Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Prev Vet Med ; 176: 104904, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32066023

ABSTRACT

Cryptosporidiosis is one of the most common zoonosis worldwide, causing intestinal infection to both humans and livestock. The purpose of this study was to assess whether the level of anti-C. parvum IgG antibodies transferred through colostrum from dams to newborn calves impacts the susceptibility to cryptosporidiosis. A number of 50 dams and their healthy newborns were included in the study. Colostrum samples were collected within 12 h after birth and anti-C. parvum IgG antibody levels were determined by single radial immunodiffusion. The health condition of the newborns was daily monitored, and fecal samples were collected at first diarrheic episode of a calf. In all dams, the anti-C. parvum IgG antibody concentration in colostrum varied between 570 and 4070 mg/dl; in dams who gave birth to calves with diarrhea and were C. parvum-positive, the antibody concentration in colostrum varied between 680 and 3680 mg/dl (Table 1). The point-biserial correlation showed a negative correlation between the levels of anti-C. parvum antibodies and manifestation of clinical cryptosporidiosis (r=-0.425). Our findings highlight the importance of IgG levels in colostrum received by neonatal calves during their first day of life for prevention of C. parvum infection.


Subject(s)
Antibodies, Protozoan/physiology , Cattle Diseases/immunology , Colostrum/immunology , Cryptosporidiosis/immunology , Cryptosporidium parvum/physiology , Diarrhea/veterinary , Immunoglobulin G/physiology , Animals , Animals, Newborn/immunology , Cattle/immunology , Cattle Diseases/parasitology , Cattle Diseases/prevention & control , Cryptosporidiosis/parasitology , Cryptosporidiosis/prevention & control , Diarrhea/immunology , Diarrhea/parasitology , Diarrhea/prevention & control , Greece
2.
Malar J ; 17(1): 370, 2018 Oct 17.
Article in English | MEDLINE | ID: mdl-30333026

ABSTRACT

BACKGROUND: Plasmodium vivax is the most geographically widespread of the human malaria parasites, causing 50,000 to 100,000 deaths annually. Plasmodium vivax parasites have the unique feature of forming dormant liver stages (hypnozoites) that can reactivate weeks or months after a parasite-infected mosquito bite, leading to new symptomatic blood stage infections. Efforts to eliminate P. vivax malaria likely will need to target the persistent hypnozoites in the liver. Therefore, research on P. vivax liver stages necessitates a marker for clearly distinguishing between actively replicating parasites and dormant hypnozoites. Hypnozoites possess a densely fluorescent prominence in the parasitophorous vacuole membrane (PVM) when stained with antibodies against the PVM-resident protein Upregulated in Infectious Sporozoites 4 (PvUIS4), resulting in a key feature recognizable for quantification of hypnozoites. Thus, PvUIS4 staining, in combination with the characteristic small size of the parasite, is currently the only hypnozoite-specific morphological marker available. RESULTS: Here, the generation and validation of a recombinant monoclonal antibody against PvUIS4 (α-rUIS4 mAb) is described. The variable heavy and light chain domains of an α-PvUIS4 hybridoma were cloned into murine IgG1 and IgK expression vectors. These expression plasmids were co-transfected into HEK293 cells and mature IgG was purified from culture supernatants. It is shown that the α-rUIS4 mAb binds to its target with high affinity. It reliably stains the schizont PVM and the hypnozoite-specific PVM prominence, enabling the visual differentiation of hypnozoites from replicating liver stages by immunofluorescence assays in different in vitro settings, as well as in liver sections from P. vivax infected liver-chimeric mice. The antibody functions reliably against all four parasite isolates tested and will be an important tool in the identification of the elusive hypnozoite. CONCLUSIONS: The α-rUIS4 mAb is a versatile tool for distinguishing replicating P. vivax liver stages from dormant hypnozoites, making it a valuable resource that can be deployed throughout laboratories worldwide.


Subject(s)
Antibodies, Protozoan/physiology , Liver/parasitology , Plasmodium vivax/isolation & purification , Sporozoites/immunology , Biomarkers/analysis
3.
Infect Immun ; 84(4): 950-963, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26787721

