Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Malar J ; 17(1): 304, 2018 Aug 20.
Article in English | MEDLINE | ID: mdl-30126436

ABSTRACT

BACKGROUND: Plasmodium enolase is a target for the growth neutralizing antibodies. Interestingly, the three invasive stages i.e. sporozoites, merozoites, and ookinetes express this protein on their cell surface. Polyclonal anti-Plasmodium falciparum enolase (Pfeno) antibodies disrupt traversal of ookinete through mosquito mid-gut wall as well as have inhibitory effect on parasite growth at erythrocytic stage. In a recent study, it was observed that immunization with a unique epitope of parasite enolase (EWGWS) could confer partial protection against mouse malaria. Further validation is needed for the protective potential of this unique epitope in otherwise highly conserved enolase. METHODS: In order to investigate the efficacy of growth inhibitory potential of the epitope of P falciparum enolase, a monoclonal antibody specific to EWGWS is generated. In vitro parasite growth inhibition assays and passive immunization of Plasmodium yoelii (or Plasmodium berghei) infected mice were used to assess the parasite growth neutralizing activity of the antibody. RESULTS: Screening a panel of monoclonal antibodies raised against recombinant Pfeno that were specific to EWGWS resulted in isolation of H12E1. This antibody recognized only EWGWS epitope containing enolases. H12E1 strongly inhibited parasite growth in culture. This inhibition was strain transcending. Passive infusion of this antibody in P. yoelii or P. berghei infected mice showed significant reduction in parasitemia as compared to controls (p < 0.001). Surface Plasmon Resonance measurements indicated high affinity binding of H12E1 to P. falciparum enolase (KD ~ 7.6 × 10-9M). CONCLUSIONS: A monoclonal antibody directed against EWGWS epitope of Pfeno was shown to inhibit the growth of blood stage malarial parasites. This inhibition was species/strain transcending and is likely to arise due to blockade of enolase on the surface of merozoites, functionally implicating Pfeno in invasion related events. Presence of enolase on the cell surface of merozoites and ookinetes could potentially result in inhibition of host cell invasions at erythrocytic and transmission stages in the parasite life cycle. It is suggested that antibodies against EWGWS epitope have the potential to confer dual stage, species and strain transcending protection against malaria.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Malaria/prevention & control , Phosphopyruvate Hydratase/immunology , Plasmodium falciparum/enzymology , Plasmodium falciparum/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Protozoan/administration & dosage , Disease Models, Animal , Immunization, Passive , Malaria/immunology , Male , Mice , Plasmodium berghei/immunology , Plasmodium yoelii/immunology
2.
PLoS Pathog ; 11(6): e1004942, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26110623

ABSTRACT

African trypanosomiasis is a deadly neglected disease caused by the extracellular parasite Trypanosoma brucei. Current therapies are characterized by high drug toxicity and increasing drug resistance mainly associated with loss-of-function mutations in the transporters involved in drug import. The introduction of new antiparasitic drugs into therapeutic use is a slow and expensive process. In contrast, specific targeting of existing drugs could represent a more rapid and cost-effective approach for neglected disease treatment, impacting through reduced systemic toxicity and circumventing resistance acquired through impaired compound uptake. We have generated nanoparticles of chitosan loaded with the trypanocidal drug pentamidine and coated by a single domain nanobody that specifically targets the surface of African trypanosomes. Once loaded into this nanocarrier, pentamidine enters trypanosomes through endocytosis instead of via classical cell surface transporters. The curative dose of pentamidine-loaded nanobody-chitosan nanoparticles was 100-fold lower than pentamidine alone in a murine model of acute African trypanosomiasis. Crucially, this new formulation displayed undiminished in vitro and in vivo activity against a trypanosome cell line resistant to pentamidine as a result of mutations in the surface transporter aquaglyceroporin 2. We conclude that this new drug delivery system increases drug efficacy and has the ability to overcome resistance to some anti-protozoal drugs.


