Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.428
Filtrar
1.
Diagn Microbiol Infect Dis ; 110(3): 116480, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39163788

RESUMO

The apical membrane antigen-1 (AMA-1) is a crucial target for malaria management and prevention strategies. While the immunogenicity of AMA-1 has been extensively studied for Plasmodium falciparum and Plasmodium vivax, there is a notable scarcity of information for Plasmodium malariae. In this study, recombinant PmAMA-1 was expressed in Escherichia coli, and its integrity was confirmed via western blotting and indirect immunofluorescence assays. Immunization of BALB/c mice with rPmAMA-1 emulsified in Freund's adjuvant resulted in significantly elevated specific IgG antibodies, predominantly IgG1. The immune response exhibited Th1, Th2, and Th17 phenotypes, with a notable Th1 bias. Antisera from immunized mice effectively recognized native PmAMA-1 on P. malariae. These results suggest that PmAMA-1 is a promising target for both vaccine development and diagnostic applications for P. malariae infections, offering dual preventive and diagnostic benefits in malaria control.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Malária , Proteínas de Membrana , Plasmodium malariae , Proteínas de Protozoários , Animais , Feminino , Camundongos , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Escherichia coli/genética , Imunoglobulina G/sangue , Malária/diagnóstico , Malária/prevenção & controle , Malária/imunologia , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Proteínas de Membrana/imunologia , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Plasmodium malariae/imunologia , Plasmodium malariae/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/genética
2.
Immunobiology ; 229(5): 152837, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39089130

RESUMO

It is known that conventional antigen presentation involves phagocytosis of antigens followed by its internalization in endocytic compartments and presentation of epitopes through MHC class II molecules for CD4 T cells. However, since 1976 a cross-presentation pathway has been studied, in which CD8 T cells are activated via MHC class I with antigens acquired through phagocytosis or endocytosis by dendritic cells (DCs). Among some important molecules involved in the cross-presentation, the C-type lectin receptor of the Dectin-1 cluster (CLECs), particularly the CLEC9A receptor, not only is expressed in dendritic cells but also presents a pivotal role in this context. In special, CLEC12A has been highlighted as a malaria pigment hemozoin (HZ) receptor. During Plasmodium infection, hemozoin crystals defend the parasite against heme toxicity within erythrocytes, as well as the released native HZ elicits pro-inflammatory responses and can induce cross-presentation. Particularly, this crystal can be synthesized from hematin anhydride and mimics the native form, and the gaps generated between the nanocrystal domains during its synthesis allow for substance coupling followed by its coating. Therefore, this study aimed to assess whether synthetic hemozoin (sHz) or hematin anhydride could be a nanocarrier and promote cross-presentation in dendritic cells. Firstly, it was verified that sHz can carry coated and coupled antigens, the compounds can associate to LAMP1-positive vesicles and decrease overall intracellular pH, which can potentially enhance the cross-presentation of ovalbumin and Leishmania infantum antigens. Thus, this study adds important data in the molecular intricacies of antigen presentation by showing not only the sHz immunomodulatory properties but also its potential applications as an antigen carrier.


Assuntos
Apresentação de Antígeno , Apresentação Cruzada , Células Dendríticas , Hemeproteínas , Hemeproteínas/imunologia , Apresentação Cruzada/imunologia , Animais , Células Dendríticas/imunologia , Camundongos , Nanopartículas/química , Humanos , Malária/imunologia , Lectinas Tipo C/metabolismo , Lectinas Tipo C/imunologia , Ovalbumina/imunologia
3.
Front Cell Infect Microbiol ; 14: 1438019, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39149419

RESUMO

The malaria-causing parasites have to complete a complex infection cycle in the mosquito vector that also involves attack by the insect's innate immune system, especially at the early stages of midgut infection. However, Anopheles immunity to the late Plasmodium sporogonic stages, such as oocysts, has received little attention as they are considered to be concealed from immune factors due to their location under the midgut basal lamina and for harboring an elaborate cell wall comprising an external layer derived from the basal lamina that confers self-properties to an otherwise foreign structure. Here, we investigated whether Plasmodium berghei oocysts and sporozoites are susceptible to melanization-based immunity in Anopheles gambiae. Silencing of the negative regulator of melanization response, CLIPA14, increased melanization prevalence without significantly increasing the numbers of melanized oocysts, while co-silencing CLIPA14 with CLIPA2, a second negative regulator of melanization, resulted in a significant increase in melanized oocysts and melanization prevalence. Only late-stage oocysts were found to be melanized, suggesting that oocyst rupture was a prerequisite for melanization-based immune attack, presumably due to the loss of the immune-evasive features of their wall. We also found melanized sporozoites inside oocysts and in the hemocoel, suggesting that sporozoites at different maturation stages are susceptible to melanization. Silencing the melanization promoting factors TEP1 and CLIPA28 rescued oocyst melanization in CLIPA2/CLIPA14 co-silenced mosquitoes. Interestingly, silencing of CTL4, that protects early stage ookinetes from melanization, had no effect on oocysts and sporozoites, indicating differential regulation of immunity to early and late sporogonic stages. Similar to previous studies addressing ookinete stage melanization, the melanization of Plasmodium falciparum oocysts was significantly lower than that observed for P. berghei. In summary, our results provide conclusive evidence that late sporogonic malaria parasite stages are susceptible to melanization, and we reveal distinct regulatory mechanisms for ookinete and oocyst melanization.


