Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Front Med (Lausanne) ; 11: 1342476, 2024.
Article in English | MEDLINE | ID: mdl-38808136

ABSTRACT

Human Immunodeficiency Virus (HIV) remains a global health challenge, and novel approaches to improve HIV control are significantly important. The cell and gene therapy product AGT103-T was previously evaluated (NCT04561258) for safety, immunogenicity, and persistence in seven patients for up to 180 days post infusion. In this study, we sought to investigate the impact of AGT103-T treatment upon analytical treatment interruptions (ATIs). Six patients previously infused with AGT103-T were enrolled into an ATI study (NCT05540964), wherein they suspended their antiretroviral therapy (ART) until their viral load reached 100,000 copies/mL in two successive visits, or their CD4 count was reduced to below 300 cells/µL. During the ATI, all patients experienced viral rebound followed by a notable expansion in HIV specific immune responses. The participants demonstrated up to a five-fold increase in total CD8 counts over baseline approximately 1-2 weeks followed by the peak viremia. This coincided with a rise in HIV-specific CD8 T cells, which was attributed to the increase in antigen availability and memory recall. Thus, the protocol was amended to include a second ATI with the first ATI serving as an "auto-vaccination." Four patients participated in a second ATI. During the second ATI, the Gag-specific CD8 T cells were either maintained or rose in response to viral rebound and the peak viremia was substantially decreased. The patients reached a viral set point ranging from 7,000 copies/mL to 25,000 copies/mL. Upon resuming ART, all participants achieved viral control more rapidly than during the first ATI, with CD4 counts remaining within 10% of baseline measurements and without any serious adverse events or evidence of drug resistance. In summary, the rise in CD8 counts and the viral suppression observed in 100% of the study participants are novel observations demonstrating that AGT103-T gene therapy when combined with multiple ATIs, is a safe and effective approach for achieving viral control, with viral setpoints consistently below 25,000 copies/mL and relatively stable CD4 T cell counts. We conclude that HIV cure-oriented cell and gene therapy trials should include ATI and may benefit from designs that include multiple ATIs when induction of CD8 T cells is required to establish viral control.

2.
Microbiol Spectr ; 11(6): e0094023, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37982627

ABSTRACT

IMPORTANCE: Malaria transmission by Anopheles gambiae mosquitoes is very effective, in part because the parasite expresses a surface protein called Pfs47 that allows it to evade the mosquito immune system. Here we investigate how this protein changes the response of mosquito midgut epithelial cells to invasion by the parasite. Pfs47 is known to interact with P47Rec, a mosquito midgut receptor. We found that Pf47Rec inhibits caspase-mediated apoptosis by interacting with the Hsc70-3. This disrupts nitration of midgut epithelial cells invaded by the parasite and the release of hemocyte-derived microvesicles, which are critical for effective activation of the mosquito complement system that eliminates the parasite.


Subject(s)
Anopheles , Malaria , Plasmodium , Animals , Humans , Plasmodium falciparum , Anopheles/parasitology , Heat-Shock Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 120(5): e2213626120, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36689648

ABSTRACT

Plasmodium falciparum malaria originated when Plasmodium praefalciparum, a gorilla malaria parasite transmitted by African sylvan anopheline mosquitoes, adapted to humans. Pfs47, a protein on the parasite surface mediates P. falciparum evasion of the mosquito immune system by interacting with a midgut receptor and is critical for Plasmodium adaptation to different anopheline species. Genetic analysis of 4,971 Pfs47 gene sequences from different continents revealed that Asia and Papua New Guinea harbor Pfs47 haplotypes more similar to its ortholog in P. praefalciparum at sites that determine vector compatibility, suggesting that ancestral P. falciparum readily adapted to Asian vectors. Consistent with this observation, Pfs47-receptor gene sequences from African sylvan malaria vectors, such as Anopheles moucheti and An. marshallii, were found to share greater similarity with those of Asian vectors than those of vectors of the African An. gambiae complex. Furthermore, experimental infections provide direct evidence that transformed P. falciparum parasites carrying Pfs47 orthologs of P. praefalciparum or P. reichenowi were more effective at evading the immune system of the Asian malaria vector An. dirus than An. gambiae. We propose that high compatibility of ancestral P. falciparum Pfs47 with the receptors of Asian vectors facilitated the early dispersal of human malaria to the Asian continent, without having to first adapt to sub-Saharan vectors of the An. gambiae complex.