Subject(s)
Drug Resistance/drug effects , Molecular Targeted Therapy/methods , Pentamidine/administration & dosage , Trypanocidal Agents/administration & dosage , Trypanosomiasis, African/drug therapy , Animals , Antibodies, Protozoan/administration & dosage , Chitosan/administration & dosage , Chitosan/pharmacokinetics , Disease Models, Animal , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Electrophoretic Mobility Shift Assay , Female , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Nanoparticles/therapeutic use , Pentamidine/pharmacokinetics , Real-Time Polymerase Chain Reaction , Trypanocidal Agents/pharmacokinetics
3.
Poult Sci ; 95(2): 439-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26772659

ABSTRACT

Eimeria spp. must be controlled in floor-reared poultry to prevent the onset of coccidiosis. Here we use an oral antibody to chicken IL-10 to prevent growth depression due to Eimeria spp. infection. Egg antibody directed against an antigenic peptide of IL-10 was produced in laying hens and measured using an ELISA. In the first experiment, egg yolk powder containing antibody to chicken IL-10 (vlpramqt conjugate) (anti-IL-10 yolk powder) was fed at 3.4 g/kg feed to determine growth response following mixed Eimeria spp. challenge. Chicks were fed either anti-IL-10 antibodies or control antibodies and challenged (d3) with either sterile saline or a 10× attenuated Eimeria spp. vaccine. Control-fed and Eimeria-challenged chicks grew 8.8% slower than those challenged with saline (P < 0.04), whereas anti-IL-10-fed Eimeria challenged chicks were not different from untreated controls. In the second trial a dose response was performed with doses of either 0 (control antibody), 0.34-, or 3.4-g anti-IL-10 yolk powder/kg feed. Control-fed, Eimeria-challenged chicks grew 10.6% slower than control saline-challenged chicks (P < 0.05); however, anti-IL-10-fed chicks fed either dose of anti-IL-10 were not different from saline-challenged chicks. Finally, the effect of anti-IL-10 on acquired immunity was investigated. Chicks were fed control or anti-IL-10 yolk powder and vaccinated with a 1× dose of Eimeria vaccine at d 3. After 14 d, antibody was removed from the diet. Chicks were either saline or 10× Eimeria challenged at d 17. We found that the anti-IL-10-fed chickens did not show a reduction in growth due to challenge; hence anti-IL-10 does not appear to affect adaptive immunity during the primary immunization. Overall, use of an antibody to IL-10 is a novel method in preventing adverse effects of Eimeria spp. infection in poultry.


Subject(s)
Antibodies, Protozoan/pharmacology , Avian Proteins/metabolism , Chickens , Coccidiosis/veterinary , Interleukin-10/metabolism , Poultry Diseases/prevention & control , Animal Feed/analysis , Animals , Antibodies, Protozoan/administration & dosage , Chickens/growth & development , Coccidiosis/parasitology , Coccidiosis/prevention & control , Diet/veterinary , Eimeria/physiology , Female , Poultry Diseases/parasitology
4.
Vet Res ; 45: 25, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24571471

ABSTRACT

Although IL17A is associated with the immunological control of various infectious diseases, its role in host response to Eimeria infections is not well understood. In an effort to better dissect the role of IL17A in host-pathogen interactions in avian coccidiosis, a neutralizing antibody (Ab) to chicken IL17A was used to counteract IL17A bioactivity in vivo. Chickens infected with Eimeria tenella and treated intravenously with IL17A Ab, exhibited reduced intracellular schizont and merozoite development, diminished lesion score, compared with untreated controls. Immunohistological evaluation of cecal lesions in the parasitized tissues indicated reduced migration and maturation of second-generation schizonts and reduced lesions in lamina propria and submucosa. In contrast, untreated and infected chickens had epithelial cells harboring second-generation schizonts, which extend into the submucosa through muscularis mucosa disruptions, maturing into second generation merozoites. Furthermore, IL17A Ab treatment was associated with increased parameters of Th1 immunity (IL2- and IFNγ- producing cells), reduced levels of reactive oxygen species (ROS), and diminished levels of serum matrix metalloproteinase-9 (MMP-9). Finally, schizonts from untreated and infected chickens expressed S100, Wiskott-Aldrich syndrome protein family member 3 (WASF3), and heat shock protein-70 (HSP70) proteins as merozoites matured, whereas the expression of these proteins was absent in IL17A Ab-treated chickens. These results provide the first evidence that the administration of an IL17A neutralizing Ab to E. tenella-infected chickens inhibits the migration of parasitized epithelial cells, markedly reduces the production of ROS and MMP-9, and decreases cecal lesions, suggesting that IL17A might be a potential therapeutic target for coccidiosis control.