ABSTRACT

Severe malaria (SM) is a life-threatening complication of infection with Plasmodium falciparum Epidemiological observations have long indicated that immunity against SM is acquired relatively rapidly, but prospective studies to investigate its immunological basis are logistically challenging and have rarely been undertaken. We investigated the merozoite targets and antibody-mediated mechanisms associated with protection against SM in Kenyan children aged 0 to 2 years. We designed a unique prospective matched case-control study of well-characterized SM clinical phenotypes nested within a longitudinal birth cohort of children (n= 5,949) monitored over the first 2 years of life. We quantified immunological parameters in sera collected before the SM event in cases and their individually matched controls to evaluate the prospective odds of developing SM in the first 2 years of life. Anti-AMA1 antibodies were associated with a significant reduction in the odds of developing SM (odds ratio [OR] = 0.37; 95% confidence interval [CI] = 0.15 to 0.90; P= 0.029) after adjustment for responses to all other merozoite antigens tested, while those against MSP-2, MSP-3, Plasmodium falciparum Rh2 [PfRh2], MSP-119, and the infected red blood cell surface antigens were not. The combined ability of total IgG to inhibit parasite growth and mediate the release of reactive oxygen species from neutrophils was associated with a marked reduction in the odds of developing SM (OR = 0.07; 95% CI = 0.006 to 0.82;P= 0.03). Assays of these two functional mechanisms were poorly correlated (Spearman rank correlation coefficient [rs] = 0.12;P= 0.07). Our data provide epidemiological evidence that multiple antibody-dependent mechanisms contribute to protective immunity via distinct targets whose identification could accelerate the development of vaccines to protect against SM.


Subject(s)
Antibodies, Protozoan/physiology , Malaria, Falciparum/immunology , Plasmodium falciparum/physiology , Aging , Antigens, Protozoan/immunology , Case-Control Studies , Child , Child, Preschool , Growth Inhibitors/metabolism , Humans , Infant , Kenya/epidemiology , Merozoites/immunology , Odds Ratio , Respiratory Burst/physiology
4.
Vet Parasitol ; 214(1-2): 213-8, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26428018

ABSTRACT

Ovine babesiosis, caused by the intra-erythrocytic protozoan parasite Babesia ovis, is an infectious and economically important tick-borne disease of sheep. Diagnostic testing is an essential tool used for the control of the disease. In order to identify and characterize the immunoreactive proteins which are useful in serological diagnosis of the disease, a complementary DNA (cDNA) expression library was constructed from B. ovis merozoite mRNA. A cDNA clone designated as BoSA2 was identified by immunoscreening of a cDNA library using immune sheep serum. The sequence of the BoSA2 cDNA had a partial open reading frame of 1156 nucleotides encoding a polypeptide of 384 amino acid residues. Theoretical molecular mass for the mature protein was 43.5 kDa. The sequence of the BoSA2 was inserted into the expression vector pGEX-4T-1 and then expressed in Escherichia coli DH5α cells as a glutathione S-transferase (GST)-tagged fusion protein. This recombinant fusion protein (rBoSA2) was purified by GST-affinity chromatography. Immunoreactivity of the rBoSA2 was evaluated by indirect enzyme-linked immunosorbent assay (ELISA) using the sera from the animals naturally and experimentally infected with B. ovis. ELISA results demonstrated that this antigen was useful for the diagnosis of ovine babesiosis. The localization of the BoSA2 protein was shown in and on the parasite and in the cytoplasm of the infected erythrocyte by confocal laser microscope. To our knowledge, rBoSA2 is the second immunoreactive recombinant protein of B. ovis until the present.


Subject(s)
Antibodies, Protozoan/physiology , Babesia/metabolism , Protozoan Proteins/metabolism , Sheep Diseases/parasitology , Animals , Antibodies, Protozoan/blood , Babesia/genetics , Babesia/immunology , Blotting, Western , DNA, Complementary , Enzyme-Linked Immunosorbent Assay/veterinary , Escherichia coli , Gene Expression Regulation , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins , Sheep , Sheep Diseases/blood , Sheep Diseases/immunology
5.
ACS Chem Biol ; 10(2): 460-5, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25360987

ABSTRACT

Surface display of engineered proteins has many useful applications. The expression of a synthetic chimeric antigen receptor composed of an extracellular tumor-specific antibody fragment linked to a cytosolic activating motif in engineered T cells is now considered a viable approach for the treatment of leukemias. The risk of de novo tumor development, inherent in the transfer of genetically engineered cells, calls for alternative approaches for the functionalization of the lymphocyte plasma membrane. We demonstrate the conjugation of LPXTG-tagged probes and LPXTG-bearing proteins to endogenous acceptors at the plasma membrane in a single step using sortase A. We successfully conjugated biotin probes not only to mouse hematopoietic cells but also to yeast cells, 293T cells, and Toxoplasma gondii. Installation of single domain antibodies on activated CD8 T cell redirects cell-specific cytotoxicity to cells that bear the relevant antigen. Likewise, conjugation of Toxoplasma gondii with single domain antibodies targets the pathogen to cells that express the antigen recognized by these single domain antibodies. This simple and robust enzymatic approach enables engineering of the plasma membrane for research or therapy under physiological reaction conditions that ensure the viability of the modified cells.