Subject(s)
Anopheles , Malaria, Falciparum , Malaria , Plasmodium , Animals , Humans , Plasmodium falciparum/genetics , Anopheles/genetics , Mosquito Vectors/parasitology , Malaria, Falciparum/parasitology , Gorilla gorilla
4.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Article in English | MEDLINE | ID: mdl-34711682

ABSTRACT

Immune priming in Anopheles gambiae is mediated by the systemic release of a hemocyte differentiation factor (HDF), a complex of lipoxin A4 bound to Evokin, a lipid carrier. HDF increases the proportion of circulating granulocytes and enhances mosquito cellular immunity. Here, we show that Evokin is present in hemocytes and fat-body cells, and messenger RNA (mRNA) expression increases significantly after immune priming. The double peroxidase (DBLOX) enzyme, present in insects but not in vertebrates, is essential for HDF synthesis. DBLOX is highly expressed in oenocytes in the fat-body tissue, and these cells increase in number in primed mosquitoes. We provide direct evidence that the histone acetyltransferase AgTip60 (AGAP001539) is also essential for a sustained increase in oenocyte numbers, HDF synthesis, and immune priming. We propose that oenocytes may function as a population of cells that are reprogrammed, and orchestrate and maintain a broad, systemic, and long-lasting state of enhanced immune surveillance in primed mosquitoes.


Subject(s)
Culicidae/immunology , Histone Acetyltransferases/metabolism , Immunologic Memory/immunology , Animals , Anopheles/immunology , Anopheles/metabolism , Culicidae/metabolism , Female , Granulocytes/metabolism , Hemocytes/immunology , Immunity, Innate/immunology , Insect Proteins/genetics , Insecta/metabolism , Lipoxins/metabolism , Malaria/immunology , Male , Peroxidase/metabolism , Plasmodium/metabolism , Plasmodium berghei/metabolism
5.
Trends Parasitol ; 36(11): 880-883, 2020 11.
Article in English | MEDLINE | ID: mdl-33036937

ABSTRACT

Malaria eradication is a global priority but requires innovative strategies. Humoral immune responses attack different parasite stages, and antibody-based therapy may prevent malaria infection or transmission. Here, we discuss targets of monoclonal antibodies in mosquito sexual stages of Plasmodium.


Subject(s)
Antibodies, Monoclonal/immunology , Culicidae/parasitology , Life Cycle Stages/immunology , Malaria/prevention & control , Malaria/transmission , Plasmodium falciparum/immunology , Animals , Culicidae/immunology , Disease Eradication , Humans , Malaria/parasitology
6.
Front Microbiol ; 11: 1496, 2020.
Article in English | MEDLINE | ID: mdl-32719666

ABSTRACT

An effective vaccine to reduce malaria transmission is central to control and ultimately achieve disease eradication. Recently, we demonstrated that antibodies targeting the Plasmodium falciparum surface protein P47 (Pfs47) reduce parasite transmission to Anopheles gambiae mosquitoes. Here, Plasmodium berghei (Pb) was used as a model to assess the in vivo efficacy of a P47-targeted transmission blocking vaccine (Pbs47). Mice were immunized following a prime/boost regimen and infected with P. berghei. The effect of immunization on infectivity to mosquitoes was evaluated by direct feeding on P. berghei-infected mice. The key region in Pbs47 where antibody binding confers protection was mapped, and the immunogenicity of this protective antigen was enhanced by conjugation to a virus-like particle. Passive immunization with 100 and 50 µg/mL of anti-Pbs47 IgG reduced oocyst density by 77 and 67%, respectively. Furthermore, affinity purified Pbs47-specific IgG significantly reduced oocyst density by 88 and 77%, respectively at doses as low as 10 and 1 µg/mL. These studies suggest that P47 is a promising transmission blocking target and show that antibodies to the same specific region in Pfs47 and Pbs47 confer protection.