Assuntos
Anopheles , Melaninas , Oocistos , Plasmodium berghei , Esporozoítos , Animais , Anopheles/parasitologia , Anopheles/imunologia , Plasmodium berghei/imunologia , Oocistos/metabolismo , Melaninas/metabolismo , Esporozoítos/imunologia , Esporozoítos/metabolismo , Mosquitos Vetores/parasitologia , Mosquitos Vetores/imunologia , Proteínas de Insetos/metabolismo , Proteínas de Insetos/genética , Proteínas de Insetos/imunologia , Malária/imunologia , Malária/parasitologia , Inativação Gênica , Imunidade Inata , Feminino
4.
Infect Genet Evol ; 123: 105650, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39089500

RESUMO

Despite the critical role of the Anopheles innate immune system in defending against Plasmodium infection, there is still limited information about the key immune mechanisms in Anopheles. This review assesses recent findings on the expression characteristics of immune-related genes in Anopheles following exposure to Plasmodium. A literature review, unrestricted by publication date, was conducted to evaluate immune-related gene expression in different organs of Anopheles after Plasmodium infection. Mosquito immune responses in the midgut are essential for reducing parasite populations. Additionally, innate immune responses in the salivary glands and hemocytes circulating in the hemocoel play key roles in defense against the parasite. Transcriptomic analysis of the mosquito's innate immune response to Plasmodium infection provides valuable insights into key immune mechanisms in mosquito defense. A deeper understanding of immune mechanisms in different organs of Anopheles following Plasmodium infection will aid in discovering critical targets for designing novel control strategies.


Assuntos
Anopheles , Imunidade Inata , Malária , Plasmodium , Animais , Anopheles/parasitologia , Anopheles/genética , Anopheles/imunologia , Malária/imunologia , Malária/parasitologia , Plasmodium/imunologia , Plasmodium/genética , Perfilação da Expressão Gênica , Mosquitos Vetores/parasitologia , Mosquitos Vetores/genética , Mosquitos Vetores/imunologia , Interações Hospedeiro-Parasita/imunologia , Transcriptoma
5.
Front Immunol ; 15: 1426316, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39211036

RESUMO

γδ T cells facilitate the CD4+ T helper 1 (Th1) cell response against Plasmodium infection by activating conventional dendritic cells (cDCs), although the underlying mechanism remains elusive. Our study revealed that γδ T cells promote the complete maturation and production of interleukin-12 and CXCR3-ligands specifically in type 1 cDCs (cDC1), with minimal impact on cDC2 and monocyte derived DCs (Mo-DCs). During the initial infection phase, γδ T cell activation and temporal accumulation in the splenic white pulp, alongside cDC1, occur via CCR7-signaling. Furthermore, cDC1/γδ T cell interactions in the white pulp are amplified through CXCR3 signaling in γδ T cells, optimizing Th1 cell priming by cDC1. We also demonstrated how transitional Th1 cells arise in the white pulp before establishing their presence in the red pulp as fully differentiated Th1 cells. Additionally, we elucidate the reciprocal activation between γδ T cells and cDC1s. These findings suggest that Th1 cell priming is orchestrated by this reciprocal activation in the splenic white pulp during the early phase of blood-stage Plasmodium infection.


Assuntos
Células Dendríticas , Ativação Linfocitária , Malária , Células Th1 , Células Th1/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Camundongos , Ativação Linfocitária/imunologia , Malária/imunologia , Malária/parasitologia , Camundongos Endogâmicos C57BL , Receptores CXCR3/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores CCR7/metabolismo , Receptores CCR7/imunologia , Transdução de Sinais , Baço/imunologia , Diferenciação Celular/imunologia , Feminino
6.
Nat Commun ; 15(1): 7206, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174515