Subject(s)
Antibodies, Protozoan/physiology , Protozoan Proteins/metabolism , Toxoplasma/cytology , Toxoplasma/physiology , Animals , Antigens , Bioengineering , CD8-Positive T-Lymphocytes , Gene Expression Regulation, Enzymologic , Humans , Mice , Protein Structure, Tertiary , Protozoan Proteins/genetics
6.
Infect Immun ; 82(5): 1860-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24566620

ABSTRACT

Protective immunity to Plasmodium falciparum malaria acquired after natural exposure is largely antibody mediated. IgG-specific P. falciparum EMP1 (PfEMP1) proteins on the infected erythrocyte surface are particularly important. The transient antibody responses and the slowly acquired protective immunity probably reflect the clonal antigenic variation and allelic polymorphism of PfEMP1. However, it is likely that other immune-evasive mechanisms are also involved, such as interference with formation and maintenance of immunological memory. We measured PfEMP1-specific antibody levels by enzyme-linked immunosorbent assay (ELISA) and memory B-cell frequencies by enzyme-linked immunosorbent spot (ELISPOT) assay in a cohort of P. falciparum-exposed nonpregnant Ghanaian women. The antigens used were a VAR2CSA-type PfEMP1 (IT4VAR04) with expression restricted to parasites infecting the placenta, as well as two commonly recognized PfEMP1 proteins (HB3VAR06 and IT4VAR60) implicated in rosetting and not pregnancy restricted. This enabled, for the first time, a direct comparison in the same individuals of immune responses specific for a clinically important parasite antigen expressed only during well-defined periods (pregnancy) to responses specific for comparable antigens expressed independent of pregnancy. Our data indicate that PfEMP1-specific B-cell memory is adequately acquired even when antigen exposure is infrequent (e.g., VAR2CSA-type PfEMP1). Furthermore, immunological memory specific for VAR2CSA-type PfEMP1 can be maintained for many years without antigen reexposure and after circulating antigen-specific IgG has disappeared. The study provides evidence that natural exposure to P. falciparum leads to formation of durable B-cell immunity to clinically important PfEMP1 antigens. This has encouraging implications for current efforts to develop PfEMP1-based vaccines.


Subject(s)
Antigens, Protozoan/physiology , B-Lymphocytes/physiology , Malaria, Falciparum/immunology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Adult , Antibodies, Protozoan/physiology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/physiology , Ghana/epidemiology , Humans , Immunoglobulin G/blood , Malaria, Falciparum/epidemiology , Pregnancy , Pregnancy Complications, Parasitic/epidemiology , Pregnancy Complications, Parasitic/immunology , Protozoan Proteins/genetics
7.
J Immunol ; 185(11): 6939-46, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21037092

ABSTRACT

There are >2 million new cases of leishmaniasis annually, and no effective vaccine has been developed to prevent infection. In murine infection, Leishmania mexicana, which lives intracellularly in host macrophages, has developed pathways to hijack host IgG to induce a suppressive IL-10 response through FcγRs, the cell-surface receptors for IgG. To guide vaccine development away from detrimental Ab responses, which can accompany attempts to induce cell-mediated immunity, it is crucial to know which isotypes of IgG are pathogenic in this infection. We found that IgG1 and IgG2a/c induce IL-10 from macrophages in vitro equally well but through different FcγR subtypes: IgG1 through FcγRIII and IgG2a/c through FcγRI primarily, but also through FcγRIII. In sharp contrast, mice lacking IgG1 develop earlier and stronger IgG2a/c, IgG3, and IgM responses to L. mexicana infection and yet are more resistant to the infection. Thus, IgG1, but not IgG2a/c or IgG3, is pathogenic in vivo, in agreement with prior studies indicating that FcγRIII is required for chronic disease. This calls into question the assumption that macrophages, which should secrete IL-10 in response to IgG1 and IgG2a/c immune complexes, are the most important source of IL-10 generated by IgG-FcγR engagement in L. mexicana infection. Further investigations are required to better determine the cell type responsible for this immunosuppressive FcγRIII-induced IL-10 pathway and whether IgG2a/c is protective.


Subject(s)
Antibodies, Protozoan/adverse effects , Immunoglobulin G/adverse effects , Leishmania mexicana/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/physiology , Bone Marrow Cells/immunology , Bone Marrow Cells/parasitology , Bone Marrow Cells/pathology , Cells, Cultured , Chronic Disease , Female , Immunoglobulin Class Switching , Immunoglobulin G/blood , Immunoglobulin G/physiology , Immunoglobulin M/adverse effects , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Immunophenotyping , Leishmaniasis, Cutaneous/pathology , Macrophages/immunology , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
8.
J Infect Dis ; 198(11): 1609-19, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18928377