7.
Proc Natl Acad Sci U S A ; 117(5): 2597-2605, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31969456

ABSTRACT

The surface protein Pfs47 allows Plasmodium falciparum parasites to survive and be transmitted by making them "undetectable" to the mosquito immune system. P. falciparum parasites express Pfs47 haplotypes compatible with their sympatric vectors, while those with incompatible haplotypes are eliminated by the mosquito. We proposed that Pfs47 serves as a "key" that mediates immune evasion by interacting with a mosquito receptor "the lock," which differs in evolutionarily divergent anopheline mosquitoes. Recombinant Pfs47 (rPfs47) was used to identify the mosquito Pfs47 receptor protein (P47Rec) using far-Western analysis. rPfs47 bound to a single 31-kDa band and the identity of this protein was determined by mass spectrometry. The mosquito P47Rec has two natterin-like domains and binds to Pfs47 with high affinity (17 to 32 nM). P47Rec is a highly conserved protein with submicrovillar localization in midgut cells. It has structural homology to a cytoskeleton-interacting protein and accumulates at the site of ookinete invasion. Silencing P47Rec expression reduced P. falciparum infection, indicating that the interaction of Pfs47 with the receptor is critical for parasite survival. The binding specificity of P47Rec from distant anophelines (Anopheles gambiae, Anopheles dirus, and Anopheles albimanus) with Pfs47-Africa (GB4) and Pfs47-South America (7G8) haplotypes was evaluated, and it is in agreement with the previously documented compatibility between P. falciparum parasites expressing different Pfs47 haplotypes and these three anopheline species. Our findings give further support to the role of Pfs47 in the adaptation of P. falciparum to different vectors.


Subject(s)
Anopheles/immunology , Anopheles/parasitology , Insect Proteins/immunology , Membrane Glycoproteins/immunology , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Anopheles/genetics , Host-Parasite Interactions , Immune Evasion , Insect Proteins/genetics , Kinetics , Membrane Glycoproteins/genetics , Mosquito Vectors/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics
8.
Sci Rep ; 9(1): 16833, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727945

ABSTRACT

We recently characterized Pfs47, a protein expressed on the surface of sexual stages and ookinetes of Plasmodium falciparum, as a malaria transmission-blocking vaccine (TBV) target. Mice immunization induced antibodies that conferred strong transmission-reducing activity (TRA) at a concentration of 200 µg/mL. Here, we sought to optimize the Pfs47 vaccine to elicit higher titers of high-affinity antibodies, capable of inducing strong TRA at a lower concentration. We report the development and evaluation of a Pfs47-based virus-like particle (VLP) vaccine generated by conjugating our 58 amino acid Pfs47 antigen to Acinetobacter phage AP205-VLP using the SpyCatcher:SpyTag adaptor system. AP205-Pfs47 complexes (VLP-P47) formed particles of ~22 nm diameter that reacted with polyclonal anti-Pfs47 antibodies, indicating that the antigen was accessible on the surface of the particle. Mice immunized with VLP-P47 followed by a boost with Pfs47 monomer induced significantly higher antibody titers, with higher binding affinity to Pfs47, than mice that received two immunizations with either VLP-P47 (VLP-P47/VLP-P47) or the Pfs47 monomer (P47/P47). Purified IgG from VLP-P47/P47 mice had strong TRA (83-98%) at concentrations as low as 5 µg/mL. These results indicate that conjugating the Pfs47 antigen to AP205-VLP significantly enhanced antigenicity and confirm the potential of Pfs47 as a TBV candidate.


