRB1 and suppress the function of NK cells.


Assuntos
Receptor B1 de Leucócitos Semelhante a Imunoglobulina/química , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/imunologia , Malária Falciparum/imunologia , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Sítios de Ligação/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Ligantes , Bicamadas Lipídicas , Ativação Linfocitária , Malária Falciparum/parasitologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mimetismo Molecular/imunologia , Mutação , Perforina/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Transdução de Sinais
9.
Bioessays ; 46(7): e2400053, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38713161

RESUMO

Trypanosoma brucei is the causal agent of African Trypanosomiasis in humans and other animals. It maintains a long-term infection through an antigenic variation based population survival strategy. To proliferate in a mammal, T. brucei acquires iron and haem through the receptor mediated uptake of host transferrin and haptoglobin-hemoglobin respectively. The receptors are exposed to host antibodies but this does not lead to clearance of the infection. Here we discuss how the trypanosome avoids this fate in the context of recent findings on the structure and cell biology of the receptors.


Assuntos
Trypanosoma brucei brucei , Tripanossomíase Africana , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/metabolismo , Humanos , Animais , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/parasitologia , Haptoglobinas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/imunologia , Transferrina/metabolismo , Hemoglobinas/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/imunologia , Interações Hospedeiro-Parasita/imunologia , Ferro/metabolismo , Anticorpos Antiprotozoários/imunologia
10.
Clin Microbiol Rev ; 37(2): e0007123, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38656211

RESUMO

SUMMARYMalaria remains one of the biggest health problems in the world. While significant reductions in malaria morbidity and mortality had been achieved from 2000 to 2015, the favorable trend has stalled, rather significant increases in malaria cases are seen in multiple areas. In 2022, there were 249 million estimated cases, and 608,000 malaria-related deaths, mostly in infants and children aged under 5 years, globally. Therefore, in addition to the expansion of existing anti-malarial control measures, it is critical to develop new tools, such as vaccines and monoclonal antibodies (mAbs), to fight malaria. In the last 2 years, the first and second malaria vaccines, both targeting Plasmodium falciparum circumsporozoite proteins (PfCSP), have been recommended by the World Health Organization to prevent P. falciparum malaria in children living in moderate to high transmission areas. While the approval of the two malaria vaccines is a considerable milestone in vaccine development, they have much room for improvement in efficacy and durability. In addition to the two approved vaccines, recent clinical trials with mAbs against PfCSP, blood-stage vaccines against P. falciparum or P. vivax, and transmission-blocking vaccine or mAb against P. falciparum have shown promising results. This review summarizes the development of the anti-PfCSP vaccines and mAbs, and recent topics in the blood- and transmission-blocking-stage vaccine candidates and mAbs. We further discuss issues of the current vaccines and the directions for the development of next-generation vaccines.


Assuntos
Anticorpos Monoclonais , Vacinas Antimaláricas , Vacinas Antimaláricas/imunologia , Humanos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Plasmodium falciparum/imunologia , Malária/prevenção & controle , Malária/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/imunologia , Anticorpos Antiprotozoários/imunologia , Proteínas de Protozoários/imunologia , Ensaios Clínicos como Assunto
11.
Proc Natl Acad Sci U S A ; 119(35): e2209729119, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35994647

RESUMO

Glutaminyl cyclase (QC) modifies N-terminal glutamine or glutamic acid residues of target proteins into cyclic pyroglutamic acid (pGlu). Here, we report the biochemical and functional analysis of Plasmodium QC. We show that sporozoites of QC-null mutants of rodent and human malaria parasites are recognized by the mosquito immune system and melanized when they reach the hemocoel. Detailed analyses of rodent malaria QC-null mutants showed that sporozoite numbers in salivary glands are reduced in mosquitoes infected with QC-null or QC catalytically dead mutants. This phenotype can be rescued by genetic complementation or by disrupting mosquito melanization or phagocytosis by hemocytes. Mutation of a single QC-target glutamine of the major sporozoite surface protein (circumsporozoite protein; CSP) of the rodent parasite Plasmodium berghei also results in melanization of sporozoites. These findings indicate that QC-mediated posttranslational modification of surface proteins underlies evasion of killing of sporozoites by the mosquito immune system.