Subject(s)
Antibodies, Protozoan/pharmacology , Chickens , Coccidiosis/veterinary , Eimeria tenella/physiology , Interleukin-17/administration & dosage , Poultry Diseases/prevention & control , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/pharmacology , Antibodies, Protozoan/administration & dosage , Cecum/drug effects , Cecum/parasitology , Coccidiosis/parasitology , Coccidiosis/prevention & control , Epithelial Cells/drug effects , Epithelial Cells/parasitology , Poultry Diseases/parasitology , Schizonts/drug effects , Schizonts/growth & development , Schizonts/physiology
5.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33332286

ABSTRACT

CIS43 is a potent neutralizing human mAb that targets a highly conserved "junctional" epitope in the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP). Enhancing the durability of CIS43 in vivo will be important for clinical translation. Here, 2 approaches were used to improve the durability of CIS43 in vivo while maintaining potent neutralization. First, the Fc domain was modified with the LS mutations (CIS43LS) to increase CIS43 binding affinity for the neonatal Fc receptor (FcRn). CIS43LS and CIS43 showed comparable in vivo protective efficacy. CIS43LS had 9- to 13-fold increased binding affinity for human (6.2 nM versus 54.2 nM) and rhesus (25.1 nM versus 325.8 nM) FcRn at endosomal pH 6.0 compared with CIS43. Importantly, the half-life of CIS43LS in rhesus macaques increased from 22 days to 39 days compared with CIS43. The second approach for sustaining antibody levels of CIS43 in vivo is through adeno-associated virus (AAV) expression. Mice administered once with AAV-expressing CIS43 had sustained antibody levels of approximately 300 µg/mL and mediated protection against sequential malaria challenges up to 36 weeks. Based on these data, CIS43LS has advanced to phase I clinical trials, and AAV delivery provides a potential next-generation approach for malaria prevention.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Amino Acid Substitution , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/genetics , Antibodies, Protozoan/administration & dosage , Antibodies, Protozoan/blood , Antibodies, Protozoan/genetics , Dependovirus/genetics , Female , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Macaca mulatta , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Protozoan Proteins/immunology
6.
J Exp Med ; 172(6): 1633-41, 1990 Dec 01.
Article in English | MEDLINE | ID: mdl-2258697

ABSTRACT

IgG extracted from the sera of African adults immune to malaria were injected intravenously into eight Plasmodium falciparum-infected nonimmune Thai patients. Clinical and parasitological improvement was reproducibly obtained in each case. After the disappearance of the transferred Ig, recrudescent parasites were equally susceptible to the same Ig preparation. High levels of antibodies to most parasite proteins were detected by Western blots in the receivers' sera (taken before transfer) as in the donors' Ig, thus indicating that the difference was qualitative rather than quantitative between donors and receivers. In vitro, the clinically effective Ig had no detectable inhibitory effect on either penetration or intra-erythrocytic development of the parasite. On the contrary, they sometimes increased parasite growth. In contrast, these IgG, as the receivers' Ig collected 4 d after transfer, but not those collected before transfer, proved able to exert an antibody-dependent cellular inhibitory (ADCI) effect in cooperation with normal blood monocytes. Results were consistent among the seven isolates studied in vitro, as with the recrudescent parasites. Thus, the results obtained in the ADCI assay correlate closely with clinical and parasitological observations.


Subject(s)
Antibodies, Protozoan/immunology , Immunoglobulin G/administration & dosage , Malaria/blood , Monocytes/physiology , Plasmodium falciparum/immunology , Adult , Animals , Antibodies, Protozoan/administration & dosage , Antibodies, Protozoan/isolation & purification , Antibody-Dependent Cell Cytotoxicity , Humans , Immunoglobulin G/isolation & purification , Malaria/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/pathogenicity
7.
J Exp Med ; 192(11): 1653-60, 2000 Dec 04.
Article in English | MEDLINE | ID: mdl-11104807

ABSTRACT

We have recently described that sustained Plasmodium falciparum growth could be obtained in immunodeficient mice. We now report the potential of this new mouse model by assaying the effect of the passive transfer of antibodies (Abs) which in humans have had a well-established effect.Our results show that the total African adult hyperimmune immunoglobulin Gs (HI-IgGs) strongly reduce P. falciparum parasitemia similarly to that reported in humans, but only when mice are concomitantly reconstituted with human monocytes (HuMNs). In contrast, neither HI-IgGs nor HuMNs alone had any direct effect upon parasitemia. We assessed the in vivo effect of epitope-specific human Abs affinity-purified on peptides derived either from the ring erythrocyte surface antigen (RESA) or the merozoite surface protein 3 (MSP3). The inoculation of low concentrations of anti-synthetic peptide from MSP3, but not of anti-RESA Abs, consistently suppressed P. falciparum in the presence of HuMNs. Parasitemia decrease was stronger and faster than that observed using HI-IgGs and as fast as that induced by chloroquine. Our observations demonstrate that this mouse model is of great value to evaluate the protective effect of different Abs with distinct specificity in the same animal, a step hardly accessible and therefore never performed before in humans.


Subject(s)
Antibodies, Protozoan/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adult , Amino Acid Sequence , Animals , Antibodies, Protozoan/administration & dosage , Antigens, Protozoan/immunology , Antigens, Surface/immunology , Disease Models, Animal , Humans , Immunization, Passive , Immunocompromised Host , Malaria, Falciparum/blood , Malaria, Falciparum/prevention & control , Male , Mice , Molecular Sequence Data , Monocytes/immunology , Plasmodium falciparum/growth & development , Protozoan Proteins/immunology
8.
J Exp Med ; 191(6): 1063-8, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10727468

ABSTRACT

We show here that maintenance of Leishmania infections with Leishmania mexicana complex parasites (Leishmania amazonensis and Leishmania pifanoi) is impaired in the absence of circulating antibody. In these studies, we used mice genetically altered to contain no circulating antibody, with and without functional B cells. This experimental design allowed us to rule out a critical role for B cell antigen presentation in Leishmania pathogenesis. In addition, we show that mice lacking the common gamma chain of Fc receptors (FcgammaRI, FcepsilonRI, and FcgammaRIII) are similarly refractory to infection with these parasites. These observations establish a critical role for antibody in the pathogenesis associated with infection by members of the L. mexicana complex.


Subject(s)
Leishmania mexicana/growth & development , Leishmania mexicana/metabolism , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Receptors, Fc/physiology , Animals , Antibodies, Protozoan/administration & dosage , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/blood , Immunization, Passive , Leishmania mexicana/immunology , Leishmaniasis, Cutaneous/etiology , Leishmaniasis, Cutaneous/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Fc/deficiency , Receptors, Fc/genetics , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/physiology
9.
Antimicrob Agents Chemother ; 54(4): 1385-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20086143

ABSTRACT

At present no completely effective treatments are available for Cryptosporidium parvum infections in humans and livestock. Based on previous data showing the neutralizing potential of a panel of monoclonal antibodies developed against C. parvum, and based on the fact that innate immune peptides and enzymes have anticryptosporidial activity, we engineered several of these antibodies into antibody-biocide fusion proteins. We hypothesized that the combination of high-affinity antibody targeting with innate immune molecule-mediated killing would result in a highly effective new antiprotozoal agent. To test this hypothesis, we expressed antibody-biocide fusion proteins in a mammalian cell culture system and used the resulting products for in vitro and in vivo efficacy experiments. Antibody-biocide fusion proteins efficiently bound to, and destroyed, C. parvum sporozoites in vitro through a membrane-disruptive mechanism. When antibody-biocide fusion proteins were administered orally to neonatal mice in a prophylactic model of cryptosporidiosis, the induction of infection was reduced by as much as 81% in the mucosal epithelium of the gut, as determined on the basis of histopathological scoring of infectious stages. Several versions of antibody fusion proteins that differed in antigen specificity and in the biocide used had strong inhibitory effects on the initiation of infection. The results lay the groundwork for the development of a new class of antimicrobials effective against Cryptosporidium.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Protozoan/administration & dosage , Cryptosporidiosis/immunology , Cryptosporidiosis/prevention & control , Cryptosporidium parvum , Animals , Animals, Newborn , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , Antibodies, Protozoan/genetics , Cryptosporidium parvum/immunology , Immunity, Innate , Mice , Mice, Inbred ICR , Molecular Sequence Data , Protein Engineering , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sporozoites/immunology
10.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29914622