Subject(s)
Antibodies, Protozoan/metabolism , Malaria, Falciparum/prevention & control , Membrane Glycoproteins/immunology , Protozoan Proteins/immunology , Vaccines, Virus-Like Particle/administration & dosage , Animals , Bacteriophages/genetics , Bacteriophages/immunology , Female , Immunization, Secondary , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Male , Mice , Vaccines, Virus-Like Particle/immunology
9.
Methods Mol Biol ; 1955: 119-134, 2019.
Article in English | MEDLINE | ID: mdl-30868523

ABSTRACT

The surface coat of Trypanosoma cruzi is covered with glycosylphosphatidylinositol (GPI)-anchored glycoproteins (GAGPs) that contribute to parasite protection and to the establishment of a persistent infection in both the insect vector and the mammalian host. Multiple GAGPs that vary by amino acid sequence and/or posttranslational modifications are co-expressed on the parasite surface coat, hence curtailing structural/functional analyses on these molecules. Studies in our lab have indicated that GAGP-tagged variants expressed by transfected parasites undergo analogous posttranslational processing than endogenous ones and therefore constitute suitable tools to overcome these limitations. In this chapter, we detail the entire methodological pipeline for the efficient homologous expression of GAGPs in T. cruzi: from a simple strategy for the simultaneously cloning and tagging of the gene of interest to the biochemical validation of the parasite-expressed product.


Subject(s)
GPI-Linked Proteins/genetics , Protozoan Proteins/genetics , Trypanosoma cruzi/genetics , Chagas Disease/parasitology , Cloning, Molecular/methods , Gene Expression , Humans , Recombinant Proteins/genetics , Transfection/methods
10.
Methods Mol Biol ; 1955: 135-146, 2019.
Article in English | MEDLINE | ID: mdl-30868524

ABSTRACT

Trypanosoma cruzi, the protozoan agent of Chagas disease, has evolved an innovative metabolic pathway by which protective sialic acid (SA) residues are scavenged from host sialylglycoconjugates and transferred onto parasite surface mucin-like molecules (or surface glycoconjugates from host target cells) by means of a unique trans-sialidase (TS) enzyme. TS-induced changes in the glycoprotein sialylation profile of both parasite and host cells are crucial for the establishment of a persistent T. cruzi infection and for the development of Chagas disease-associated pathogenesis. In this chapter, we describe a novel metabolic labeling method developed in our labs that enables straightforward identification and molecular characterization of SA acceptors of the TS-catalyzed reaction.


Subject(s)
Glycoproteins/metabolism , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Protozoan Proteins/metabolism , Trypanosoma cruzi/physiology , Animals , Blotting, Western/methods , Chagas Disease/metabolism , Chagas Disease/parasitology , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Host-Parasite Interactions , Humans , Metabolic Networks and Pathways , Staining and Labeling/methods , Trypanosoma cruzi/enzymology
11.
PLoS Negl Trop Dis ; 13(3): e0007245, 2019 03.
Article in English | MEDLINE | ID: mdl-30870417

ABSTRACT

BACKGROUND: TolT was originally described as a Trypanosoma cruzi molecule that accumulated on the trypomastigote flagellum bearing similarity to bacterial TolA colicins receptors. Preliminary biochemical studies indicated that TolT resolved in SDS-PAGE as ~3-5 different bands with sizes between 34 and 45 kDa, and that this heterogeneity could be ascribed to differences in polypeptide glycosylation. However, the recurrent identification of TolT-deduced peptides, and variations thereof, in trypomastigote proteomic surveys suggested an intrinsic TolT complexity, and prompted us to undertake a thorough reassessment of this antigen. METHODS/PRINCIPLE FINDINGS: Genome mining exercises showed that TolT constitutes a larger-than-expected family of genes, with at least 12 polymorphic members in the T. cruzi CL Brener reference strain and homologs in different trypanosomes. According to structural features, TolT deduced proteins could be split into three robust groups, termed TolT-A, TolT-B, and TolT-C, all of them showing marginal sequence similarity to bacterial TolA proteins and canonical signatures of surface localization/membrane association, most of which were herein experimentally validated. Further biochemical and microscopy-based characterizations indicated that this grouping may have a functional correlate, as TolT-A, TolT-B and TolT-C molecules showed differences in their expression profile, sub-cellular distribution, post-translational modification(s) and antigenic structure. We finally used a recently developed fluorescence magnetic beads immunoassay to validate a recombinant protein spanning the central and mature region of a TolT-B deduced molecule for Chagas disease serodiagnosis. CONCLUSION/SIGNIFICANCE: This study unveiled an unexpected genetic and biochemical complexity within the TolT family, which could be exploited for the development of novel T. cruzi biomarkers with diagnostic/therapeutic applications.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Polymorphism, Genetic , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Computational Biology , Glycosylation , Immunoassay , Membrane Proteins/classification , Protozoan Proteins/classification
12.
Educ Res Int ; 20192019.
Article in English | MEDLINE | ID: mdl-32071792