Assuntos
Aminoaciltransferases , Culicidae , Malária , Processamento de Proteína Pós-Traducional , Esporozoítos , Aminoaciltransferases/imunologia , Animais , Culicidae/imunologia , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Humanos , Malária/genética , Malária/imunologia , Malária/parasitologia , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia
12.
J Infect Dis ; 229(5): 1565-1573, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38298126

RESUMO

Plasmodium falciparum glutamic acid-rich protein (PfGARP) is a recently characterized cell surface antigen encoded by Plasmodium falciparum, the causative agent of severe human malaria pathophysiology. Previously, we reported that the human erythrocyte band 3 (SLC4A1) serves as a host receptor for PfGARP. Antibodies against PfGARP did not affect parasite invasion and growth. We surmised that PfGARP may play a role in the rosetting and adhesion of malaria. Another study reported that antibodies targeting PfGARP exhibit potent inhibition of parasite growth. This inhibition occurred without the presence of any immune or complement components, suggesting the activation of an inherent density-dependent regulatory system. Here, we used polyclonal antibodies against PfGARP and a monoclonal antibody mAb7899 to demonstrate that anti-PfGARP polyclonal antibodies, but not mAb7899, exerted potent inhibition of parasite growth in infected erythrocytes independent of PfGARP. These findings suggest that an unknown malaria protein(s) is the target of growth arrest by polyclonal antibodies raised against PfGARP.


Assuntos
Anticorpos Antiprotozoários , Eritrócitos , Plasmodium falciparum , Proteínas de Protozoários , Plasmodium falciparum/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Humanos , Eritrócitos/parasitologia , Eritrócitos/imunologia , Proteínas de Protozoários/imunologia , Anticorpos Antiprotozoários/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Animais , Malária Falciparum/imunologia , Malária Falciparum/parasitologia
13.
J Infect Dis ; 229(6): 1883-1893, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38330357

RESUMO

BACKGROUND: Malaria is preventable yet causes >600 000 deaths annually. RTS,S, the first marketed malaria vaccine, has modest efficacy, but improvements are needed for eradication. METHODS: We conducted an open-label, dose escalation phase 1 study of a full-length recombinant circumsporozoite protein vaccine (rCSP) administered with adjuvant glucopyranosyl lipid A-liposome Quillaja saponaria 21 formulation (GLA-LSQ) on days 1, 29, and 85 or 1 and 490 to healthy, malaria-naive adults. The primary end points were safety and reactogenicity. The secondary end points were antibody responses and Plasmodium falciparum parasitemia after homologous controlled human malaria infection. RESULTS: Participants were enrolled into 4 groups receiving rCSP/GLA-LSQ: 10 µg × 3 (n = 20), 30 µg × 3 (n = 10), 60 µg × 3 (n = 10), or 60 µg × 2 (n = 9); 10 participants received 30 µg rCSP alone × 3, and there were 6 infectivity controls. Participants experienced no serious adverse events. Rates of solicited and unsolicited adverse events were similar among groups. All 26 participants who underwent controlled human malaria infection 28 days after final vaccinations developed malaria. Increasing vaccine doses induced higher immunoglobulin G titers but did not achieve previously established RTS,S benchmarks. CONCLUSIONS: rCSP/GLA-LSQ had favorable safety results. However, tested regimens did not induce protective immunity. Further investigation could assess whether adjuvant or schedule adjustments improve efficacy. CLINICAL TRIALS REGISTRATION: NCT03589794.


Assuntos
Adjuvantes Imunológicos , Anticorpos Antiprotozoários , Lipídeo A , Lipossomos , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Proteínas de Protozoários , Humanos , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/efeitos adversos , Malária Falciparum/prevenção & controle , Malária Falciparum/imunologia , Adulto , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Feminino , Masculino , Adjuvantes Imunológicos/administração & dosagem , Adulto Jovem , Lipídeo A/análogos & derivados , Lipídeo A/administração & dosagem , Lipídeo A/imunologia , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Quillaja/química , Adolescente , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Pessoa de Meia-Idade , Glucosídeos
14.
Infect Immun ; 92(7): e0001524, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38842304