RESUMO

Apical membrane antigen-1 (AMA1) is a conserved malarial vaccine candidate essential for the formation of tight junctions with the rhoptry neck protein (RON) complex, enabling Plasmodium parasites to invade human erythrocytes, hepatocytes, and mosquito salivary glands. Despite its critical role, extensive surface polymorphisms in AMA1 have led to strain-specific protection, limiting the success of AMA1-based interventions beyond initial clinical trials. Here, we identify an i-body, a humanised single-domain antibody-like molecule that recognises a conserved pan-species conformational epitope in AMA1 with low nanomolar affinity and inhibits the binding of the RON2 ligand to AMA1. Structural characterisation indicates that the WD34 i-body epitope spans the centre of the conserved hydrophobic cleft in AMA1, where interacting residues are highly conserved among all Plasmodium species. Furthermore, we show that WD34 inhibits merozoite invasion of erythrocytes by multiple Plasmodium species and hepatocyte invasion by P. falciparum sporozoites. Despite a short half-life in mouse serum, we demonstrate that WD34 transiently suppressed P. berghei infections in female BALB/c mice. Our work describes the first pan-species AMA1 biologic with inhibitory activity against multiple life-cycle stages of Plasmodium. With improved pharmacokinetic characteristics, WD34 could be a potential immunotherapy against multiple species of Plasmodium.


Assuntos
Antígenos de Protozoários , Eritrócitos , Fígado , Proteínas de Membrana , Camundongos Endogâmicos BALB C , Proteínas de Protozoários , Animais , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/metabolismo , Feminino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Humanos , Eritrócitos/parasitologia , Eritrócitos/imunologia , Fígado/parasitologia , Fígado/imunologia , Fígado/metabolismo , Vacinas Antimaláricas/imunologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Reações Cruzadas/imunologia , Plasmodium falciparum/imunologia , Plasmodium berghei/imunologia , Epitopos/imunologia , Hepatócitos/parasitologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Plasmodium/imunologia , Merozoítos/imunologia , Merozoítos/metabolismo
7.
EMBO Mol Med ; 16(9): 2060-2079, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39103697

RESUMO

Malaria vaccination approaches using live Plasmodium parasites are currently explored, with either attenuated mosquito-derived sporozoites or attenuated blood-stage parasites. Both approaches would profit from the availability of attenuated and avirulent parasites with a reduced blood-stage multiplication rate. Here we screened gene-deletion mutants of the rodent parasite P. berghei and the human parasite P. falciparum for slow growth. Furthermore, we tested the P. berghei mutants for avirulence and resolving blood-stage infections, while preserving sporozoite formation and liver infection. Targeting 51 genes yielded 18 P. berghei gene-deletion mutants with several mutants causing mild infections. Infections with the two most attenuated mutants either by blood stages or by sporozoites were cleared by the immune response. Immunization of mice led to protection from disease after challenge with wild-type sporozoites. Two of six generated P. falciparum gene-deletion mutants showed a slow growth rate. Slow-growing, avirulent P. falciparum mutants will constitute valuable tools to inform on the induction of immune responses and will aid in developing new as well as safeguarding existing attenuated parasite vaccines.


Assuntos
Vacinas Antimaláricas , Malária , Plasmodium berghei , Plasmodium falciparum , Esporozoítos , Vacinas Atenuadas , Animais , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Esporozoítos/imunologia , Plasmodium berghei/imunologia , Plasmodium berghei/genética , Plasmodium falciparum/imunologia , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Malária/prevenção & controle , Malária/parasitologia , Malária/imunologia , Camundongos , Vacinação/métodos , Humanos , Deleção de Genes , Feminino
8.
Int Immunopharmacol ; 140: 112843, 2024 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-39098224

RESUMO

Neutrophils and macrophages confine pathogens by entrapping them in extracellular traps (ETs) through activating TLR9 function. However, plasmodial parasites secreted TatD-like DNases (TatD) to counteract ETs-mediated immune clearance. We found that TLR9 mutant mice increased susceptibility to rodent malaria, suggesting TLR9 is a key protein for host defense. We found that the proportion of neutrophils and macrophages in response to plasmodial parasite infection in the TLR9 mutant mice was significantly reduced compared to that of the WT mice. Importantly, PbTatD can directly bind to the surface TLR9 (sTLR9) on macrophages, which blocking the phosphorylation of mitogen-activated protein kinase and nuclear factor-κB, negatively regulated the signaling of ETs formation by both macrophages and neutrophils. Such, P. berghei TatD is a parasite virulence factor that can inhibit the proliferation of macrophages and neutrophils through directly binding to TLR9 receptors on the cell surface, thereby blocking the activation of the downstream MyD88-NF-kB pathways.


Assuntos
Desoxirribonucleases , Imunidade Inata , Macrófagos , Malária , Neutrófilos , Plasmodium berghei , Transdução de Sinais , Animais , Humanos , Camundongos , Desoxirribonucleases/metabolismo , Armadilhas Extracelulares/imunologia , Armadilhas Extracelulares/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Malária/imunologia , Malária/parasitologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Neutrófilos/imunologia , NF-kappa B/metabolismo , Plasmodium berghei/imunologia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Receptor Toll-Like 9/metabolismo
9.
PLoS Negl Trop Dis ; 18(8): e0012424, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39150978

RESUMO

The risk of severe malaria from the zoonotic parasite Plasmodium knowlesi approximates that from P. falciparum. In severe falciparum malaria, neutrophil activation contributes to inflammatory pathogenesis, including acute lung injury (ALI). The role of neutrophil activation in the pathogenesis of severe knowlesi malaria has not been examined. We evaluated 213 patients with P. knowlesi mono-infection (138 non-severe, 75 severe) and 49 Plasmodium-negative controls from Malaysia. Markers of neutrophil activation (soluble neutrophil elastase [NE], citrullinated histone [CitH3] and circulating neutrophil extracellular traps [NETs]) were quantified in peripheral blood by microscopy and immunoassays. Findings were correlated with malaria severity, ALI clinical criteria, biomarkers of parasite biomass, haemolysis, and endothelial activation. Neutrophil activation increased with disease severity, with median levels higher in severe than non-severe malaria and controls for NE (380[IQR:210-930]ng/mL, 236[139-448]ng/mL, 218[134-307]ng/mL, respectively) and CitH3 (8.72[IQR:3.0-23.1]ng/mL, 4.29[1.46-9.49]ng/mL, 1.53[0.6-2.59]ng/mL, respectively)[all p<0.01]. NETs were higher in severe malaria compared to controls (126/µL[IQR:49-323] vs 51[20-75]/µL, p<0.001). In non-severe malaria, neutrophil activation fell significantly upon discharge from hospital (p<0.03). In severe disease, NETs, NE, and CitH3 were correlated with parasitaemia, cell-free haemoglobin and angiopoietin-2 (all Pearson's r>0.24, p<0.05). Plasma NE and angiopoietin-2 were higher in knowlesi patients with ALI than those without (p<0.008); neutrophilia was associated with an increased risk of ALI (aOR 3.27, p<0.01). In conclusion, neutrophil activation is increased in ALI and in proportion to disease severity in knowlesi malaria, is associated with endothelial activation, and may contribute to disease pathogenesis. Trials of adjunctive therapies to regulate neutrophil activation are warranted in severe knowlesi malaria.


Assuntos
Lesão Pulmonar Aguda , Armadilhas Extracelulares , Malária , Ativação de Neutrófilo , Neutrófilos , Plasmodium knowlesi , Índice de Gravidade de Doença , Humanos , Masculino , Feminino , Malária/imunologia , Malária/sangue , Malária/parasitologia , Adulto , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/parasitologia , Lesão Pulmonar Aguda/patologia , Pessoa de Meia-Idade , Neutrófilos/imunologia , Armadilhas Extracelulares/imunologia , Malásia , Biomarcadores/sangue , Adulto Jovem , Elastase de Leucócito/sangue , Histonas/sangue , Adolescente
10.
Nat Commun ; 15(1): 7487, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39209843

RESUMO

CelTOS is a malaria vaccine antigen that is conserved in Plasmodium and other apicomplexan parasites and plays a role in cell-traversal. The structural basis and mechanisms of CelTOS-induced protective immunity to parasites are unknown. Here, CelTOS-specific monoclonal antibodies (mAbs) 7g7 and 4h12 demonstrated multistage activity, protecting against liver infection and preventing parasite transmission to mosquitoes. Both mAbs demonstrated cross-species activity with sterile protection against in vivo challenge with transgenic parasites containing either P. falciparum or P. vivax CelTOS, and with transmission reducing activity against P. falciparum. The mAbs prevented CelTOS-mediated pore formation providing insight into the protective mechanisms. X-ray crystallography and mutant-library epitope mapping revealed two distinct broadly conserved neutralizing epitopes. 7g7 bound to a parallel dimer of CelTOS, while 4h12 bound to a novel antiparallel dimer architecture. These findings inform the design of antibody therapies and vaccines and raise the prospect of a single intervention to simultaneously combat P. falciparum and P. vivax malaria.


Assuntos
Anticorpos Monoclonais , Anticorpos Antiprotozoários , Vacinas Antimaláricas , Plasmodium falciparum , Plasmodium vivax , Anticorpos Monoclonais/imunologia , Animais , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Vacinas Antimaláricas/imunologia , Anticorpos Antiprotozoários/imunologia , Camundongos , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/parasitologia , Cristalografia por Raios X , Epitopos/imunologia , Malária Vivax/prevenção & controle , Malária Vivax/imunologia , Malária Vivax/parasitologia , Antígenos de Protozoários/imunologia , Humanos , Feminino , Mapeamento de Epitopos , Malária/imunologia , Malária/prevenção & controle , Malária/parasitologia , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/química
11.
Viruses ; 16(7)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39066199

RESUMO

Human immunodeficiency virus (HIV) and malaria, caused by infection with Plasmodium spp., are endemic in similar geographical locations. As a result, there is high potential for HIV/Plasmodium co-infection, which increases the pathology of both diseases. However, the immunological mechanisms underlying the exacerbated disease pathology observed in co-infected individuals are poorly understood. Moreover, there is limited data available on the impact of Plasmodium co-infection on antiretroviral (ART)-treated HIV infection. Here, we used the rhesus macaque (RM) model to conduct a pilot study to establish a model of Plasmodium fragile co-infection during ART-treated simian immunodeficiency virus (SIV) infection, and to begin to characterize the immunopathogenic effect of co-infection in the context of ART. We observed that P. fragile co-infection resulted in parasitemia and anemia, as well as persistently detectable viral loads (VLs) and decreased absolute CD4+ T-cell counts despite daily ART treatment. Notably, P. fragile co-infection was associated with increased levels of inflammatory cytokines, including monocyte chemoattractant protein 1 (MCP-1). P. fragile co-infection was also associated with increased levels of neutrophil elastase, a plasma marker of neutrophil extracellular trap (NET) formation, but significant decreases in markers of neutrophil degranulation, potentially indicating a shift in the neutrophil functionality during co-infection. Finally, we characterized the levels of plasma markers of gastrointestinal (GI) barrier permeability and microbial translocation and observed significant correlations between indicators of GI dysfunction, clinical markers of SIV and Plasmodium infection, and neutrophil frequency and function. Taken together, these pilot data verify the utility of using the RM model to examine ART-treated SIV/P. fragile co-infection, and indicate that neutrophil-driven inflammation and GI dysfunction may underlie heightened SIV/P. fragile co-infection pathogenesis.


Assuntos
Coinfecção , Inflamação , Macaca mulatta , Malária , Neutrófilos , Plasmodium , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Coinfecção/tratamento farmacológico , Coinfecção/parasitologia , Coinfecção/virologia , Malária/tratamento farmacológico , Malária/imunologia , Malária/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Projetos Piloto , Neutrófilos/imunologia , Antirretrovirais/uso terapêutico , Carga Viral , Biomarcadores/sangue , Citocinas/sangue , Modelos Animais de Doenças , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia
12.
BMC Immunol ; 25(1): 47, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39039450

RESUMO

BACKGROUND: There is currently insufficient data regarding immune parameters and relationship with severity of malaria infection in Enugu, Nigeria where the economic and social costs of the disease and its management are extremely high. This study was conducted to determine the relationship between malaria severity and some immune-inflammatory markers among malaria-infected children in Enugu, Nigeria. METHODS: The study adopted a case control design. Eligible children were categorized into three groups - complicated, uncomplicated and healthy children. Pro-inflammatory cytokines -interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α); and anti-inflammatory cytokine - interleukin-10 (IL-10) were assayed using enzyme-linked immunosorbent assay (ELISA) technique, while immune cell ratios - neutrophil lymphocyte ratio (NLR) and monocyte lymphocyte ratio (MLR) were calculated from full blood count results. RESULTS: The overall mean age of the participants was 7.3 ± 3.4 (range: 6 months - 12 years) and the male-female ratio was 1:1. There was no significant difference between the ages of the three groups (P = 0.44). The Mean levels of IFN-γ, TNF-α, and NLR were higher in complicated than uncomplicated malaria (266.9 ± 66.3pg/ml vs. 62.5 ± 6.4pg/ml, p < 0.001; 140.3 ± 30.0pg/ml vs. 42.0 ± 9.0pg/ml, p < 0.001; and 32.9 ± 16.2pg/ml vs. 17.8 ± 6.0pg/ml, p < 0.001, respectively); and higher in uncomplicated malaria than healthy children (62.5 ± 6.4pg/ml vs. 40.6 ± 9.1pg/ml, p < 0.001; 42.0 ± 9.0pg/ml vs. 105.7 ± 32.1, p < 0.001; 17.8 ± 6.0pg/ml vs. 18.7 ± 6.2pg/ml, p < 0.001, respectively). On the other hand, the mean level of IL-10 is higher in uncomplicated than complicated malaria (105.73 ± 32.06pg/ml vs. 40.60 ± 9.11pg/ml, p < 0.001). There was a positive correlation between NLR and IFN-γ (r = 0.815; p = 0.003), as well as NLR and TNF-α (r = 0.745; p = 0.002). CONCLUSION: Complicated malaria is associated with higher levels of pro-inflammatory cytokines while uncomplicated malaria is associated with higher levels of anti-inflammatory cytokines. NLR correlates positively with pro-inflammatory cytokines, and could be useful in evaluation for the severity of malaria infection.


Assuntos
Biomarcadores , Malária , Humanos , Masculino , Nigéria/epidemiologia , Feminino , Pré-Escolar , Criança , Biomarcadores/sangue , Lactente , Malária/imunologia , Malária/sangue , Estudos de Casos e Controles , Interferon gama/sangue , Interferon gama/metabolismo , Fator de Necrose Tumoral alfa/sangue , Citocinas/sangue , Neutrófilos/imunologia , Inflamação/imunologia , Inflamação/sangue , Interleucina-10/sangue , Linfócitos/imunologia , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo
13.
Expert Rev Vaccines ; 23(1): 715-729, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39042099

RESUMO

INTRODUCTION: The use of novel adjuvants in human vaccines continues to expand as their contribution to preventing disease in challenging populations and caused by complex pathogens is increasingly understood. AS01 is a family of liposome-based vaccine Adjuvant Systems containing two immunostimulants: 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01-containing vaccines have been approved and administered to millions of individuals worldwide. AREAS COVERED: Here, we report advances in our understanding of the mode of action of AS01 that contributed to the development of efficacious vaccines preventing disease due to malaria, herpes zoster, and respiratory syncytial virus. AS01 induces early innate immune activation that induces T cell-mediated and antibody-mediated responses with optimized functional characteristics and induction of immune memory. AS01-containing vaccines appear relatively impervious to baseline immune status translating into high efficacy across populations. Currently licensed AS01-containing vaccines have shown acceptable safety profiles in clinical trials and post-marketing settings. EXPERT OPINION: Initial expectations that adjuvantation with AS01 could support effective vaccine responses and contribute to disease control have been realized. Investigation of the utility of AS01 in vaccines to prevent other challenging diseases, such as tuberculosis, is ongoing, together with efforts to fully define its mechanisms of action in different vaccine settings.


Adjuvants are added to vaccines to increase the immune response produced after vaccination. Adjuvant Systems contain two or more molecules that stimulate the immune system. AS01 is an Adjuvant System that contains two components, MPL and QS-21, that stimulate the immune system. AS01 is included in three approved vaccines: a malaria vaccine for children, a herpes zoster vaccine for older adults, and a respiratory syncytial virus vaccine also for older adults. Vaccines containing AS01 have been extensively evaluated in clinical trials and administered to millions of individuals during market use. These vaccines are effective in preventing disease and have acceptable safety in different age groups. Experiments have been done to investigate how AS01 works in vaccines to produce an efficient immune response that helps to protect against the disease being targeted. A key effect of AS01 is to encourage specific immune cells to produce chemicals that stimulate the immune system. We now know that this effect is due to co-operation between MPL and QS-21. Experiments have shown that AS01 induces a sophisticated immune 'gene signature' in blood within 24 h after vaccination, and people who developed this 'gene signature' had a stronger response to vaccination. AS01 seems to be able to stimulate the immune system of most people ­ even if they are older or have a weakened immune system. This means that AS01 could be included in other vaccines against other challenging diseases, such as tuberculosis, or could be used in the treatment of some disease, such as chronic hepatitis B.


Assuntos
Adjuvantes Imunológicos , Adjuvantes de Vacinas , Saponinas , Humanos , Saponinas/imunologia , Saponinas/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Lipídeo A/análogos & derivados , Lipídeo A/imunologia , Lipídeo A/farmacologia , Animais , Imunidade Inata/efeitos dos fármacos , Vacinas contra Vírus Sincicial Respiratório/imunologia , Lipossomos , Malária/prevenção & controle , Malária/imunologia , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Combinação de Medicamentos
14.
Cell Rep ; 43(8): 114533, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39052480

RESUMO

Ghana and other parts of West Africa have experienced lower COVID-19 mortality rates than other regions. This phenomenon has been hypothesized to be associated with previous exposure to infections such as malaria. This study investigated the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the influence of previous malaria exposure. Blood samples were collected from individuals with asymptomatic or symptomatic COVID-19 (n = 217). A variety of assays were used to characterize the SARS-CoV-2-specific immune response, and malaria exposure was quantified using Plasmodium falciparum ELISA. The study found evidence of attenuated immune responses to COVID-19 among asymptomatic individuals, with elevated proportions of non-classical monocytes and greater memory B cell activation. Symptomatic patients displayed higher P. falciparum-specific T cell recall immune responses, whereas asymptomatic individuals demonstrated elevated P. falciparum antibody levels. Summarily, this study suggests that P. falciparum exposure-associated immune modulation may contribute to reduced severity of SARS-CoV-2 infection among people living in malaria-endemic regions.


Assuntos
COVID-19 , Malária Falciparum , Plasmodium falciparum , SARS-CoV-2 , Humanos , COVID-19/imunologia , SARS-CoV-2/imunologia , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Plasmodium falciparum/imunologia , Malária Falciparum/imunologia , Malária Falciparum/epidemiologia , Imunidade Celular , Doenças Endêmicas , Adulto Jovem , Idoso , Gana/epidemiologia , Linfócitos T/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Adolescente , Malária/imunologia , Monócitos/imunologia
15.
PLoS One ; 19(7): e0306664, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38968270

RESUMO

BACKGROUNDS: Malaria, a preventive and treatable disease, is still responsible for annual deaths reported in most tropical regions, principally in sub-Saharan Africa. Subunit recombinant transmission-blocking vaccines (TBVs) have been proposed as promising vaccines to succeed in malaria elimination and eradication. Here, a provisional study was designed to assess the immunogenicity and functional activity of alanyl aminopeptidase N (APN1) of Anopheles stephensi, as a TBV candidate, administered with MPL, CpG, and QS21 adjuvants in the murine model. METHODOLOGY/PRINCIPAL FINDINGS: The mouse groups were immunized with recombinant APN1 (rAPN1) alone or formulated with CpG, MPL, QS-21, or a combination of adjuvants (CMQ), and the elicited immune responses were evaluated after the third immunization. The standard membrane feeding assay (SMFA) measured the functional activity of antibodies against bacterial-expressed APN1 protein in adjuvanted vaccine groups on transmission of P. falciparum (NF54) to An. stephensi mosquitoes. Evaluation of mice vaccinated with rAPN1 formulated with distinct adjuvants manifested a significant increase in the high-avidity level of anti-APN1 IgG and IgG subclasses; however, rAPN1 induced the highest level of high-avidity anti-APN1 IgG1, IgG2a, and IgG2b antibodies in the immunized vaccine group 5 (APN1/CMQ). In addition, vaccine group 5 (receiving APN1/CMQ), had still the highest level of anti-APN1 IgG antibodies relative to other immunized groups after six months, on day 180. The SMFA data indicates a trend towards higher transmission-reducing activity in groups 2 and 5, which received the antigen formulated with CpG or a combination of three adjuvants. CONCLUSIONS/SIGNIFICANCE: The results have shown the capability of admixture to stimulate high-affinity and long-lasting antibodies against the target antigen to hinder Plasmodium parasite development in the mid-gut of An. stephensi. The attained results authenticated APN1/CMQ and APN1/CpG as a potent APN1-based TBV formulation which will be helpful in designing a vaccine in the future.


Assuntos
Adjuvantes Imunológicos , Anopheles , Antígenos CD13 , Vacinas Antimaláricas , Saponinas , Animais , Anopheles/parasitologia , Anopheles/imunologia , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Camundongos , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Saponinas/farmacologia , Saponinas/administração & dosagem , Antígenos CD13/imunologia , Antígenos CD13/metabolismo , Feminino , Plasmodium falciparum/imunologia , Malária/prevenção & controle , Malária/transmissão , Malária/imunologia , Malária/parasitologia , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Camundongos Endogâmicos BALB C , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Malária Falciparum/imunologia , Malária Falciparum/parasitologia
16.
PLoS Pathog ; 20(7): e1012352, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39024388

RESUMO

CD4+ T cells are central mediators of protective immunity to blood-stage malaria, particularly for their capacity in orchestrating germinal center reaction and generating parasite-specific high-affinity antibodies. T follicular helper (Tfh) cells are predominant CD4+ effector T cell subset implicated in these processes, yet the factors and detailed mechanisms that assist Tfh cell development and function during Plasmodium infection are largely undefined. Here we provide evidence that receptor for activated C kinase 1 (RACK1), an adaptor protein of various intracellular signals, is not only important for CD4+ T cell expansion as previously implied but also plays a prominent role in Tfh cell differentiation and function during blood-stage Plasmodium yoelii 17XNL infection. Consequently, RACK1 in CD4+ T cells contributes significantly to germinal center formation, parasite-specific IgG production, and host resistance to the infection. Mechanistic exploration detects specific interaction of RACK1 with STAT3 in P. yoelii 17XNL-responsive CD4+ T cells, ablation of RACK1 leads to defective STAT3 phosphorylation, accompanied by substantially lower amount of STAT3 protein in CD4+ T cells, whereas retroviral overexpression of RACK1 or STAT3 in RACK1-deficient CD4+ T cells greatly restores STAT3 activity and Bcl-6 expression under the Tfh polarization condition. Further analyses suggest RACK1 positively regulates STAT3 stability by inhibiting the ubiquitin-proteasomal degradation process, thus promoting optimal STAT3 activity and Bcl-6 induction during Tfh cell differentiation. These findings uncover a novel mechanism by which RACK1 participates in posttranslational regulation of STAT3, Tfh cell differentiation, and subsequent development of anti-Plasmodium humoral immunity.


Assuntos
Diferenciação Celular , Malária , Plasmodium yoelii , Receptores de Quinase C Ativada , Fator de Transcrição STAT3 , Células T Auxiliares Foliculares , Animais , Receptores de Quinase C Ativada/metabolismo , Fator de Transcrição STAT3/metabolismo , Malária/imunologia , Malária/parasitologia , Camundongos , Plasmodium yoelii/imunologia , Células T Auxiliares Foliculares/imunologia , Células T Auxiliares Foliculares/metabolismo , Camundongos Endogâmicos C57BL , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Camundongos Knockout , Centro Germinativo/imunologia
18.
Nat Rev Immunol ; 24(9): 637-653, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38862638

RESUMO

Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/imunologia , Animais , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Imunidade Adaptativa/imunologia , Malária/imunologia , Malária/parasitologia , Interações Hospedeiro-Parasita/imunologia
19.
Nat Commun ; 15(1): 5497, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38944658

RESUMO

Children in malaria-endemic regions can experience repeated Plasmodium infections over short periods of time. Effects of re-infection on multiple co-existing CD4+ T cell subsets remain unresolved. Here, we examine antigen-experienced CD4+ T cells during re-infection in mice, using scRNA-seq/TCR-seq and spatial transcriptomics. TCR transgenic TEM cells initiate rapid Th1/Tr1 recall responses prior to proliferating, while GC Tfh counterparts are refractory, with TCM/Tfh-like cells exhibiting modest non-proliferative responses. Th1-recall is a partial facsimile of primary Th1-responses, with no upregulated effector-associated genes being unique to recall. Polyclonal, TCR-diverse, CD4+ T cells exhibit similar recall dynamics, with individual clones giving rise to multiple effectors including highly proliferative Th1/Tr1 cells, as well as GC Tfh and Tfh-like cells lacking proliferative capacity. Thus, we show substantial diversity in recall responses mounted by multiple co-existing CD4+ T cell subsets in the spleen, and present graphical user interfaces for studying gene expression dynamics and clonal relationships during re-infection.


Assuntos
Linfócitos T CD4-Positivos , Malária , Reinfecção , Animais , Malária/imunologia , Malária/parasitologia , Linfócitos T CD4-Positivos/imunologia , Camundongos , Reinfecção/imunologia , Células Th1/imunologia , Camundongos Endogâmicos C57BL , Baço/imunologia , Baço/parasitologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/genética , Camundongos Transgênicos , Feminino , Memória Imunológica
20.
Parasites Hosts Dis ; 62(2): 193-204, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38835260

RESUMO

Malaria is a global disease affecting a large portion of the world's population. Although vaccines have recently become available, their efficacies are suboptimal. We generated virus-like particles (VLPs) that expressed either apical membrane antigen 1 (AMA1) or microneme-associated antigen (MIC) of Plasmodium berghei and compared their efficacy in BALB/c mice. We found that immune sera acquired from AMA1 VLP- or MIC VLP-immunized mice specifically interacted with the antigen of choice and the whole P. berghei lysate antigen, indicating that the antibodies were highly parasite-specific. Both VLP vaccines significantly enhanced germinal center B cell frequencies in the inguinal lymph nodes of mice compared with the control, but only the mice that received MIC VLPs showed significantly enhanced CD4+ T cell responses in the blood following P. berghei challenge infection. AMA1 and MIC VLPs significantly suppressed TNF-α and interleukin-10 production but had a negligible effect on interferon-γ. Both VLPs prevented excessive parasitemia buildup in immunized mice, although parasite burden reduction induced by MIC VLPs was slightly more effective than that induced by AMA1. Both VLPs were equally effective at preventing body weight loss. Our findings demonstrated that the MIC VLP was an effective inducer of protection against murine experimental malaria and should be the focus of further development.


Assuntos
Antígenos de Protozoários , Vacinas Antimaláricas , Proteínas de Membrana , Plasmodium berghei , Proteínas de Protozoários , Vacinas de Partículas Semelhantes a Vírus , Animais , Feminino , Camundongos , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/imunologia , Malária/prevenção & controle , Malária/imunologia , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Proteínas de Membrana/imunologia , Camundongos Endogâmicos BALB C , Parasitemia/imunologia , Parasitemia/prevenção & controle , Plasmodium berghei/imunologia , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...