ABSTRACT

BACKGROUND: Placental malaria (PM) and maternal infection with human immunodeficiency virus (HIV) type 1 have been shown to affect infant morbidity and immune responses to Plasmodium falciparum. We studied the effects of PM and HIV infection on the antimalarial antibody responses and morbidity outcomes of infants throughout the first year of life. METHODS: A total of 411 Kenyan infants who were born to mothers who were singly or dually infected with PM and/or HIV had their levels of immunoglobulin G antibody to 6 P. falciparum antigens/epitopes (apical membrane antigen-1, erythrocyte-binding antigen-175; liver-stage antigen-1 [LSA-1], circumsporozoite protein [CSP], merozoite surface protein-2, and rhoptry-associated protein-1 [RAP-1]) and to tetanus toxoid (TT) tested using enzyme-linked immunosorbent assay. RESULTS: PM had little effect on the antibody responses of infants, whereas maternal HIV infection resulted in decreased levels of antibody to LSA-1, CSP, and RAP-1 epitopes at birth, compared with the absence of PM and maternal HIV infection (P = .0063). Levels of antibodies to TT were significantly reduced in infants born to mothers coinfected with HIV and PM, compared with the levels noted in infants born to HIV-negative mothers (P = .0003). In HIV-infected infants, levels of antibody to TT were reduced, but levels of antibody to malarial antigens were not. Antimalarial antibody levels were positively associated with malaria-related morbidity outcomes. CONCLUSION: Infant HIV infection and maternal coinfection with HIV and PM negatively influence antibody responses to TT, but not those to malarial antigens, in infants. Antimalarial antibodies rarely showed protective associations with morbidity in infants and were more often a marker for malaria exposure and risk of infection.


Subject(s)
Antibodies, Protozoan/physiology , HIV Infections/complications , Malaria, Falciparum/immunology , Placenta/parasitology , Aging , Animals , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Kenya , Longitudinal Studies , Plasmodium falciparum , Pregnancy , Pregnancy Complications, Parasitic
9.
Malar J ; 6: 107, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17686163

ABSTRACT

BACKGROUND: Mosquito stage malaria vaccines are designed to induce an immune response in the human host that will block the parasite's growth in the mosquito and consequently block transmission of the parasite. A mosquito membrane-feeding assay (MFA) is used to test transmission-blocking activity (TBA), but in this technique cannot accommodate many samples. A clear understanding of the relationship between antibody levels and TBA may allow ELISA determinations to be used to predict TBA and assist in planning vaccine development. METHODS: Rabbit anti-Pfs25 sera and monkey anti-Pvs25 sera were generated and the antibody titers were determined by a standardized ELISA. The biological activity of the same sera was tested by MFA using Plasmodium gametocytes (cultured Plasmodium falciparum or Plasmodium vivax from malaria patients) and Anopheles mosquitoes. RESULTS: Anti-Pfs25 and anti-Pvs25 sera showed that ELISA antibody units correlate with the percent reduction in the oocyst density per mosquito (Spearman Rank correlations: 0.934 and 0.616, respectively), and fit a hyperbolic curve when percent reduction in oocyst density is plotted against antibody units of the tested sample. Antibody levels also correlated with the number of mosquitoes that failed to become infected, and this proportion can be calculated from the reduction in oocyst numbers and the distribution of oocysts per infected mosquito in control group. CONCLUSION: ELISA data may be used as a surrogate for the MFA to evaluate transmission-blocking vaccine efficacy. This will facilitate the evaluation of transmission-blocking vaccines and implementation of this malaria control strategy.


Subject(s)
Antibodies, Protozoan/blood , Antibodies, Protozoan/physiology , Antigens, Protozoan/immunology , Antigens, Surface/immunology , Immunization , Malaria Vaccines/immunology , Malaria, Falciparum/blood , Malaria, Vivax/blood , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Animals , Anopheles/parasitology , Anopheles/physiology , Cell Count , Culicidae/parasitology , Culicidae/physiology , Disease Transmission, Infectious/prevention & control , Female , Humans , Immunization Schedule , Injections, Intramuscular , Macaca mulatta , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Male , Oocytes/cytology , Rabbits , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
10.
J Immunol ; 179(1): 557-65, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579077

ABSTRACT

Chronic inflammation during placental malaria (PM) is most frequent in first time mothers and is associated with poor maternal and fetal outcomes. In the first genome-wide analysis of the local human response to sequestered malaria parasites, we identified genes associated with chronic PM and then localized the corresponding proteins and immune cell subsets in placental cryosections. B cell-related genes were among the most highly up-regulated transcripts in inflamed tissue. The B cell chemoattractant CXCL13 was up-regulated >1,000-fold, and B cell-activating factor was also detected. Both proteins were expressed by intervillous macrophages. Ig L and H chain transcription increased significantly, and heavy depositions of IgG3 and IgM were observed in intervillous spaces. The B cell phenotype was heterogeneous, including naive (CD27-negative), mature (CD138-positive), and cycling (Ki-67-positive) cells. B cells expressed T-bet but not Bcl-6, suggesting T cell-independent activation without germinal center formation. Genes for the Fc binding proteins FcgammaRIa, FcgammaRIIIa, and C1q were highly up-regulated, and the proteins localized to intervillous macrophages. Birth weight was inversely correlated with transcript levels of CXCL13, IgG H chain, and IgM H chain. The iron regulatory peptide hepcidin was also expressed but was not associated with maternal anemia. The results suggest that B cells and macrophages contribute to chronic PM in a process resembling lymphoid neogenesis. We propose a model where the production of Ig during chronic malaria may enhance inflammation by attracting and activating macrophages that, in turn, recruit B cells to further produce Ig in the intervillous spaces.