RESUMO

Strain-transcending antibodies against virulence-associated subsets of P. falciparum-infected erythrocyte surface antigens could protect children from severe malaria. However, the evidence supporting the existence of such antibodies is incomplete and inconsistent. One subset of surface antigens associated with severe malaria, rosette-mediating Plasmodium falciparum Erythrocyte Membrane Protein one (PfEMP1) variants, cause infected erythrocytes to bind to uninfected erythrocytes to form clusters of cells (rosettes) that contribute to microvascular obstruction and pathology. Here, we tested plasma from 80 individuals living in malaria-endemic regions for IgG recognition of the surface of four P. falciparum rosetting strains using flow cytometry. Broadly reactive plasma samples were then used in antibody elution experiments in which intact IgG was eluted from the surface of infected erythrocytes and transferred to heterologous rosetting strains to look for strain-transcending antibodies. We found that seroprevalence (percentage of positive plasma samples) against allopatric rosetting strains was high in adults (63%-93%) but lower in children (13%-48%). Strain-transcending antibodies were present in nine out of eleven eluted antibody experiments, with six of these recognizing multiple heterologous rosetting parasite strains. One eluate had rosette-disrupting activity against heterologous strains, suggesting PfEMP1 as the likely target of the strain-transcending antibodies. Naturally acquired strain-transcending antibodies to rosetting P. falciparum strains in humans have not been directly demonstrated previously. Their existence suggests that such antibodies could play a role in clinical protection and raises the possibility that conserved epitopes recognized by strain-transcending antibodies could be targeted therapeutically by monoclonal antibodies or vaccines.


Assuntos
Anticorpos Antiprotozoários , Imunoglobulina G , Malária Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/imunologia , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/sangue , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Criança , Adulto , Imunoglobulina G/imunologia , Imunoglobulina G/sangue , Pré-Escolar , Adolescente , Proteínas de Protozoários/imunologia , Eritrócitos/parasitologia , Eritrócitos/imunologia , Antígenos de Protozoários/imunologia , Feminino , Masculino , Adulto Jovem , Pessoa de Meia-Idade , Estudos Soroepidemiológicos , Formação de Roseta , Citometria de Fluxo
15.
BMC Med ; 22(1): 170, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38649867

RESUMO

BACKGROUND: The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS: In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS: Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS: R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION: The trial is registered with ClinicalTrials.gov under identifier NCT04862416.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Glicoproteínas de Membrana , Plasmodium falciparum , Proteínas de Protozoários , Adolescente , Adulto , Animais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Adjuvantes Imunológicos/administração & dosagem , Hidróxido de Alumínio/administração & dosagem , Anticorpos Antiprotozoários , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Malária Falciparum/imunologia , Países Baixos , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia
16.
Immunity ; 42(3): 580-90, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25786180

RESUMO

Antibodies play major roles in immunity to malaria; however, a limited understanding of mechanisms mediating protection is a major barrier to vaccine development. We have demonstrated that acquired human anti-malarial antibodies promote complement deposition on the merozoite to mediate inhibition of erythrocyte invasion through C1q fixation and activation of the classical complement pathway. Antibody-mediated complement-dependent (Ab-C') inhibition was the predominant invasion-inhibitory activity of human antibodies; most antibodies were non-inhibitory without complement. Inhibitory activity was mediated predominately via C1q fixation, and merozoite surface proteins 1 and 2 were identified as major targets. Complement fixation by antibodies was very strongly associated with protection from both clinical malaria and high-density parasitemia in a prospective longitudinal study of children. Ab-C' inhibitory activity could be induced by human immunization with a candidate merozoite surface-protein vaccine. Our findings demonstrate that human anti-malarial antibodies have evolved to function by fixing complement for potent invasion-inhibitory activity and protective immunity.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Complemento C1q/metabolismo , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Merozoítos/imunologia , Parasitemia/prevenção & controle , Plasmodium falciparum/imunologia , Adolescente , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Criança , Pré-Escolar , Testes de Fixação de Complemento , Via Clássica do Complemento , Eritrócitos/imunologia , Eritrócitos/parasitologia , Feminino , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunoglobulina G/biossíntese , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Masculino , Proteína 1 de Superfície de Merozoito/antagonistas & inibidores , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Parasitemia/imunologia , Parasitemia/parasitologia , Estudos Prospectivos , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia
17.
Malar J ; 23(1): 163, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783317

RESUMO

BACKGROUND: Plasmodium vivax represents the most geographically widespread human malaria parasite affecting civilian and military populations in endemic areas. Targeting the pre-erythrocytic (PE) stage of the parasite life cycle is especially appealing for developing P. vivax vaccines as it would prevent disease and transmission. Here, naturally acquired immunity to a panel of P. vivax PE antigens was explored, which may facilitate vaccine development and lead to a better understanding of naturally acquired PE immunity. METHODS: Twelve P. vivax PE antigens orthologous to a panel of P. falciparum antigens previously identified as highly immunogenic in protected subjects after immunization with radiation attenuated sporozoites (RAS) were used for evaluation of humoral and cellular immunity by ELISA and IFN-γ ELISpot. Samples from P. vivax infected individuals (n = 76) from a low endemic malaria region in the Peruvian Amazon Basin were used. RESULTS: In those clinical samples, all PE antigens evaluated showed positive IgG antibody reactivity with a variable prevalence of 58-99% in recently P. vivax diagnosed patients. The magnitude of the IgG antibody response against PE antigens was lower compared with blood stage antigens MSP1 and DBP-II, although antibody levels persisted better for PE antigens (average decrease of 6% for PE antigens and 43% for MSP1, p < 0.05). Higher IgG antibodies was associated with one or more previous malaria episodes only for blood stage antigens (p < 0.001). High IgG responders across PE and blood stage antigens showed significantly lower parasitaemia compared to low IgG responders (median 1,921 vs 4,663 par/µl, p < 0.05). In a subgroup of volunteers (n = 17),positive IFN-γ T cell response by ELISPOT was observed in 35% vs 9-35% against blood stage MSP1 and PE antigens, respectively, but no correlation with IgG responses. CONCLUSIONS: These results demonstrate clear humoral and T cell responses against P. vivax PE antigens in individuals naturally infected with P. vivax. These data identify novel attractive PE antigens suitable for use in the potential development and selection of new malaria vaccine candidates which can be used as a part of malaria prevention strategies in civilian and military populations living in P. vivax endemic areas.


Assuntos
Antígenos de Protozoários , Malária Vivax , Plasmodium vivax , Proteínas de Protozoários , Plasmodium vivax/imunologia , Peru/epidemiologia , Humanos , Malária Vivax/imunologia , Malária Vivax/epidemiologia , Adulto , Masculino , Adulto Jovem , Adolescente , Feminino , Pessoa de Meia-Idade , Proteínas de Protozoários/imunologia , Antígenos de Protozoários/imunologia , Imunoglobulina G/sangue , Anticorpos Antiprotozoários/sangue , Ensaio de Imunoadsorção Enzimática , Criança , Idoso , ELISPOT
18.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38673969

RESUMO

This study presents an evaluation of seventeen newly produced recombinant trivalent chimeric proteins (containing the same immunodominant fragment of SAG1 and SAG2 of Toxoplasma gondii antigens, and an additional immunodominant fragment of one of the parasite antigens, such as AMA1, GRA1, GRA2, GRA5, GRA6, GRA7, GRA9, LDH2, MAG1, MIC1, MIC3, P35, and ROP1) as a potential alternative to the whole-cell tachyzoite lysate (TLA) used in the detection of infection in small ruminants. These recombinant proteins, obtained by genetic engineering and molecular biology methods, were tested for their reactivity with specific anti-Toxoplasma IgG antibodies contained in serum samples of small ruminants (192 samples of sheep serum and 95 samples of goat serum) using an enzyme-linked immunosorbent assay (ELISA). The reactivity of six recombinant trivalent chimeric proteins (SAG1-SAG2-GRA5, SAG1-SAG2-GRA9, SAG1-SAG2-MIC1, SAG1-SAG2-MIC3, SAG1-SAG2-P35, and SAG1-SAG2-ROP1) with IgG antibodies generated during T. gondii invasion was comparable to the sensitivity of TLA-based IgG ELISA (100%). The obtained results show a strong correlation with the results obtained for TLA. This suggests that these protein preparations may be a potential alternative to TLA used in commercial tests and could be used to develop a cheaper test for the detection of parasite infection in small ruminants.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Ensaio de Imunoadsorção Enzimática , Cabras , Imunoglobulina G , Toxoplasma , Animais , Toxoplasma/imunologia , Toxoplasma/genética , Imunoglobulina G/imunologia , Imunoglobulina G/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Ovinos , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/sangue , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/genética , Toxoplasmose Animal/diagnóstico , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/parasitologia , Doenças dos Ovinos/parasitologia , Doenças dos Ovinos/diagnóstico , Doenças dos Ovinos/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/genética , Doenças das Cabras/parasitologia , Doenças das Cabras/diagnóstico , Doenças das Cabras/imunologia
19.
J Vector Borne Dis ; 61(2): 203-210, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38922654

RESUMO

BACKGROUND OBJECTIVES: Plasmodium knowlesi, a simian malaria species, is now known to infect humans. Due to disadvantages in the current diagnosis methods, many efforts have been placed into developing new methods to diagnose the disease. This study assessed the ability of the PkRAP-1 sandwich enzyme-linked immunosorbent (ELISA) to detect P knowlesi antigens in whole blood specimens. METHODS: Western blot assay was conducted to evaluate the ability of raised mouse and rabbit anti-PkRAP-1 polyclonal antibodies to bind to the native proteins in P. knowlesi lysate. The polyclonal antibodies were then used in sandwich ELISA to detect P. knowlesi. In the sandwich ELISA, mouse and rabbit polyclonal antibodies were used as the capture and detection antibodies, respectively. The limit of detection (LOD) of the assay was determined using P. knowlesi A1H1 culture and purified recombinant PkRAP-1. RESULTS: Western blot results showed positive reactions towards the proteins in P. knowlesi lysate. The LOD of the assay from three technical replicates was 0.068% parasitaemia. The assay performance in detecting P. knowlesi was 83% sensitivity and 70% specificity with positive and negative predictive values of 74% and 80%, respectively. The anti-PkRAP-1 polyclonal antibodies did not cross-react with P. falciparum and healthy samples, but P. vivax by detecting all 12 samples. INTERPRETATION CONCLUSION: PkRAP-1 has the potential as a biomarker for the development of a new diagnostic tool for P. knowlesi detection. Further studies need to be conducted to establish the full potential of the usage of anti-PkRAP-1 antibodies for P. knowlesi detection.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Ensaio de Imunoadsorção Enzimática , Malária , Plasmodium knowlesi , Proteínas de Protozoários , Sensibilidade e Especificidade , Plasmodium knowlesi/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Animais , Malária/diagnóstico , Malária/sangue , Anticorpos Antiprotozoários/sangue , Coelhos , Camundongos , Proteínas de Protozoários/imunologia , Humanos , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/sangue , Western Blotting/métodos , Limite de Detecção
20.
Immunol Rev ; 293(1): 230-252, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31562653

RESUMO

Malaria, a mosquito-borne infectious disease caused by parasites of the genus Plasmodium continues to be a major health problem worldwide. The unicellular Plasmodium-parasites have the unique capacity to infect and replicate within host erythrocytes. By expressing variant surface antigens Plasmodium falciparum has evolved to avoid protective immune responses; as a result in endemic areas anti-malaria immunity develops gradually over many years of multiple and repeated infections. We are studying the role of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) expressed by asexual stages of P. falciparum responsible for the pathogenicity of severe malaria. The immunopathology of falciparum malaria has been linked to cyto-adhesion of infected erythrocytes to specific host receptors. A greater appreciation of the PfEMP1 molecules important for the development of protective immunity and immunopathology is a prerequisite for the rational discovery and development of a safe and protective anti-disease malaria vaccine. Here we review the role of ICAM-1 and EPCR receptor adhering falciparum-parasites in the development of severe malaria; we discuss our current research to understand the factors involved in the pathogenesis of cerebral malaria and the feasibility of developing a vaccine targeted specifically to prevent this disease.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Variação Antigênica , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Humanos , Imunidade , Vacinas Antimaláricas/imunologia , Malária Cerebral/prevenção & controle , Malária Falciparum/prevenção & controle , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA