Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.419
Filter
1.
Braz J Biol ; 84: e284001, 2024.
Article in English | MEDLINE | ID: mdl-39319928

ABSTRACT

The clinical manifestations of cutaneous leishmaniasis (CL) depend not only on the infecting species involved, but also on the immune response of the individual. Although not yet well understood in humans, parasite survival and persistence are related to the cytokine profile and T cell proliferation, with the Th1 profile being related to cure, and the Th2 profile to disease progression. Considering the need for studies focused on the species with the highest circulation in the state of Amazonas, this study aimed to analyze the immunoregulation stimulated by soluble antigens (SLAs) of Leishmania (L.) amazonensis and Leishmania (V.) guyanensis in human lymphocytes in vitro, in order to understand the immune response of patients with CL. Lymphoproliferation was evaluated against stimuli of SLAs from L. amazonensis (100 µg/mL), SLAs from L. guyanensis (100 µg/mL) and phytohemagglutinin (10 µg/mL) using a BrdU Cell Proliferation ELISA kit after 72 h of incubation. Quantification of the cytokines IL-1b, IL-6, IL-8, IL-10, IL-12 and TNF was performed using the BD™ cytometric bead array human Th1/Th2/Th17 cytokine kit. Our results demonstrated that soluble antigens from L. amazonensis and L. guyanensis stimulated the lymphoproliferation of PBMCs from patients primo-infected with CL. Among the cytokines dosed, the highest concentrations were of IL-6 and IL-8, thus demonstrating that the soluble antigens evaluated are capable of inducing regulatory mechanisms.


Subject(s)
Antigens, Protozoan , Cell Proliferation , Cytokines , Humans , Antigens, Protozoan/immunology , Cytokines/immunology , Cell Proliferation/drug effects , Leishmaniasis, Cutaneous/immunology , Adult , Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Male , Female , Leishmania guyanensis/immunology , Young Adult
3.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337297

ABSTRACT

We developed a protein to rapidly and accurately diagnose Chagas disease, a life-threatening illness identified by the WHO as a critical worldwide public health risk. Limitations in present day serological tests are complicating the current health situation and contributing to most infected persons being unaware of their condition and therefore untreated. To improve diagnostic testing, we developed an immunological mimic of the etiological agent, Trypanosoma cruzi, by combining ten pathogen-specific epitopes within the beta-barrel protein structure of Thermal Green Protein. The resulting multi-epitope protein, DxCruziV3, displayed high specificity and sensitivity as the antibody capture reagent in an ELISA platform with an analytical sensitivity that exceeds WHO recommendations. Within an immunochromatographic platform, DxCruziV3 showed excellent performance for the point of application diagnosis in a region endemic for multiple diseases, the municipality of Barcelos in the state of Amazonas, Brazil. In total, 167 individuals were rapidly tested using whole blood from a finger stick. As recommended by the Brazilian Ministry of Health, venous blood samples were laboratory tested by conventional assays for comparison. Test results suggest utilizing DxCruziV3 in different assay platforms can confidently diagnose chronic infections by T. cruzi. Rapid and more accurate results will benefit everyone but will have the most noticeable impact in resource-limited rural areas where the disease is endemic.


Subject(s)
Chagas Disease , Enzyme-Linked Immunosorbent Assay , Epitopes , Serologic Tests , Trypanosoma cruzi , Chagas Disease/diagnosis , Chagas Disease/blood , Chagas Disease/immunology , Humans , Enzyme-Linked Immunosorbent Assay/methods , Trypanosoma cruzi/immunology , Serologic Tests/methods , Epitopes/immunology , Chronic Disease , Male , Sensitivity and Specificity , Female , Adult , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Middle Aged , Antigens, Protozoan/immunology , Antigens, Protozoan/blood , Brazil/epidemiology
4.
Parasit Vectors ; 17(1): 402, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334233

ABSTRACT

BACKGROUND: Laboratory diagnosis of American cutaneous leishmaniasis (ACL) requires a tool amenable to the epidemiological status of ACL in Brazil. Montenegro skin test (MST), an efficient immunological tool used for laboratory diagnosis of ACL, induces delayed-type hypersensitivity (DTH) response to the promastigote antigens of Leishmania; however, human immune responses against infection are modulated by the amastigote of the parasite. Leishmania (V.) lainsoni induces strong cellular immunity in humans; therefore, the antigenic reactivity of its axenic amastigote (AMA antigen) to MST was evaluated for the laboratory diagnosis of ACL. METHODS: Among 70 individuals examined, 60 had a laboratory-confirmed diagnosis of ACL; 53 had localized cutaneous leishmaniasis (LCL), and 7 had mucosal leishmaniasis (ML). Patients were treated at the Evandro Chagas Institute's leishmaniasis clinic, Pará State, Brazil. Ten healthy individuals with no history of ACL (control group) were also examined. Leishmania (V.) braziliensis promastigote antigen (PRO) was used to compare the reactivity with that of AMA antigen. Paired Student's t-test, kappa agreement, and Spearman test were used to evaluate the reactivity of AMA and PRO. RESULTS: The mean reactivity of AMA in ACL patients was 19.4 mm ± 13.3, which was higher (P < 0.001) than that of PRO: 12.1 mm ± 8.1. MST reactivity according to the clinical forms revealed that AMA reactivity in LCL and ML, 18.8 mm ± 13.3 and 24.3 mm ± 13.7, was higher (P < 0.001) than that of PRO, 11.8 mm ± 8.2 and 14.6 mm ± 8.4, respectively. CONCLUSION: AMA reactivity was higher than that of PRO, indicating that AMA is a promising alternative for optimizing MST in the laboratory diagnosis of ACL.


Subject(s)
Antigens, Protozoan , Leishmania , Leishmaniasis, Cutaneous , Skin Tests , Humans , Antigens, Protozoan/immunology , Skin Tests/methods , Adult , Female , Leishmaniasis, Cutaneous/diagnosis , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Male , Brazil , Leishmania/immunology , Middle Aged , Young Adult , Adolescent
5.
Vet Parasitol ; 331: 110299, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232468

ABSTRACT

Canine Visceral Leishmaniasis (CVL) is the most fatal form of Leishmania infection in dogs and is caused by L. infantum in the Americas. This parasite follows a zoonotic life cycle, raising concerns within domestic households, where dogs act as the primary reservoir of the parasite. Accurately detecting infected dogs is vital for effective epidemiological control in both canine and human populations. However, existing diagnostic methods in Brazil have limitations, particularly in detecting asymptomatic and oligosymptomatic dogs, leading to ineffective disease control. To address this challenge, we evaluated a novel recombinant antigen from L. infantum, the rLiNTPDase2. Previous studies have confirmed its high performance via ELISA, leading us to assess its suitability for a Lateral Flow Immunochromatographic Assay (LFIA), which is ideal for point-of-care testing. Standardization of the assay involved testing two nitrocellulose membranes (HF135 and HF120, Millipore), three blocking protocols, and five sample dilutions (1:10, 1:20, 1:40, 1:80, and 1:160). Following the chosen conditions (HF120 membrane, 1-minute blocking protocol, and 1:80 sample dilution), we validated our assay with a sample size of 78 dogs, comprising 32 negatives and 46 positives, including symptomatic (n=23), oligosymptomatic (n=17), and asymptomatic (n=6) cases. The results revealed a sensitivity of 86.9 %, specificity of 62.5 %, and accuracy of 76.9 %, which is consistent with ELISA performance for the same samples. Compared to DPP-LVC, our assay demonstrated promising results in detecting asymptomatic and oligosymptomatic cases. This study underscores the suitability of the rLiNTPDase2 antigen for the LFIA format, suggesting its potential as a novel point-of-care diagnostic test for CVL.


Subject(s)
Antigens, Protozoan , Dog Diseases , Leishmaniasis, Visceral , Sensitivity and Specificity , Animals , Dogs , Leishmaniasis, Visceral/veterinary , Leishmaniasis, Visceral/diagnosis , Leishmaniasis, Visceral/parasitology , Dog Diseases/diagnosis , Dog Diseases/parasitology , Antigens, Protozoan/immunology , Antigens, Protozoan/analysis , Chromatography, Affinity/veterinary , Chromatography, Affinity/methods , Leishmania infantum/enzymology , Leishmania infantum/immunology , Recombinant Proteins/immunology , Enzyme-Linked Immunosorbent Assay/veterinary , Enzyme-Linked Immunosorbent Assay/methods
6.
Vet Parasitol ; 331: 110297, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236398

ABSTRACT

Studies in various species have demonstrated different results on the effects of T. gondii infection on sperm quality. It has also been demonstrated that in some stages of the disease, there is elimination of cellular debris or even the intact parasite in the semen. The present work aimed to evaluate the in vitro effects of the presence of soluble T. gondii antigens in bovine semen on sperm integrity. The spermatozoa were treated with T. gondii antigens in double serial dilutions classified as high, medium and low doses (8, 4, 2 µg/ml) in "TALP-Sperm" and "TALP-Fert" media. The results showed that T. gondii antigens affect sperm motility and mitochondrial activity, and cause changes in sperm chromatin integrity, as well as damage to the sperm membrane and acrosome. Finally, spermatozoa treated with T. gondii antigens were evaluated in the in vitro production of embryos (IVEP). The use of semen contaminated with antigens in IVEP routines did not lead to a decrease in the fertilization of oocytes, as sperm undergo selection before in vitro fertilization, which eliminates the most altered sperm. However, early embryonic development was affected, probably by structural changes that were not eliminated in the selection process. The results demonstrated that the presence of soluble T. gondii antigens in bovine semen alters sperm integrity and vital characteristics for the fertilization process and embryonic development and therefore causes fertility problems in males.


Subject(s)
Antigens, Protozoan , Fertility , Spermatozoa , Toxoplasma , Animals , Cattle , Male , Spermatozoa/immunology , Toxoplasma/immunology , Toxoplasma/physiology , Antigens, Protozoan/immunology , Sperm Motility , Fertilization in Vitro/veterinary , Semen/parasitology , Semen/immunology
7.
Front Cell Infect Microbiol ; 14: 1451063, 2024.
Article in English | MEDLINE | ID: mdl-39258252

ABSTRACT

Background: Transmission-blocking vaccines (TBVs) can effectively prevent the community's spread of malaria by targeting the antigens of mosquito sexual stage parasites. At present, only a few candidate antigens have demonstrated transmission-blocking activity (TBA) potential in P. vivax. Quiescin-sulfhydryl oxidase (QSOX) is a sexual stage protein in the rodent malaria parasite Plasmodium berghei and is associated with a critical role in protein folding by introducing disulfides into unfolded reduced proteins. Here, we reported the immunogenicity and transmission-blocking potency of the PvQSOX in P. vivax. Methods and findings: The full-length recombinant PvQSOX protein (rPvQSOX) was expressed in the Escherichia coli expression system. The anti-rPvQSOX antibodies were generated following immunization with the rPvQSOX in rabbits. A parasite integration of the pvqsox gene into the P. berghei pbqsox gene knockout genome was developed to express full-length PvQSOX protein in P. berghei (Pv-Tr-PbQSOX). In western blot, the anti-rPvQSOX antibodies recognized the native PvQSOX protein expressed in transgenic P. berghei gametocyte and ookinete. In indirect immunofluorescence assays, the fluorescence signal was detected in the sexual stages, including gametocyte, gamete, zygote, and ookinete. Anti-rPvQSOX IgGs obviously inhibited the ookinetes and oocysts development both in vivo and in vitro using transgenic parasites. Direct membrane feeding assays of anti-rPvQSOX antibodies were conducted using four field P. vivax isolates (named isolates #1-4) in Thailand. Oocyst density in mosquitoes was significantly reduced by 32.00, 85.96, 43.52, and 66.03% with rabbit anti-rPvQSOX antibodies, respectively. The anti-rPvQSOX antibodies also showed a modest reduction of infection prevalence by 15, 15, 20, and 22.22%, respectively, as compared to the control, while the effect was insignificant. The variation in the DMFA results may be unrelated to the genetic polymorphisms. Compared to the P.vivax Salvador (Sal) I strain sequences, the pvqsox in isolate #1 showed no amino acid substitution, whereas isolates #2, #3, and #4 all had the M361I substitution. Conclusions: Our results suggest that PvQSOX could serve as a potential P. vivax TBVs candidate, which warrants further evaluation and optimization.


Subject(s)
Antibodies, Protozoan , Malaria Vaccines , Malaria, Vivax , Plasmodium berghei , Plasmodium vivax , Recombinant Proteins , Plasmodium vivax/immunology , Plasmodium vivax/genetics , Plasmodium vivax/enzymology , Animals , Rabbits , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Malaria, Vivax/immunology , Plasmodium berghei/immunology , Plasmodium berghei/genetics , Plasmodium berghei/enzymology , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Mice , Escherichia coli/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Female , Humans , Immunogenicity, Vaccine , Mice, Inbred BALB C
8.
BMC Med ; 22(1): 388, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267089

ABSTRACT

BACKGROUND: Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) proteins are expressed on the surface of infected erythrocytes, mediating parasite sequestration in the vasculature. PfEMP1 is a major target of protective antibodies, but the features of the antibody response are poorly defined. METHODS: In Malawian children with cerebral or uncomplicated malaria, we characterized the antibody response to 39 recombinant PfEMP1 Duffy binding like (DBL) domains or cysteine-rich interdomain regions (CIDRs) in detail, including measures of antibody classes, subclasses, and engagement with Fcγ receptors and complement. Using elastic net regularized logistic regression, we identified a combination of seven antibody targets and Fc features that best distinguished between children with cerebral and uncomplicated malaria. To confirm the role of the selected targets and Fc features, we measured antibody-dependent neutrophil and THP-1 cell phagocytosis of intercellular adhesion molecule-1 (ICAM-1) and endothelial protein C (EPCR) co-binding infected erythrocytes. RESULTS: The selected features distinguished between children with cerebral and uncomplicated malaria with 87% accuracy (median, 80-96% interquartile range) and included antibody to well-characterized DBLß3 domains and a less well-characterized CIDRγ12 domain. The abilities of antibodies to engage C1q and FcγRIIIb, rather than levels of IgG, correlated with protection. In line with a role of FcγRIIIb binding antibodies to DBLß3 domains, antibody-dependent neutrophil phagocytosis of ICAM-1 and EPCR co-binding IE was higher in uncomplicated malaria (15% median, 8-38% interquartile range) compared to cerebral malaria (7%, 30-15%, p < 0.001). CONCLUSIONS: Antibodies associated with protection from cerebral malaria target a subset of PfEMP1 domains. The Fc features of protective antibody response include engagement of FcγRIIIb and C1q, and ability to induce antibody-dependent neutrophil phagocytosis of infected erythrocytes. Identifying the targets and Fc features of protective immunity could facilitate the development of PfEMP1-based therapeutics for cerebral malaria.


Subject(s)
Antibodies, Protozoan , Malaria, Cerebral , Plasmodium falciparum , Protozoan Proteins , Humans , Malaria, Cerebral/immunology , Malawi , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Protozoan Proteins/immunology , Child, Preschool , Plasmodium falciparum/immunology , Male , Female , Child , Infant , Intercellular Adhesion Molecule-1/immunology , Endothelial Protein C Receptor/immunology , Phagocytosis , Erythrocytes/parasitology , Erythrocytes/immunology , Malaria, Falciparum/immunology , Antigens, Protozoan/immunology
9.
Front Immunol ; 15: 1400308, 2024.
Article in English | MEDLINE | ID: mdl-39234242

ABSTRACT

Tropical theileriosis is a lymphoproliferative disease caused by Theileria annulata and is transmitted by Ixodid ticks of the genus Hyalomma. It causes significant losses in livestock, especially in exotic cattle. The existing methods for controlling it, chemotherapeutic agents and a vaccine based on an attenuated schizont stage parasite, have several limitations. A promising solution to control this disease is the use of molecular vaccines based on potential immunogenic proteins of T. annulata. For this purpose, we selected five antigenic sequences of T. annulata, i.e. SPAG-1, Tams, TaSP, spm2, and Ta9. These were subjected to epitope prediction for cytotoxic T lymphocytes, B-cells, and helper T lymphocytes. CTL and B-cell epitopes with a higher score whereas those of HTL with a lower score, were selected for the construct. A single protein was constructed using specific linkers and evaluated for high antigenicity and low allergenicity. The construct was acidic, hydrophobic, and thermostable in nature. Secondary and tertiary structures of this construct were drawn using the PSIPRED and RaptorX servers, respectively. A Ramachandran plot showed a high percentage of residues in this construct in favorable, allowed, and general regions. Molecular docking studies suggested that the complex was stable and our construct could potentially be a good candidate for immunization trials. Furthermore, we successfully cloned it into the pET-28a plasmid and transformed it into the BL21 strain. A restriction analysis was performed to confirm the transformation of our plasmid. After expression and purification, recombinant protein of 49 kDa was confirmed by western blotting. An ELISA detected increased specific antibody levels in the sera of the immunized animals compared with the control group, and flow cytometric analysis showed a stronger cell-mediated immune response. We believe our multi-epitope recombinant protein has the potential for the large-scale application for disease prevention globally in the bovine population. This study will act as a model for similar parasitic challenges.


Subject(s)
Immunity, Cellular , Immunity, Humoral , Recombinant Proteins , Theileria annulata , Theileriasis , Theileria annulata/immunology , Theileria annulata/genetics , Animals , Cattle , Theileriasis/immunology , Theileriasis/parasitology , Theileriasis/prevention & control , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Protozoan Vaccines/immunology , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Computer Simulation , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood
10.
Parasites Hosts Dis ; 62(3): 302-312, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39218629

ABSTRACT

Plasmodium falciparum apical membrane antigen-1 (PfAMA-1) is a major candidate for the blood-stage malaria vaccine. Genetic polymorphisms of global pfama-1suggest that the genetic diversity of the gene can disturb effective vaccine development targeting this antigen. This study was conducted to explore the genetic diversity and gene structure of pfama-1 among P. falciparum isolates collected in the Khyber Pakhtunkhwa (KP) province of Pakistan. A total of 19 full-length pfama-1 sequences were obtained from KP-Pakistan P. falciparum isolates, and genetic polymorphism and natural selection were investigated. KP-Pakistan pfama-1 exhibited genetic diversity, wherein 58 amino acid changes were identified, most of which were located in ectodomains, and domains I, II, and III. The amino acid changes commonly found in the ectodomain of global pfama-1 were also detected in KP-Pakistan pfama-1. Interestingly, 13 novel amino acid changes not reported in the global population were identified in KP-Pakistan pfama-1. KP-Pakistan pfama-1 shared similar levels of genetic diversity with global pfama-1. Evidence of natural selection and recombination events were also detected in KP-Pakistan pfama-1.


Subject(s)
Antigens, Protozoan , Malaria, Falciparum , Membrane Proteins , Plasmodium falciparum , Polymorphism, Genetic , Protozoan Proteins , Pakistan , Plasmodium falciparum/genetics , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Membrane Proteins/genetics , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/epidemiology , Genetic Variation/genetics , Selection, Genetic , Phylogeny , Recombination, Genetic/genetics
11.
Hum Vaccin Immunother ; 20(1): 2374147, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39090779

ABSTRACT

Entamoeba histolytica, the causative agent of amebiasis, is one of the top three parasitic causes of mortality worldwide. However, no vaccine exists against amebiasis. Using a lead candidate vaccine containing the LecA fragment of Gal-lectin and GLA-3M-052 liposome adjuvant, we immunized rhesus macaques via intranasal or intramuscular routes. The vaccine elicited high-avidity functional humoral responses as seen by the inhibition of amebic attachment to mammalian target cells by plasma and stool antibodies. Importantly, antigen-specific IFN-γ-secreting peripheral blood mononuclear cells (PBMCs) and IgG/IgA memory B cells (BMEM) were detected in immunized animals. Furthermore, antigen-specific antibody and cellular responses were maintained for at least 8 months after the final immunization as observed by robust LecA-specific BMEM as well as IFN-γ+ PBMC responses. Overall, both intranasal and intramuscular immunizations elicited a durable and functional response in systemic and mucosal compartments, which supports advancing the LecA+GLA-3M-052 liposome vaccine candidate to clinical testing.


Subject(s)
Administration, Intranasal , Antibodies, Protozoan , Entamoeba histolytica , Entamoebiasis , Interferon-gamma , Leukocytes, Mononuclear , Liposomes , Macaca mulatta , Protozoan Vaccines , Animals , Entamoeba histolytica/immunology , Liposomes/immunology , Liposomes/administration & dosage , Protozoan Vaccines/immunology , Protozoan Vaccines/administration & dosage , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Leukocytes, Mononuclear/immunology , Entamoebiasis/prevention & control , Entamoebiasis/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Injections, Intramuscular , Immunogenicity, Vaccine , Adjuvants, Vaccine/administration & dosage , Adjuvants, Immunologic/administration & dosage , B-Lymphocytes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Antigens, Protozoan/immunology , Immunity, Humoral , Immunologic Memory , Protozoan Proteins/immunology
12.
J Infect ; 89(4): 106252, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182654

ABSTRACT

OBJECTIVES: Acquisition of antibodies to Plasmodium falciparum variant surface antigens (VSA) expressed on infected red blood cells (iRBCs) is associated with naturally acquired immunity to malaria. We have previously shown that antibodies to VSA on iRBCs are associated with protection against parasite growth in the context of controlled human malaria infection (CHMI). This study explored whether antibodies to recombinant antigens derived from PfEMP1 domains were independently associated with protection during CHMI in semi-immune Kenyan adults. METHODS: We used a multiplex bead assay to measure levels of IgG antibody against a panel of 27 recombinant PfEMP1 antigens derived from the PfEMP1 repertoire of the 3D7 parasite clone. We measured IgG levels in plasma samples collected from the CHMI participants before inoculation with Sanaria® PfSPZ Challenge, on the day of diagnosis, and 35 days post-inoculation. Univariable and multivariable Cox regression analysis was used to evaluate the relationship between the levels of antibodies to the antigens and CHMI outcome. We also adjusted for previous data including antibodies to VSA on iRBCs, and we assessed the kinetics of antibody acquisition to the different PfEMP1 recombinant antigens over time. RESULTS: All study participants had detectable antibodies to multiple PfEMP1 proteins before inoculation. All PfEMP1 antigens were associated with protection against parasite growth to the threshold criteria for treatment in CHMI, albeit with substantial collinearity. However, individual PfEMP1 antigens were not independently associated with protection following adjustment for breadth of reactivity to VSA on iRBCs and schizont extract. In addition, antibodies to PfEMP1 antigens derived from group B PfEMP1 were induced and sustained in the participants who could not control parasite growth. CONCLUSION: This study shows that the breadth of antibody response to VSA on iRBCs, and not to specific PfEMP1 antigens, is predictive of protection against malaria in CHMI.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Immunoglobulin G , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Humans , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Kenya , Antigens, Protozoan/immunology , Adult , Plasmodium falciparum/immunology , Male , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Young Adult , Antigens, Surface/immunology , Middle Aged , Adolescent
13.
J Infect ; 89(4): 106257, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39216830

ABSTRACT

Children with hemoglobin AC or AS have decreased susceptibility to clinical malaria. Parasite variant surface antigen (VSA) presentation on the surface of infected erythrocytes is altered in erythrocytes with hemoglobin C (Hb AC) or sickle trait (Hb AS) mutations in vitro. The protective role of incomplete or altered VSA presentation against clinical malaria in individuals with Hb AC or AS is unclear. Using a high-throughput protein microarray, we sought to use serological responses to VSAs as a measure of host exposure to VSAs among Malian children with Hb AC, Hb AS, or wildtype hemoglobin (Hb AA). In uncomplicated malaria, when compared to Hb AA children, Hb AC children had significantly lower serological responses to extracellular Plasmodium falciparum erythrocyte membrane protein-1 (PfEMP1) domains but did not differ in responses to intracellular PfEMP1 domains and other VSAs, including members of the repetitive interspersed family (RIFIN) and subtelomeric variable open reading frame (STEVOR) family. Healthy children with Hb AC and Hb AS genotypes recognized fewer extracellular PfEMP1s compared to children with Hb AA, especially CD36-binding PfEMP1s. These reduced serologic responses may reflect reduced VSA presentation or lower parasite exposure in children with Hb AC or AS and provide insights into mechanisms of protection.


Subject(s)
Antigens, Protozoan , Hemoglobin C , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Sickle Cell Trait , Humans , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Child, Preschool , Child , Plasmodium falciparum/immunology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Hemoglobin C/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/blood , Sickle Cell Trait/genetics , Sickle Cell Trait/blood , Sickle Cell Trait/immunology , Male , Female , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Hemoglobin, Sickle/genetics , Mali/epidemiology , Infant , Antigens, Surface/immunology , Antigens, Surface/genetics , Protein Array Analysis , Adolescent
14.
BMC Infect Dis ; 24(1): 886, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210269

ABSTRACT

BACKGROUND: The highly expressed surface antigen 1 (SAG1)-related sequence (SRS) proteins of T. gondii tachyzoites, as a widespread zoonotic parasite, are critical for host cell invasion and represent promising vaccine targets. In this study, we employed a computer-aided multi-method approach for in silico design and evaluation of TgVax452, an epitope-based candidate vaccine against T. gondii tachyzoite-specific SRS proteins. METHODS: Using immunoinformatics web-based tools, structural modeling, and static/dynamic molecular simulations, we identified and screened B- and T-cell immunodominant epitopes and predicted TgVax452's antigenicity, stability, safety, adjuvanticity, and physico-chemical properties. RESULTS: The designed protein possessed 452 residues, a MW of 44.07 kDa, an alkaline pI (6.7), good stability (33.20), solubility (0.498), and antigenicity (0.9639) with no allergenicity. Comprehensive molecular dynamic (MD) simulation analyses confirmed the stable interaction (average potential energy: 3.3799 × 106 KJ/mol) between the TLR4 agonist residues (RS09 peptide) of the TgVax452 in interaction with human TLR4, potentially activating innate immune responses. Also, a dramatic increase was observed in specific antibodies (IgM and IgG), cytokines (IFN-γ), and lymphocyte responses, based on C-ImmSim outputs. Finally, we optimized TgVax452's codon adaptation and mRNA secondary structure for efficient expression in E. coli BL21 expression machinery. CONCLUSION: Our findings suggest that TgVax452 is a promising candidate vaccine against T. gondii tachyzoite-specific SRS proteins and requires further experimental studies for its potential use in preclinical trials.


Subject(s)
Antigens, Protozoan , Computational Biology , Epitopes, T-Lymphocyte , Protozoan Proteins , Protozoan Vaccines , Toxoplasma , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Toxoplasma/immunology , Toxoplasma/genetics , Toxoplasma/chemistry , Protozoan Vaccines/immunology , Protozoan Vaccines/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/chemistry , Animals , Mice , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Female , Antibodies, Protozoan/immunology , Mice, Inbred BALB C , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/chemistry , Humans , Molecular Dynamics Simulation , Immunodominant Epitopes/immunology , Immunodominant Epitopes/genetics , Immunodominant Epitopes/chemistry , Toxoplasmosis/prevention & control , Toxoplasmosis/immunology , Immunoinformatics
15.
Acta Parasitol ; 69(3): 1613-1620, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39164549

ABSTRACT

Visceral Leishmaniasis is a serious public health problem caused by Leishmania species parasites. Approximately 500 thousand people get Visceral Leishmaniasis (VL) every year. An effective and reliable vaccine against the disease has still not been formulated. Choosing the right adjuvant is important to increase immunogenicity in vaccines prepared with total antigens. In this study, we investigate the ideal adjuvant for use in vaccine formulations against VL. For this purpose, Leishmania antigens (FTLA) obtained from L. infantum parasites by the freeze-thaw method and three different adjuvants (alum-saponin and calcium phosphate) were used. The effectiveness of the formulations was investigated in vitro by cell viability analysis and determination of nitric oxide and cytokine production abilities in J774 macrophage cells. According to the study results, it was determined that formulations prepared with calcium phosphate produced 72% more NO and approximately 7.2 times more IL-12 cytokine. The results obtained showed that calcium phosphate salts can be used as ideal adjuvants in vaccine research against leishmaniasis.


Subject(s)
Antigens, Protozoan , Leishmania infantum , Leishmaniasis Vaccines , Animals , Mice , Leishmaniasis Vaccines/immunology , Leishmania infantum/immunology , Antigens, Protozoan/immunology , Cell Line , Macrophages/immunology , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Nitric Oxide/metabolism , Calcium Phosphates , Cytokines/metabolism , Adjuvants, Vaccine , Leishmaniasis, Visceral/prevention & control , Leishmaniasis, Visceral/immunology , Saponins/pharmacology , Alum Compounds/administration & dosage , Adjuvants, Immunologic/administration & dosage , Cell Survival/drug effects
16.
Diagn Microbiol Infect Dis ; 110(3): 116480, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39163788

ABSTRACT

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.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Malaria , Membrane Proteins , Plasmodium malariae , Protozoan Proteins , Animals , Female , Mice , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Escherichia coli/genetics , Immunoglobulin G/blood , Malaria/diagnosis , Malaria/prevention & control , Malaria/immunology , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Membrane Proteins/immunology , Membrane Proteins/genetics , Mice, Inbred BALB C , Plasmodium malariae/immunology , Plasmodium malariae/genetics , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/genetics
17.
Vaccine ; 42(21): 126178, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39096765

ABSTRACT

American Tegumentary Leishmaniasis (ATL) is a disease of high severity and incidence in Brazil, in addition to being a worldwide concern in public health. Leishmania amazonensis is one of the etiological agents of ATL, and the inefficiency of control measures, associated with the high toxicity of the treatment and the lack of effective immunoprophylactic strategies, makes the development of vaccines indispensable and imminent. In this light, the present study proposes to elaborate a chimeric protein (rChiP), based on the fusion of multiple epitopes of CD4+/CD8+ T cells, identified in the immunoproteome of the parasites L. amazonensis and L. braziliensis. The designed chimeric protein was tested in the L. amazonensis murine model of infection using the following formulations: 25 µg of the rChiP in saline (rChiP group) and 25 µg of the rChiP plus 25 µg of MPLA-PHAD® (rChiP+MPLA group). After completing immunization, CD4+ and CD8+ T cells, stimulated with SLa-Antigen or rChiP, showed an increased production of nitric oxide and intracytoplasmic pro-inflammatory cytokines, in addition to the generation of central and effector memory T cells. rChiP and rChiP+MPLA formulations were able to promote an effective protection against L. amazonensis infection determined by a reduction in the development of skin lesions and lower parasitic burden. Reduction in the development of skin lesions and lower parasitic burden in the vaccinated groups were associated with an increase of nitrite, CD4+/CD8+IFN-γ+TNF-α+ and CD4+/CD8+CD44highCD62Lhigh/low T cells, IgGTotal, IgG2a, and lower rates of IgG1 and CD4+/CD8+IL-10+. This data suggests that proposed formulations could be considered potential tools to prevent ATL.


Subject(s)
Adjuvants, Immunologic , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Immunologic Memory , Leishmaniasis Vaccines , Leishmaniasis, Cutaneous , Animals , Leishmaniasis, Cutaneous/prevention & control , Leishmaniasis, Cutaneous/immunology , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Mice , Leishmaniasis Vaccines/immunology , Female , Adjuvants, Immunologic/administration & dosage , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Leishmania braziliensis/immunology , Lipid A/analogs & derivatives , Lipid A/immunology , Antibodies, Protozoan/immunology , Cytokines/metabolism , Cytokines/immunology , Disease Models, Animal , Antigens, Protozoan/immunology
18.
Nat Commun ; 15(1): 7206, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174515

ABSTRACT

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.


Subject(s)
Antigens, Protozoan , Erythrocytes , Liver , Membrane Proteins , Mice, Inbred BALB C , Protozoan Proteins , Animals , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Female , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Humans , Erythrocytes/parasitology , Erythrocytes/immunology , Liver/parasitology , Liver/immunology , Liver/metabolism , Malaria Vaccines/immunology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Cross Reactions/immunology , Plasmodium falciparum/immunology , Plasmodium berghei/immunology , Epitopes/immunology , Hepatocytes/parasitology , Hepatocytes/immunology , Hepatocytes/metabolism , Plasmodium/immunology , Merozoites/immunology , Merozoites/metabolism
19.
Nat Commun ; 15(1): 7487, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39209843

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Antibodies, Protozoan , Malaria Vaccines , Plasmodium falciparum , Plasmodium vivax , Antibodies, Monoclonal/immunology , Animals , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Mice , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/parasitology , Crystallography, X-Ray , Epitopes/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Antigens, Protozoan/immunology , Humans , Female , Epitope Mapping , Malaria/immunology , Malaria/prevention & control , Malaria/parasitology , Mice, Inbred BALB C , Protozoan Proteins/immunology , Protozoan Proteins/chemistry
20.
J Phys Chem B ; 128(35): 8400-8408, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39172501

ABSTRACT

Plasmodium falciparum apical membrane antigen 1 (PfAMA1) is a surface protein found in two stages of the malaria life cycle. This is a protein involved in a reorientation movement of the parasite so that cell invasion occurs in the so-called "moving junction", relevant when the membranes of the parasite and the host are in contact. The structure of a conformational epitope of domain III of PfAMA1 in complex with the monoclonal antibody Fab F8.12.19 is experimentally known. Here, we used molecular dynamics with enhanced sampling by Hamiltonian replica exchange molecular dynamics (HREMD) to understand the effect of intermolecular interactions, conformational variability, and intrinsically disordered regions on the mechanism of antigen-antibody interaction. Clustering methods and the analysis of conformational variability were used in order to understand the influence of the presence of the partner protein in the complex. The free-state epitope accesses a broader conformational pool, including disordered conformations not seen in the bound state. The simulations suggest an extended conformational selection mechanism in which the antibody stabilizes a conformational set of the epitope existing in the free state. The stabilization of the active conformation occurs mainly through hydrogen bonds: Tyr(H33)-Asp493, His(L94)-Val510, Ser(L93)-Glu511, Tyr(H56)-Asp485, and Tyr(H35)-Asp493. The antibody has a structure with few flexible regions, and only the complementarity determining region (CDR) H3 shows greater plasticity in the presence of the epitope.


Subject(s)
Antigens, Protozoan , Membrane Proteins , Molecular Dynamics Simulation , Plasmodium falciparum , Protozoan Proteins , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/chemistry , Protein Conformation , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology
SELECTION OF CITATIONS
SEARCH DETAIL