Subject(s)
Gene Expression Profiling , Lymphopoiesis/immunology , Malaria, Falciparum/immunology , Placenta Diseases/immunology , Adolescent , Adult , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/physiology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Movement/genetics , Cell Movement/immunology , Chemokine CXCL13 , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Chronic Disease , Female , Humans , Lymphopoiesis/genetics , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Malaria, Falciparum/genetics , Middle Aged , Placenta Diseases/genetics , Pregnancy , Up-Regulation/immunology
11.
Infect Immun ; 75(3): 1177-85, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17158893

ABSTRACT

A successful vaccine against Plasmodium vivax malaria would significantly improve the health and quality of the lives of more than 1 billion people around the world. A subunit vaccine is the only option in the absence of long-term culture of P. vivax parasites. The circumsporozoite protein that covers the surface of Plasmodium sporozoites is one of the best-studied malarial antigens and the most promising vaccine in clinical trials. We report here the development of a novel "immunologically optimal" recombinant vaccine expressed in Escherichia coli that encodes a chimeric CS protein encompassing repeats from the two major alleles, VK210 and VK247. This molecule is widely recognized by sera from patients naturally exposed to P. vivax infection and induces a highly potent immune response in genetically disparate strains of mice. Antibodies from immunized animals recognize both VK210 and VK247 sporozoites. Furthermore, these antibodies appear to be protective in nature since they cause the agglutination of live sporozoites, an in vitro surrogate of sporozoite infectivity. These results strongly suggest that recombinant CS is biologically active and highly immunogenic across major histocompatibility complex strains and raises the prospect that in humans this vaccine may induce protective immune responses.


Subject(s)
Antibodies, Protozoan/genetics , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Recombinant Fusion Proteins/immunology , Sporozoites/immunology , Animals , Antibodies, Protozoan/metabolism , Antibodies, Protozoan/physiology , Female , Humans , Malaria Vaccines/genetics , Malaria, Vivax/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Recombinant Fusion Proteins/genetics , Sporozoites/metabolism , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
12.
Microbes Infect ; 7(3): 375-84, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15784182

ABSTRACT

Trypanosoma cruzi expresses oligopeptidase B and cathepsin B that have important functions in the interaction with mammalian host cells. In this study, we demonstrated that sera from both chagasic rabbits and humans have specific antibodies to highly purified native oligopeptidase B and cathepsin B. Levels of antibodies to cathepsin B were higher than those observed to oligopeptidase B by absorbance values recorded upon ELISA. We next showed that 90% and 30% of sera from individuals with mucocutaneous leishmaniasis have antibodies that recognize oligopeptidase B and cathepsin B as antigens, respectively. In addition, 55% and 40% of sera from kala-azar patients have antibodies to oligopeptidase B and cathepsin B, respectively. Sera from malaria patients did not recognize the proteases as antigens. Despite high levels of specific antibodies, sera from T. cruzi-infected patients did not inhibit the activities of either oligopeptidase B or cathepsin B. Furthermore, sera or IgG purified from either infected or non-infected individuals enhanced the enzymatic activity of the secreted oligopeptidase B. Oligopeptidase B secreted by trypomastigotes and cathepsin B released upon parasite lysis retain their enzymatic activities and may be associated with Chagas' disease pathogenesis by hydrolyzing host proteins and inducing host immune responses.


Subject(s)
Antibodies, Protozoan/physiology , Cathepsin B/immunology , Immunoglobulin G/physiology , Serine Endopeptidases/immunology , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/immunology , Animals , Cathepsin B/physiology , Chagas Disease/immunology , Humans , Rabbits , Serine Endopeptidases/physiology
13.
J Immunol ; 172(10): 6167-74, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15128804

ABSTRACT

A Plasmodium falciparum chimeric protein 2.9 (PfCP-2.9) was constructed consisting of the C-terminal regions of two leading malaria vaccine candidates, domain III of apical membrane ag-1 (AMA-1) and 19-kDa C-terminal fragment of the merozoite surface protein 1 (MSP1). The PfCP-2.9 was produced by Pichia pastoris in secreted form with a yield of 2600 mg/L and approximately 1 g/L of final product was obtained from a three-step purification process. Analysis of conformational properties of the chimeric protein showed that all six conformational mAbs interacted with the recombinant protein were reduction-sensitive, indicating that fusion of the two cysteine-rich proteins retains critical conformational epitopes. PfCP-2.9 was found to be highly immunogenic in rabbits as well as in rhesus monkeys (Macaca mulatta). The chimeric protein induced both anti-MSP1-19 and anti-AMA-1(III) Abs at levels 11- and 18-fold higher, respectively, than individual components did. Anti-PfCP-2.9 sera from both rabbits and rhesus monkeys almost completely inhibited in vitro growth of the P. falciparum FCC1/HN and 3D7 lines when tested at a 6.7-fold dilution. It was shown that the inhibition is dependent on the presence of Abs to the chimeric protein and their disulfide bond-dependent conformations. Moreover, the activity was mediated by a combination of growth-inhibitory Abs generated by the individual MSP1-19 and AMA-1(III) of PfCP-2.9. The combination of the extremely high yield of the protein and enhancement of its immune response provides a basis to develop an effective and affordable malaria vaccine.


Subject(s)
Antibodies, Protozoan/physiology , Antigens, Protozoan/genetics , Antiprotozoal Agents/immunology , Calcium-Binding Proteins/genetics , Malaria Vaccines/genetics , Membrane Proteins/genetics , Plasmodium falciparum/growth & development , Protein Kinases/genetics , Protozoan Proteins/genetics , Recombinant Fusion Proteins/genetics , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/genetics , Animals , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/immunology , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/chemical synthesis , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/immunology , Growth Inhibitors/administration & dosage , Growth Inhibitors/chemical synthesis , Growth Inhibitors/genetics , Growth Inhibitors/immunology , Macaca mulatta , Malaria Vaccines/administration & dosage , Malaria Vaccines/chemical synthesis , Malaria Vaccines/immunology , Membrane Proteins/administration & dosage , Membrane Proteins/immunology , Molecular Sequence Data , Plasmodium falciparum/immunology , Protein Kinases/administration & dosage , Protein Kinases/immunology , Protozoan Proteins/administration & dosage , Protozoan Proteins/immunology , Rabbits , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/immunology , Subtilisins/administration & dosage , Subtilisins/genetics , Subtilisins/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/chemical synthesis
14.
J Immunol ; 171(10): 5461-9, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14607952

ABSTRACT

Maternal Abs generated as a result of prior exposure to infectious agents such as the malaria parasite are transferred from the mother through the placenta to the fetus. Numerous studies have attributed the resistance to malaria infection observed in neonates and infants up to 6 mo of age to the presence of maternally derived Abs. However, recent studies have produced conflicting results suggesting that alternative protective mechanisms may be responsible. Although the presence of maternally derived Abs in the infant is not disputed, their exact role in the infant is unknown. Even less clear is the effect that maternally derived Abs, if generated in response to vaccination, may have on the infant's ability to respond to malaria infection. Studies on mouse pups were performed to determine the role of the 19-kDa region of merozoite surface protein 1 (MSP1(19)) and Plasmodium yoelii-specific Abs in neonatal malaria infection and to examine their effect on the development of a specific immune response in the pup. It was shown that P. yoelii- and MSP1(19)-specific Abs transferred to the pup from the mother act to suppress the growth of the parasite in the pup. However, the maternally derived Abs interfered with the development of the pups' own Ab response to the parasite by altering the fine specificity of the response. These results suggest that immunizing women of child-bearing age with a malaria vaccine candidate such as MSP1(19) would not prevent the infant from producing Abs in response to malaria infection, but it may affect the region of the Ag to which it responds.


Subject(s)
Animals, Newborn/immunology , Animals, Newborn/parasitology , Antibodies, Protozoan/physiology , Malaria/immunology , Maternal-Fetal Exchange/immunology , Merozoite Surface Protein 1/physiology , Plasmodium yoelii/growth & development , Plasmodium yoelii/immunology , Animals , Animals, Newborn/growth & development , Animals, Suckling/immunology , Animals, Suckling/parasitology , Antibodies, Protozoan/biosynthesis , Epitopes/administration & dosage , Epitopes/immunology , Epitopes/physiology , Female , Immunization, Passive , Malaria/physiopathology , Malaria/prevention & control , Male , Merozoite Surface Protein 1/administration & dosage , Merozoite Surface Protein 1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Weight , Pregnancy , Protein Structure, Tertiary/physiology , Species Specificity
15.
J Immunol ; 169(2): 944-51, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12097400

ABSTRACT

Merozoite surface protein 1 (MSP1) of malaria parasites undergoes proteolytic processing at least twice before invasion into a new RBC. The 42-kDa fragment, a product of primary processing, is cleaved by proteolytic enzymes giving rise to MSP1(33), which is shed from the merozoite surface, and MSP1(19), which is the only fragment carried into a new RBC. In this study, we have identified T cell epitopes on MSP1(33) of Plasmodium yoelii and have examined their function in immunity to blood stage malaria. Peptides 20 aa in length, spanning the length of MSP1(33) and overlapping each other by 10 aa, were analyzed for their ability to induce T cell proliferation in immunized BALB/c and C57BL/6 mice. Multiple epitopes were recognized by these two strains of mice. Effector functions of the dominant epitopes were then investigated. Peptides Cm15 and Cm21 were of particular interest as they were able to induce effector T cells capable of delaying growth of lethal P. yoelii YM following adoptive transfer into immunodeficient mice without inducing detectable Ab responses. Homologs of these epitopes could be candidates for inclusion in a subunit vaccine.


Subject(s)
Antibodies, Protozoan/physiology , Epitopes, T-Lymphocyte/analysis , Epitopes, T-Lymphocyte/therapeutic use , Malaria/immunology , Malaria/prevention & control , Merozoite Surface Protein 1/therapeutic use , Plasmodium yoelii/growth & development , Plasmodium yoelii/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Cell Line/transplantation , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Immunity, Innate , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Immunodominant Epitopes/therapeutic use , Injections, Subcutaneous , Malaria/blood , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria Vaccines/therapeutic use , Merozoite Surface Protein 1/administration & dosage , Merozoite Surface Protein 1/analysis , Merozoite Surface Protein 1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Molecular Sequence Data , Molecular Weight , Peptide Fragments/administration & dosage , Peptide Fragments/analysis , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , T-Lymphocyte Subsets/transplantation
16.
J Immunol ; 168(3): 1348-55, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11801675

ABSTRACT

In this study, we investigated the role of endogenous IL-12 in protective immunity against blood-stage P. chabaudi AS malaria using IL-12 p40 gene knockout (KO) and wild-type (WT) C57BL/6 mice. Following infection, KO mice developed significantly higher levels of primary parasitemia than WT mice and were unable to rapidly resolve primary infection and control challenge infection. Infected KO mice had severely impaired IFN-gamma production in vivo and in vitro by NK cells and splenocytes compared with WT mice. Production of TNF-alpha and IL-4 was not compromised in infected KO mice. KO mice produced significantly lower levels of Th1-dependent IgG2a and IgG3 but a higher level of Th2-dependent IgG1 than WT mice during primary and challenge infections. Treatment of KO mice with murine rIL-12 during the early stage of primary infection corrected the altered IgG2a, IgG3, and IgG1 responses and restored the ability to rapidly resolve primary and control challenge infections. Transfer of immune serum from WT mice to P. chabaudi AS-infected susceptible A/J mice completely protected the recipients, whereas immune serum from KO mice did not, as evidenced by high levels of parasitemia and 100% mortality in recipient mice. Furthermore, depletion of IgG2a from WT immune serum significantly reduced the protective effect of the serum while IgG1 depletion had no significant effect. Taken together, these results demonstrate the protective role of a Th1-immune response during both acute and chronic phases of blood-stage malaria and extend the immunoregulatory role of IL-12 to Ab-mediated immunity against Plasmodium parasites.


Subject(s)
Antibodies, Protozoan/physiology , Antibody-Dependent Cell Cytotoxicity/immunology , Erythrocytes/parasitology , Interleukin-12/physiology , Malaria/immunology , Malaria/prevention & control , Plasmodium chabaudi/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibody-Dependent Cell Cytotoxicity/genetics , Cells, Cultured , Cytokines/biosynthesis , Disease Progression , Erythrocytes/immunology , Female , Immune Sera/administration & dosage , Immune Sera/genetics , Immunization, Passive , Immunoglobulin G/biosynthesis , Immunoglobulin G/physiology , Injections, Intravenous , Interleukin-12/deficiency , Interleukin-12/genetics , Killer Cells, Natural/immunology , Malaria/genetics , Malaria/parasitology , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Plasmodium chabaudi/growth & development
17.
Vet Immunol Immunopathol ; 76(3-4): 309-17, 2000 Oct 31.
Article in English | MEDLINE | ID: mdl-11044562

ABSTRACT

Fecal samples obtained at intervals from six calves with acute cryptosporidiosis contained antibodies of multiple isotypes to p23. IgM-, IgA-, and IgG(1)-isotype anti-p23 appeared before IgG(2)-isotype antibodies. All anti-p23 antibodies had declined by 2 months after infection. One calf that failed to shed oocysts following initial exposure developed IgG(1)-isotype anti-p23 antibodies. One calf that died following exposure to Cryptosporidium parvum oocysts lacked detectable anti-p23 antibodies. Re-inoculation with C. parvum resulted in a brief, marked recall response to p23.


Subject(s)
Antibodies, Protozoan/immunology , Cattle Diseases/immunology , Cryptosporidiosis/veterinary , Cryptosporidium parvum/immunology , Immunity, Mucosal/physiology , Animals , Animals, Newborn , Antibodies, Protozoan/physiology , Cattle , Cattle Diseases/parasitology , Cryptosporidiosis/immunology , Cryptosporidiosis/parasitology , Cryptosporidium parvum/growth & development , Feces/parasitology , Flow Cytometry/veterinary , Immunoassay/veterinary , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/physiology , Male , Microspheres , Specific Pathogen-Free Organisms
18.
J Immunol ; 165(1): 389-96, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861076

ABSTRACT

Apical membrane Ag 1 (AMA1) is a leading malaria vaccine candidate. Homologues of AMA1 can induce protection in mice and monkeys, but the mechanism of immunity is not understood. Mice immunized with a refolded, recombinant, Plasmodium chabaudi AMA1 fragment (AMA1B) can withstand subsequent challenge with P. chabaudi adami. Here we show that CD4+ T cell depletion, but not gammadelta T cell depletion, can cause a significant drop in antiparasite immunity in either immunized normal or immunized B cell KO mice. In normal mice, this loss of immunity is not accompanied by a decline in Ab levels. These observations indicate a role for AMA1-specific Ab-independent T cell-mediated immunity. However, the loss of immunity in normal CD4+ T cell-depleted mice is temporary. Furthermore, immunized B cell KO mice cannot survive infection, demonstrating the absolute importance of B cells, and presumably Ab, in AMA1-induced immunity. CD4+ T cells specific for a cryptic conserved epitope on AMA1 can adoptively transfer protection to athymic (nu/nu) mice, the level of which is enhanced by cotransfer of rabbit anti-AMA1-specific antisera. Recipients of rabbit antisera alone do not survive. Some protected recipients of T cells plus antisera do not develop their own AMA 1-specific Ab response, suggesting that AMA 1-specific CMI alone can protect mice. These data are the first to demonstrate the specificity of any protective CMI response in malaria and have important implications for developing a malaria vaccine.


Subject(s)
Antibodies, Protozoan/physiology , Antigens, Protozoan/immunology , CD4-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria/immunology , Malaria/prevention & control , Membrane Proteins/immunology , Plasmodium chabaudi/immunology , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/biosynthesis , Antibody Specificity , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/genetics , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Female , Immune Sera/administration & dosage , Immunity, Innate , Injections, Intraperitoneal , Lymphocyte Depletion , Lymphopenia/genetics , Lymphopenia/immunology , Malaria/parasitology , Malaria Vaccines/administration & dosage , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Molecular Sequence Data , Parasitemia/immunology , Parasitemia/prevention & control , Plasmodium chabaudi/genetics , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
19.
Braz J Med Biol Res ; 33(3): 253-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10719375

ABSTRACT

Host resistance to Trypanosoma cruzi is dependent on both natural and acquired immune responses. During the acute phase of the infection the presence of IFN-gamma, TNF-alpha, IL-12 and GM-CSF has been closely associated with resistance, whereas TGF-ss and IL-10 have been associated with susceptibility. Several investigators have demonstrated that antibodies are responsible for the survival of susceptible animals in the initial phase of infection and for the maintenance of low levels of parasitemia in the chronic phase. However, how this occurs is not yet understood. Our results and other data in the literature support the hypothesis that the protective role of antibodies in the acute phase of infection is dependent mostly on their ability to induce removal of bloodstream trypomastigotes from the circulation in addition to other concomitant cell-mediated events.


Subject(s)
Antibodies, Protozoan/physiology , Chagas Disease/immunology , Trypanosoma cruzi/immunology , Animals , Cytokines/physiology , Disease Susceptibility , Mice
20.
Braz. j. med. biol. res ; 33(3): 253-8, Mar. 2000.
Article in English | LILACS | ID: lil-255044

ABSTRACT

Host resistance to Trypanosoma cruzi is dependent on both natural and acquired immune responses. During the acute phase of the infection the presence of IFN-gama, TNF-alpha, IL-12 and GM-CSF has been closely associated with resistance, whereas TGF-beta and IL-10 have been associated with susceptibility. Several investigators have demonstrated that antibodies are responsible for the survival of susceptible animals in the initial phase of infection and for the maintenance of low levels of parasitemia in the chronic phase. However, how this occurs is not yet understood. Our results and other data in the literature support the hypothesis that the protective role of antibodies in the acute phase of infection is dependent mostly on their ability to induce removal of bloodstream trypomastigotes from the circulation in addition to other concomitant cell-mediated events.


Subject(s)
Animals , Mice , Antibodies, Protozoan/physiology , Trypanosoma cruzi/immunology , Cytokines/physiology , Disease Susceptibility
SELECTION OF CITATIONS
SEARCH DETAIL
...