ABSTRACT

Successful researchers in the biological sciences communicate their work to a global audience and must do so in English to be widely recognized and cited. This applies equally to scientific talks, posters, and published articles; thus, scientific English must be prioritized in nonnative English-speaking (NNES) academic institutions to prepare their trainees for successful careers. Here, we propose strategies for integrating scientific English into PhD programs operating in NNES countries. Many graduate students from NNES countries strive for an international career and encounter English as an important barrier. Based on our own experiences as NNES postdoctoral fellows at a US institution, or as a US mentor of these trainees, we contend that conventional learning processes at home institutions do not sufficiently prioritize scientific English as the medium for regular discussions of laboratory-generated data. Principal investigators, mentors, and supervisors are key in promoting English language usage as a structured component of PhD training. If these stakeholders routinely integrate English training and education within the research laboratory program, graduates will be equipped to pursue international academic careers. The ideas presented here are intended for NNES PhD students (and their mentors) who seek an international scientific career in the biological sciences.

13.
NPJ Vaccines ; 3: 26, 2018.
Article in English | MEDLINE | ID: mdl-30002917

ABSTRACT

Transmission-blocking vaccines are based on eliciting antibody responses in the vertebrate host that disrupt parasite development in the mosquito vector and prevent malaria transmission. The surface protein Pfs47 is present in Plasmodium falciparum gametocytes and female gametes. The potential of Pfs47 as a vaccine target was evaluated. Soluble full-length recombinant protein, consisting of three domains, was expressed in E. coli as a thioredoxin fusion (T-Pfs47). The protein was immunogenic, and polyclonal and monoclonal antibodies (mAb) were obtained, but they did not confer transmission blocking activity (TBA). All fourteen mAb targeted either domains 1 or 3, but not domain 2 (D2), and immune reactivity to D2 was also very low in polyclonal mouse IgG after T-Pfs47 immunization. Disruption of the predicted disulfide bond in D2, by replacing cysteines for alanines (C230A and C260A), allowed expression of recombinant D2 protein in E. coli. A combination of mAbs targeting D2, and deletion proteins from this domain, allowed us to map a central 52 amino acid (aa) region where antibody binding confers strong TBA (78-99%). This 52 aa antigen is immunogenic and well conserved, with only seven haplotypes world-wide that share 96-98% identity. Neither human complement nor the mosquito complement-like system are required for the observed TBA. A dramatic reduction in ookinete numbers and ookinete-specific transcripts was observed, suggesting that the antibodies are interacting with female gametocytes and preventing fertilization.

14.
Curr Opin Microbiol ; 40: 168-174, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29229188

ABSTRACT

Malaria is caused by infection with Plasmodium parasites that have a complex life cycle. The parasite protein P47 is critical for disease transmission. P47 mediates mosquito immune evasion in both Plasmodium berghei (Pbs47) and Plasmodium falciparum (Pfs47), and has been shown to be important for optimal female gamete fertility in P. berghei. Pfs47 presents strong geographic structure in natural P. falciparum populations, consistent with natural selection of Pfs47 haplotypes by the mosquito immune system as the parasite adapted to new vector species worldwide. These key functions make Plasmodium P47 an attractive target to disrupt malaria transmission.


Subject(s)
Malaria, Falciparum/transmission , Mosquito Vectors/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Female , Humans , Malaria, Falciparum/parasitology , Mosquito Vectors/physiology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Reproduction
15.
Proc Natl Acad Sci U S A ; 114(47): 12566-12571, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29114059

ABSTRACT

A naturally occurring Wolbachia strain (wAnga-Mali) was identified in mosquitoes of the Anopheles gambiae complex collected in the Malian villages of Dangassa and Kenieroba. Phylogenetic analysis of the nucleotide sequence of two 16S rRNA regions showed that wAnga-Mali clusters with Wolbachia strains from supergroup A and has the highest homology to a Wolbachia strain isolated from cat fleas (Ctenocephalides). wAnga-Mali is different from two Wolbachia strains previously reported in A. gambiae from Burkina Faso (wAnga_VK5_STP and wAnga_VK5_3.1a). Quantitative analysis of Wolbachia and Plasmodium sporozoite infection in field-collected mosquitoes indicates that the prevalence and intensity of Plasmodium falciparum sporozoite infection is significantly lower in Wolbachia-infected females. The presence of Wolbachia in females from a laboratory Anopheles coluzzii (A. gambiae, M form) colony experimentally infected with P. falciparum (NF54 strain) gametocyte cultures slightly enhanced oocyst infection. However, Wolbachia infection significantly reduced the prevalence and intensity of sporozoite infection, as observed in the field. This indicates that wAnga-Mali infection does not limit early stages of Plasmodium infection in the mosquito, but it has a strong deleterious effect on sporozoites and reduces malaria transmission.


Subject(s)
Anopheles/microbiology , Host-Parasite Interactions , Insect Vectors/microbiology , Malaria, Falciparum/transmission , Plasmodium falciparum/microbiology , Wolbachia/genetics , Animals , Anopheles/parasitology , Female , Host-Pathogen Interactions , Insect Vectors/parasitology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Mali/epidemiology , Oocysts/pathogenicity , Oocysts/physiology , Phylogeny , RNA, Ribosomal, 16S/genetics , Severity of Illness Index , Sporozoites/pathogenicity , Sporozoites/physiology , Wolbachia/classification , Wolbachia/isolation & purification
16.
PLoS Negl Trop Dis ; 11(8): e0005856, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28800609

ABSTRACT

BACKGROUND: TSSA (Trypomastigote Small Surface Antigen) is an antigenic, adhesion molecule displayed on the surface of Trypanosoma cruzi trypomastigotes. TSSA displays substantial sequence identity to members of the TcMUC gene family, which code for the trypomastigote mucins (tGPI-mucins). In addition, TSSA bears sequence polymorphisms among parasite strains; and two TSSA variants expressed as recombinant molecules (termed TSSA-CL and TSSA-Sy) were shown to exhibit contrasting features in their host cell binding and signaling properties. METHODS/PRINCIPLE FINDINGS: Here we used a variety of approaches to get insights into TSSA structure/function. We show that at variance with tGPI-mucins, which rely on their extensive O-glycoslylation to achieve their protective function, TSSA seems to be displayed on the trypomastigote coat as a hypo-glycosylated molecule. This has a functional correlate, as further deletion mapping experiments and cell binding assays indicated that exposition of at least two peptidic motifs is critical for the engagement of the 'adhesive' TSSA variant (TSSA-CL) with host cell surface receptor(s) prior to trypomastigote internalization. These motifs are not conserved in the 'non-adhesive' TSSA-Sy variant. We next developed transgenic lines over-expressing either TSSA variant in different parasite backgrounds. In strict accordance to recombinant protein binding data, trypomastigotes over-expressing TSSA-CL displayed improved adhesion and infectivity towards non-macrophagic cell lines as compared to those over-expressing TSSA-Sy or parental lines. These phenotypes could be specifically counteracted by exogenous addition of peptides spanning the TSSA-CL adhesion motifs. In addition, and irrespective of the TSSA variant, over-expression of this molecule leads to an enhanced trypomastigote-to-amastigote conversion, indicating a possible role of TSSA also in parasite differentiation. CONCLUSION/SIGNIFICANCE: In this study we provided novel evidence indicating that TSSA plays an important role not only on the infectivity and differentiation of T. cruzi trypomastigotes but also on the phenotypic variability displayed by parasite strains.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Surface/chemistry , Mucins/metabolism , Trypanosoma cruzi/pathogenicity , Amino Acid Sequence , Animals , Antigens, Protozoan/genetics , Antigens, Surface/genetics , Cell Differentiation , Chagas Disease/parasitology , Chlorocebus aethiops , Gene Expression Regulation , Genes, Protozoan , HeLa Cells , Humans , Recombinant Proteins/chemistry , Trypanosoma cruzi/genetics , Vero Cells
17.
PLoS One ; 11(12): e0168279, 2016.
Article in English | MEDLINE | ID: mdl-27992481

ABSTRACT

Malaria is a life-threatening disease caused by Plasmodium falciparum parasites that is transmitted through the bites of infected anopheline mosquitoes. P. falciparum dispersal from Africa, as a result of human migration, required adaptation of the parasite to several different indigenous anopheline species. The mosquito immune system can greatly limit infection and P. falciparum evolved a strategy to evade these responses that is mediated by the Pfs47 gene. Pfs47 is a polymorphic gene with signatures of diversifying selection and a strong geographic genetic structure at a continental level. Here, we investigated the role of single four amino acid differences between the Pfs47 gene from African (GB4 and NF54) and a New World (7G8) strains that differ drastically in their ability to evade the immune system of A. gambiae L35 refractory mosquitoes. Wild type NF54 and GB4 parasites can survive in this mosquito strain, while 7G8 parasites are eliminated. Our studies indicate that replacement in any of these four single amino acids in Pfs47 from the NF54 strain by those present in 7G8, completely disrupts the ability of NF54 parasites to hide from the mosquito immune system. One of these amino acid replacements had the opposite effect on A. albimanus mosquitoes, and enhanced infection. We conclude that malaria transmission involves a complex interplay between the genetic background of the parasite and the mosquito and that Pfs47 can be critical in this interaction as it mediates Plasmodium immune evasion through molecular interactions that need to be precise in some parasite/vector combinations.


Subject(s)
Anopheles/immunology , Malaria, Falciparum/parasitology , Membrane Glycoproteins/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Anopheles/parasitology , Genotype , Host-Parasite Interactions , Humans , Immune Evasion , Malaria, Falciparum/immunology , Mutation , Plasmodium falciparum/classification , Plasmodium falciparum/genetics
18.
Proc Natl Acad Sci U S A ; 112(49): 15178-83, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26598665

ABSTRACT

Plasmodium falciparum malaria originated in Africa and became global as humans migrated to other continents. During this journey, parasites encountered new mosquito species, some of them evolutionarily distant from African vectors. We have previously shown that the Pfs47 protein allows the parasite to evade the mosquito immune system of Anopheles gambiae mosquitoes. Here, we investigated the role of Pfs47-mediated immune evasion in the adaptation of P. falciparum to evolutionarily distant mosquito species. We found that P. falciparum isolates from Africa, Asia, or the Americas have low compatibility to malaria vectors from a different continent, an effect that is mediated by the mosquito immune system. We identified 42 different haplotypes of Pfs47 that have a strong geographic population structure and much lower haplotype diversity outside Africa. Replacement of the Pfs47 haplotypes in a P. falciparum isolate is sufficient to make it compatible to a different mosquito species. Those parasites that express a Pfs47 haplotype compatible with a given vector evade antiplasmodial immunity and survive. We propose that Pfs47-mediated immune evasion has been critical for the globalization of P. falciparum malaria as parasites adapted to new vector species. Our findings predict that this ongoing selective force by the mosquito immune system could influence the dispersal of Plasmodium genetic traits and point to Pfs47 as a potential target to block malaria transmission. A new model, the "lock-and-key theory" of P. falciparum globalization, is proposed, and its implications are discussed.


Subject(s)
Anopheles/immunology , Immune Evasion , Malaria, Falciparum/transmission , Plasmodium falciparum/physiology , Animals , Anopheles/parasitology , Insect Vectors , Molecular Sequence Data
19.
Proc Natl Acad Sci U S A ; 112(5): 1273-80, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25552553

ABSTRACT

The malaria parasite, Plasmodium, must survive and develop in the mosquito vector to be successfully transmitted to a new host. The Plasmodium falciparum Pfs47 gene is critical for malaria transmission. Parasites that express Pfs47 (NF54 WT) evade mosquito immunity and survive, whereas Pfs47 knockouts (KO) are efficiently eliminated by the complement-like system. Two alternative approaches were used to investigate the mechanism of action of Pfs47 on immune evasion. First, we examined whether Pfs47 affected signal transduction pathways mediating mosquito immune responses, and show that the Jun-N-terminal kinase (JNK) pathway is a key mediator of Anopheles gambiae antiplasmodial responses to P. falciparum infection and that Pfs47 disrupts JNK signaling. Second, we used microarrays to compare the global transcriptional responses of A. gambiae midguts to infection with WT and KO parasites. The presence of Pfs47 results in broad and profound changes in gene expression in response to infection that are already evident 12 h postfeeding, but become most prominent at 26 h postfeeding, the time when ookinetes invade the mosquito midgut. Silencing of 15 differentially expressed candidate genes identified caspase-S2 as a key effector of Plasmodium elimination in parasites lacking Pfs47. We provide experimental evidence that JNK pathway regulates activation of caspases in Plasmodium-invaded midgut cells, and that caspase activation is required to trigger midgut epithelial nitration. Pfs47 alters the cell death pathway of invaded midgut cells by disrupting JNK signaling and prevents the activation of several caspases, resulting in an ineffective nitration response that makes the parasite undetectable by the mosquito complement-like system.


Subject(s)
Anopheles/immunology , Apoptosis/physiology , MAP Kinase Kinase 4/metabolism , Plasmodium falciparum/physiology , Animals , Anopheles/parasitology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Protozoan Proteins/physiology
20.
Clin Vaccine Immunol ; 22(3): 304-12, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589551

ABSTRACT

The trypomastigote small surface antigen (TSSA) is a mucin-like molecule from Trypanosoma cruzi, the etiological agent of Chagas disease, which displays amino acid polymorphisms in parasite isolates. TSSA expression is restricted to the surface of infective cell-derived trypomastigotes, where it functions as an adhesin and engages surface receptors on the host cell as a prerequisite for parasite internalization. Previous results have established TSSA-CL, the isoform encoded by the CL Brener clone, as an appealing candidate for use in serology-based diagnostics for Chagas disease. Here, we used a combination of peptide- and recombinant protein-based tools to map the antigenic structure of TSSA-CL at maximal resolution. Our results indicate the presence of different partially overlapping B-cell epitopes clustering in the central portion of TSSA-CL, which contains most of the polymorphisms found in parasite isolates. Based on these results, we assessed the serodiagnostic performance of a 21-amino-acid-long peptide that spans TSSA-CL major antigenic determinants, which was similar to the performance of the previously validated glutathione S-transferase (GST)-TSSA-CL fusion molecule. Furthermore, the tools developed for the antigenic characterization of the TSSA antigen were also used to explore other potential diagnostic applications of the anti-TSSA humoral response in Chagasic patients. Overall, our present results provide additional insights into the antigenic structure of TSSA-CL and support this molecule as an excellent target for molecular intervention in Chagas disease.


Subject(s)
Antibodies, Protozoan/blood , Chagas Disease/diagnosis , Trypanosoma cruzi/chemistry , Trypanosoma cruzi/immunology , Variant Surface Glycoproteins, Trypanosoma/chemistry , Variant Surface Glycoproteins, Trypanosoma/immunology , Amino Acid Sequence , Chagas Disease/congenital , Chagas Disease/immunology , Epitope Mapping , Epitopes, B-Lymphocyte , Glutathione Transferase , Humans , Peptide Mapping , Peptides/immunology , Protein Array Analysis , Protein Structure, Tertiary , ROC Curve , Recombinant Fusion Proteins/immunology , Variant Surface Glycoproteins, Trypanosoma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...