ABSTRACT

Anti-malarial pre-erythrocytic vaccines (PEV) target transmission by inhibiting human infection but are currently partially protective. It has been posited, but never demonstrated, that co-administering transmission-blocking vaccines (TBV) would enhance malaria control. We hypothesized a mechanism that TBV could reduce parasite density in the mosquito salivary glands, thereby enhancing PEV efficacy. This was tested using a multigenerational population assay, passaging Plasmodium berghei to Anopheles stephensi mosquitoes. A combined efficacy of 90.8% (86.7-94.2%) was observed in the PEV +TBV antibody group, higher than the estimated efficacy of 83.3% (95% CrI 79.1-87.0%) if the two antibodies acted independently. Higher PEV efficacy at lower mosquito parasite loads was observed, comprising the first direct evidence that co-administering anti-sporozoite and anti-transmission interventions act synergistically, enhancing PEV efficacy across a range of TBV doses and transmission intensities. Combining partially effective vaccines of differing anti-parasitic classes is a pragmatic, powerful way to accelerate malaria elimination efforts.


Subject(s)
Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Protozoan/administration & dosage , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Plasmodium berghei/immunology , Sporozoites/immunology , Animals , Anopheles/parasitology , Drug Synergism , Female , Humans , Malaria/immunology , Malaria/parasitology , Mice , Mosquito Vectors/parasitology , Parasite Load , Plasmodium berghei/drug effects , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Salivary Glands/parasitology , Sporozoites/chemistry , Trophozoites/chemistry , Trophozoites/immunology
11.
Sci Rep ; 8(1): 10511, 2018 Jul 12.
Article in English | MEDLINE | ID: mdl-30002416

ABSTRACT

Plasmodium vivax merozoite invasion is restricted to Duffy positive reticulocytes. Merozoite interaction with the Duffy antigen is mediated by the P. vivax Duffy binding protein (PvDBP). The receptor-binding domain of PvDBP maps to an N-terminal cysteine-rich region referred to as region II (PvDBPII). In addition, a family of P. vivax reticulocyte binding proteins (PvRBPs) mediates interactions with reticulocyte receptors. The receptor binding domain of P. vivax reticulocyte binding protein 1a (PvRBP1a) maps to a 30 kD region (PvRBP1a30). Antibodies raised against recombinant PvRBP1a30 and PvDBPII recognize the native P. vivax antigens and inhibit their binding to host receptors. Rabbit IgG purified from sera raised against PvRBP1a30 and PvDBPII were tested individually and in combination for inhibition of reticulocyte invasion by P. vivax field isolates. While anti-PvDBPII rabbit IgG inhibits invasion, anti-PvRBP1a30 rabbit IgG does not show significant invasion inhibitory activity. Combining antibodies against PvDBPII and PvRBP1a30 also does not increase invasion inhibitory activity. These studies suggest that although PvRBP1a mediates reticulocyte invasion by P. vivax merozoites, it may not be useful to include PvRBP1a30 in a blood stage vaccine for P. vivax malaria. In contrast, these studies validate PvDBPII as a promising blood stage vaccine candidate for P. vivax malaria.


Subject(s)
Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Reticulocytes/parasitology , Animals , Antibodies, Protozoan/administration & dosage , Antibodies, Protozoan/isolation & purification , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Biological Assay/methods , COS Cells , Chlorocebus aethiops , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Malaria Vaccines/administration & dosage , Malaria, Vivax/immunology , Malaria, Vivax/virology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Merozoites/immunology , Merozoites/pathogenicity , Mice , Plasmodium vivax/genetics , Plasmodium vivax/pathogenicity , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Rabbits , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Reticulocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
12.
PLoS One ; 12(12): e0189878, 2017.
Article in English | MEDLINE | ID: mdl-29244862

ABSTRACT

East Coast Fever (ECF) is the most economically important production disease among traditional beef cattle farmers in Zambia. Despite the disease control efforts by the government, donors, and farmers, ECF cases are increasing. Why does ECF oscillate over time? Can alternative approaches such as systems thinking contribute solutions to the complex ECF problem, avoid unintended consequences, and achieve sustainable results? To answer these research questions and inform the design and implementation of ECF interventions, we qualitatively investigated the influence of dynamic socio-economic, cultural, and ecological factors. We used system dynamics modelling to specify these dynamics qualitatively, and an innovative participatory framework called spatial group model building (SGMB). SGMB uses participatory geographical information system (GIS) concepts and techniques to capture the role of spatial phenomenon in the context of complex systems, allowing stakeholders to identify spatial phenomenon directly on physical maps and integrate such information in model development. Our SGMB process convened focus groups of beef value chain stakeholders in two distinct production systems. The focus groups helped to jointly construct a series of interrelated system dynamics models that described ECF in a broader systems context. Thus, a complementary objective of this study was to demonstrate the applicability of system dynamics modelling and SGMB in animal health. The SGMB process revealed policy leverage points in the beef cattle value chain that could be targeted to improve ECF control. For example, policies that develop sustainable and stable cattle markets and improve household income availability may have positive feedback effects on investment in animal health. The results obtained from a SGMB process also demonstrated that a "one-size-fits-all" approach may not be equally effective in policing ECF in different agro-ecological zones due to the complex interactions of socio-ecological context with important, and often ignored, spatial patterns.


Subject(s)
Animal Husbandry , Theileriasis/virology , Animals , Antibodies, Protozoan/administration & dosage , Cattle , Geographic Information Systems , Immunization , Theileriasis/epidemiology , Theileriasis/physiopathology , Zambia
13.
Trans R Soc Trop Med Hyg ; 83(3): 305-7, 1989.
Article in English | MEDLINE | ID: mdl-2694460

ABSTRACT

Five days after receiving a Plasmodium falciparum NF54 infectious blood meal, Anopheles stephensi mosquitoes were fed rat anti-P, falciparum sporozoite or rabbit anti-R32tet32 antibodies. Sporozoites isolated from salivary glands were tested by the inhibition of sporozoite invasion (ISI) assay using monoclonal antibody (Mab) 2A10 to P. falciparum circumsporozoite protein or sera from human volunteers immunized with P. falciparum R32tet32 or (NANP)3-TT vaccines. Whereas sporozoites from control mosquitoes were neutralized by Mab 2A10 and vaccine sera, only the Mab and not the vaccine sera neutralized sporozoites from immune-fed mosquitoes. The implications of these results in vaccine design and the impact on transmission are discussed.


Subject(s)
Anopheles/parasitology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Plasmodium falciparum/immunology , Protozoan Proteins , Animals , Antibodies, Protozoan/administration & dosage , Host-Parasite Interactions , Humans , Immune Sera , Insect Vectors/parasitology , Malaria/immunology , Neutralization Tests , Plasmodium falciparum/growth & development , Rats , Vaccines, Synthetic/immunology
14.
Clin Vaccine Immunol ; 19(1): 100-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22038847

ABSTRACT

Theileria equi immune plasma was infused into young horses (foals) with severe combined immunodeficiency. Although all foals became infected following intravenous challenge with homologous T. equi merozoite stabilate, delayed time to peak parasitemia occurred. Protective effects were associated with a predominance of passively transferred merozoite-specific IgG3.


Subject(s)
Antibodies, Protozoan/administration & dosage , Horse Diseases/prevention & control , Immunization, Passive/methods , Immunologic Factors/administration & dosage , Severe Combined Immunodeficiency/veterinary , Theileriasis/prevention & control , Animals , Antibodies, Protozoan/immunology , Horse Diseases/therapy , Horses , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Immunologic Factors/immunology , Merozoites/immunology , Parasitemia/prevention & control , Theileria/immunology , Time Factors
15.
Br J Pharmacol ; 165(7): 2341-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22013955

ABSTRACT

BACKGROUND AND PURPOSE: Nanobodies are promising antigen-binding moieties for molecular imaging and therapeutic purposes because of their favourable pharmacological and pharmacokinetic properties. However, the capability of monovalent nanobodies to reach targets in the CNS remains to be demonstrated. EXPERIMENTAL APPROACH: We have assessed the blood-brain barrier permeability of Nb_An33, a nanobody against the Trypanosoma brucei brucei variant-specific surface glycoprotein (VSG). This analysis was performed in healthy rats and in rats that were in the encephalitic stage of African trypanosomiasis using intracerebral microdialysis, single photon emission computed tomography (SPECT) or a combination of both methodologies. This enabled the quantification of unlabelled and (99m) Tc-labelled nanobodies using, respectively, a sensitive VSG-based nanobody-detection elisa, radioactivity measurement in collected microdialysates and SPECT image analysis. KEY RESULTS: The combined read-out methodologies showed that Nb_An33 was detected in the brain of healthy rats following i.v. injection, inflammation-induced damage to the blood-brain barrier, as in the late encephalitic stage of trypanosomiasis, significantly increased the efficiency of passage of the nanobody through this barrier. Complementing SPECT analyses with intracerebral microdialysis improved analysis of brain disposition. There is clear value in assessing penetration of the blood-brain barrier by monovalent nanobodies in models of CNS inflammation. Our data also suggest that rapid clearance from blood might hamper efficient targeting of specific nanobodies to the CNS. CONCLUSIONS AND IMPLICATIONS: Nanobodies can enter the brain parenchyma from the systemic circulation, especially in pathological conditions where the blood-brain barrier integrity is compromised.


Subject(s)
Antibodies, Protozoan/administration & dosage , Antibodies, Protozoan/metabolism , Blood-Brain Barrier/immunology , Nanostructures , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/parasitology , Male , Microdialysis/methods , Rats , Rats, Wistar , Technetium Tc 99m Sestamibi/pharmacokinetics , Tomography, Emission-Computed, Single-Photon , Trypanosoma brucei brucei/immunology , Trypanosomiasis, African/diagnostic imaging , Trypanosomiasis, African/immunology , Trypanosomiasis, African/parasitology , X-Ray Microtomography
17.
J Immunol ; 179(6): 4093-100, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785848

ABSTRACT

B and T lymphocyte attenuator (BTLA; CD272) is a coinhibitory receptor that is predominantly expressed on T and B cells and dampens T cell activation. In this study, we analyzed the function of BTLA during infection with Plasmodium berghei ANKA. Infection of C57BL/6 mice with this strain leads to sequestration of leukocytes in brain capillaries that is associated with a pathology resembling cerebral malaria in humans. During the course of infection, we found an induction of BTLA in several organs, which was either due to up-regulation of BTLA expression on T cells in the spleen or due to infiltration of BTLA-expressing T cells into the brain. In the brain, we observed a marked induction of BTLA and its ligand herpesvirus entry mediator during cerebral malaria, which was accompanied by an accumulation of predominantly CD8+ T cells, but also CD4+ T cells. Application of an agonistic anti-BTLA mAb caused a significantly reduced incidence of cerebral malaria compared with control mice. Treatment with this Ab also led to a decreased number of T cells that were sequestered in the brain of P. berghei ANKA-infected mice. Our findings indicate that BTLA-herpesvirus entry mediator interactions are functionally involved in T cell regulation during P. berghei ANKA infection of mice and that BTLA is a potential target for therapeutic interventions in severe malaria.


Subject(s)
Malaria, Cerebral/immunology , Malaria, Cerebral/prevention & control , Receptors, Immunologic/metabolism , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Protozoan/administration & dosage , Brain/blood supply , Brain/immunology , Brain/parasitology , Brain/pathology , Cell Movement/immunology , Cells, Cultured , Female , Ligands , Lymphocyte Activation/immunology , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microcirculation/immunology , Microcirculation/parasitology , Microcirculation/pathology , Plasmodium berghei/growth & development , Plasmodium berghei/immunology , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/immunology , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor, Member 14/deficiency , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Tumor Necrosis Factor, Member 14/physiology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , T-Lymphocytes/pathology
18.
Parasite Immunol ; 11(3): 223-30, 1989 May.
Article in English | MEDLINE | ID: mdl-2771427

ABSTRACT

The inhibitory effect of anti-sporozoite monoclonal antibodies (MoAb) on the in-vitro development of liver stages of Plasmodium cynomolgi bastianellii (NIH strain) was evaluated using primary cultures of rhesus monkey hepatocytes. MoAbs against the circumsporozoite proteins of five strains of P. cynomolgi (NIH, London, Gombak, Ceylon, Berok), and of P. knowlesi (H strain) were used. Incubation of sporozoites of P. cynomolgi bastianellii with the anti-NIH strain MoAbs entirely prevented liver-stage development; MoAbs produced against the other four strains had no apparent activity. The anti-P. knowlesi MoAbs had a partially inhibitory effect on parasite development. These functional studies complement previous immunological studies on P. cynomolgi strain specificity, and confirm the cross-reactivity observed previously between sporozoites of P. cynomolgi bastianellii and P. knowlesi (H strain).


Subject(s)
Antibodies, Protozoan/administration & dosage , Plasmodium/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Cells, Cultured , Cross Reactions , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Plasmodium/growth & development , Species Specificity
19.
J Protozool ; 38(6): 42S-43S, 1991.
Article in English | MEDLINE | ID: mdl-1818193

ABSTRACT

Leghorn hens were subcutaneously immunized with 25 micrograms of Cryptosporidium parvum oocyst emulsified in Freund's complete adjuvant. A booster dose was injected 5 weeks later. Anti-Cryptosporidium activities of yolks and sera measured by an enzyme-linked immunosorbent assay (ELISA), demonstrated high levels in both sera and egg yolks which persisted for at least 17 wk. Preparations from yolks with high, medium and low anti-Cryptosporidium ELISA activities were used in a neonatal mouse model to assess their biological activities. A significant parasite reduction (P less than or equal to 0.001) was found between the high and all other groups. Hyperimmune eggs could be used as a source for passive immunity in cryptosporidiosis.


Subject(s)
Antibodies, Protozoan/isolation & purification , Chickens/immunology , Cryptosporidium parvum/immunology , Immunization, Passive , Animals , Antibodies, Protozoan/administration & dosage , Cryptosporidiosis/prevention & control , Egg Yolk/immunology , Enzyme-Linked Immunosorbent Assay , Evaluation Studies as Topic , Female , Mice , Mice, Inbred BALB C , Pregnancy , Random Allocation
20.
Infect Immun ; 66(9): 4469-73, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9712802

ABSTRACT

Cryptosporidium parvum is an important diarrhea-causing protozoan parasite of immunocompetent and immunocompromised hosts. Immunoglobulin A (IgA) has been implicated in resistance to mucosal infections with bacteria, viruses, and parasites, but little is known about the role of IgA in the control of C. parvum infection. We assessed the role of IgA during C. parvum infection in neonatal mice. IgA-secreting hybridomas were developed by using Peyer's patch lymphocytes from BALB/c mice which had been orally inoculated with viable C. parvum oocysts. Six monoclonal antibodies (MAbs) were selected for further study based on indirect immunofluorescence assay reactivity with sporozoite and merozoite pellicles and the antigen (Ag) deposited on glass substrate by gliding sporozoites. Each MAb was secreted in dimeric form and recognized a 23-kDa sporozoite Ag in Western immunoblots. The Ag recognized comigrated in sodium dodecyl sulfate-polyacrylamide gel electrophoresis with P23, a previously defined neutralization-sensitive zoite pellicle Ag. MAbs were evaluated for prophylactic or therapeutic efficacy against C. parvum, singly and in combinations, in neonatal BALB/c mice. A combination of two MAbs given prophylactically prior to and 12 h following oocyst challenge reduced the number of intestinal parasites scored histologically by 21.1% compared to the numbers in mice given an isotype-matched control MAb (P < 0.01). Individual MAbs given therapeutically in nine doses over a 96-h period following oocyst challenge increased efficacy against C. parvum infection. Four MAbs given therapeutically each reduced intestinal infection 34.4 to 42.2% compared to isotype-matched control MAb-treated mice (P < 0.05). One MAb reduced infection 63.3 and 72. 7% in replicate experiments compared to isotype-matched control MAb-treated mice (P < 0.0001). We conclude that IgA MAbs directed to neutralization-sensitive P23 epitopes may have utility in passive immunization against murine C. parvum infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cryptosporidiosis/prevention & control , Cryptosporidium parvum/immunology , Immunization, Passive , Immunoglobulin A/immunology , Protozoan Vaccines/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Protozoan/administration & dosage , Immunity, Mucosal , Immunoglobulin A/administration & dosage , Mice , Mice, Inbred BALB C , Peyer's Patches , Protozoan Vaccines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL