Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 7(5): e37019, 2012.
Article in English | MEDLINE | ID: mdl-22615877

ABSTRACT

Adjuvants potentiate antigen-specific protective immune responses and can be key elements promoting vaccine effectiveness. We previously reported that the Onchocerca volvulus recombinant protein rOv-ASP-1 can induce activation and maturation of naïve human DCs and therefore could be used as an innate adjuvant to promote balanced Th1 and Th2 responses to bystander vaccine antigens in mice. With a few vaccine antigens, it also promoted a Th1-biased response based on pronounced induction of Th1-associated IgG2a and IgG2b antibody responses and the upregulated production of Th1 cytokines, including IL-2, IFN-γ, TNF-α and IL-6. However, because it is a protein, the rOv-ASP-1 adjuvant may also induce anti-self-antibodies. Therefore, it was important to verify that the host responses to self will not affect the adjuvanticity of rOv-ASP-1 when it is used in subsequent vaccinations with the same or different vaccine antigens. In this study, we have established rOv-ASP-1's adjuvanticity in mice during the course of two sequential vaccinations using two vaccine model systems: the receptor-binding domain (RBD) of SARS-CoV spike protein and a commercial influenza virus hemagglutinin (HA) vaccine comprised of three virus strains. Moreover, the adjuvanticity of rOv-ASP-1 was retained with an efficacy similar to that obtained when it was used for a first vaccination, even though a high level of anti-rOv-ASP-1 antibodies was present in the sera of mice before the administration of the second vaccine. To further demonstrate its utility as an adjuvant for human use, we also immunized non-human primates (NHPs) with RBD plus rOv-ASP-1 and showed that rOv-ASP-1 could induce high titres of functional and protective anti-RBD antibody responses in NHPs. Notably, the rOv-ASP-1 adjuvant did not induce high titer antibodies against self in NHPs. Thus, the present study provided a sound scientific foundation for future strategies in the development of this novel protein adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Helminth/immunology , Helminth Proteins/immunology , Influenza Vaccines/immunology , Onchocerca volvulus/immunology , Onchocerca volvulus/metabolism , Recombinant Proteins/immunology , Animals , Antibody Formation , Female , HEK293 Cells , Hemagglutinins/immunology , Humans , Mice , Mice, Inbred BALB C , Primates/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Vaccination/methods
2.
Protein Expr Purif ; 83(2): 145-51, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22503665

ABSTRACT

The enzyme Necator americanus glutathione S-transferase 1 (Na-GST-1) belongs to a unique Nu class of GSTs and is a lead candidate antigen in a bivalent human hookworm vaccine. Here we describe the expression of Na-GST-1 in the yeast Pichia pastoris at the 20 L manufacturing scale and its purification process performed by three chromatographic steps, comprised of a Q Sepharose XL anion exchange column, followed by a Butyl Sepharose HP hydrophobic affinity column and a Superdex 75 size-exclusion column. Approximately 1.5 g of recombinant protein was recovered at an overall process yield of 51%, with a purity grade of 98% and the absence of detectable host cell protein. By mass spectrometry the recombinant protein exhibits a mass of 23,676Da, which closely matches the predicted molecular mass of the protein. The expression and purification methods described here are suitable for further scale-up product development and for its use to design formulation processes suitable to generate a vaccine for clinical testing.


Subject(s)
Antigens, Helminth/isolation & purification , Glutathione Transferase/isolation & purification , Helminth Proteins/isolation & purification , Necator americanus/enzymology , Recombinant Proteins/isolation & purification , Animals , Antigens, Helminth/genetics , Antigens, Helminth/metabolism , Chromatography, Gel , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Helminth Proteins/genetics , Helminth Proteins/metabolism , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Infect Immun ; 78(4): 1552-63, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20145100

ABSTRACT

Hookworm glutathione S-transferases (GSTs) are critical for parasite blood feeding and survival and represent potential targets for vaccination. Three cDNAs, each encoding a full-length GST protein from the human hookworm Necator americanus (and designated Na-GST-1, Na-GST-2, and Na-GST-3, respectively) were isolated from cDNA based on their sequence similarity to Ac-GST-1, a GST from the dog hookworm Ancylostoma caninum. The open reading frames of the three N. americanus GSTs each contain 206 amino acids with 51% to 69% sequence identity between each other and Ac-GST-1. Sequence alignment with GSTs from other organisms shows that the three Na-GSTs belong to a nematode-specific nu-class GST family. All three Na-GSTs, when expressed in Pichia pastoris, exhibited low lipid peroxidase and glutathione-conjugating enzymatic activities but high heme-binding capacities, and they may be involved in the detoxification and/or transport of heme. In two separate vaccine trials, recombinant Na-GST-1 formulated with Alhydrogel elicited 32 and 39% reductions in adult hookworm burdens (P < 0.05) following N. americanus larval challenge relative to the results for a group immunized with Alhydrogel alone. In contrast, no protection was observed in vaccine trials with Na-GST-2 or Na-GST-3. On the basis of these and other preclinical data, Na-GST-1 is under possible consideration for further vaccine development.


Subject(s)
Antigens, Helminth/immunology , Antigens, Helminth/metabolism , Glutathione Transferase/immunology , Glutathione Transferase/metabolism , Heme/metabolism , Necator americanus/enzymology , Necator americanus/immunology , Necatoriasis/prevention & control , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Amino Acid Sequence , Animals , Antigens, Helminth/genetics , Cloning, Molecular , Cricetinae , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , DNA, Helminth/genetics , DNA, Helminth/isolation & purification , Gene Expression , Glutathione/metabolism , Glutathione Transferase/genetics , Humans , Lipid Peroxidation , Molecular Sequence Data , Necator americanus/genetics , Necatoriasis/immunology , Open Reading Frames , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Vaccines, Subunit/immunology
4.
Article in English | MEDLINE | ID: mdl-20124715

ABSTRACT

Human hookworms are among the most pathogenic soil-transmitted helminths. These parasitic nematodes have co-evolved with the host and are able to maintain a high worm burden for decades without killing the human host. However, it is possible to develop vaccines against laboratory-challenge hookworm infections using either irradiated third-state infective larvae (L3) or enzymes from the adult parasites. In an effort to control hookworm infection globally, the Human Hookworm Vaccine Initiative, a product-development partnership with the Sabin Vaccine Institute to develop new control tools including vaccines, has identified a battery of protein antigens, including surface-associated antigens (SAAs) from L3. SAA proteins are characterized by a 13 kDa conserved domain of unknown function. SAA proteins are found on the surface of infective L3 stages (and some adult stages) of different nematode parasites, suggesting that they may play important roles in these organisms. The atomic structures and function of SAA proteins remain undetermined and in an effort to remedy this situation recombinant Na-SAA-2 from the most prevalent human hookworm parasite Necator americanus has been expressed, purified and crystallized. Useful X-ray data have been collected to 2.3 A resolution from a crystal that belonged to the monoclinic space group C2 with unit-cell parameters a = 73.88, b = 35.58, c = 42.75 A, beta = 116.1 degrees .


Subject(s)
Antigens, Helminth/chemistry , Necator americanus/chemistry , Amino Acid Sequence , Animals , Antigens, Helminth/genetics , Crystallography, X-Ray , Humans , Molecular Sequence Data , Necator americanus/genetics , Sequence Alignment , Sequence Homology, Amino Acid , X-Ray Diffraction
5.
Mol Biochem Parasitol ; 162(2): 142-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18804124

ABSTRACT

Ac-TMP-2, an immunodominant hookworm antigen encoding a tissue inhibitor of metalloproteinase (TIMP) was cloned by immunoscreening an Ancylostoma caninum larval cDNA library with sera pooled from dogs immunized with irradiated A. caninum third stage larvae (ir-L3). The open reading frame of Ac-tmp-2 cDNA encoded a 244 amino acids (predicted molecular weight of 27.7 kDa), which shared a common N-terminus with other vertebrate and invertebrate TIMPs, including Ac-TMP-1, the most abundant adult hookworm secreted protein. However Ac-TMP-2 also contains an unusual multicopy (ten) repeat of the amino acid sequence, KTVEENDE. By immunoblotting, Ac-TMP-2 was detected only in adult hookworms and their excretory secretory products although the corresponding mRNA was also detected in L3. Immunolocalization with specific antiserum showed that native Ac-TMP-2 was located in adult worm's esophagus and cephalic glands. Recombinant Ac-TMP-2 expressed in bacteria was highly immunogenic and recognized by ir-L3 immunized dog immune sera. The recombinant Ac-TMP-2 protein inhibited the human matrix metalloproteinases, MMP-2, MMP-7 and MMP-13. As an immunodominant protein having a possible role in the parasite-host relationship of canine hookworm infection, recombinant Ac-TMP-2 represents a plausible target for vaccine development.


Subject(s)
Ancylostoma/metabolism , Antigens, Helminth/chemistry , Antigens, Helminth/genetics , Helminth Proteins/chemistry , Helminth Proteins/genetics , Tissue Inhibitor of Metalloproteinases/chemistry , Tissue Inhibitor of Metalloproteinases/genetics , Amino Acid Sequence , Ancylostoma/enzymology , Animals , Antigens, Helminth/analysis , Cloning, Molecular , Dogs , Helminth Proteins/analysis , Humans , Matrix Metalloproteinases, Secreted/antagonists & inhibitors , Matrix Metalloproteinases, Secreted/metabolism , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinases/analysis
6.
Exp Parasitol ; 118(1): 32-40, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17645877

ABSTRACT

We have previously reported the successful adaptation of human hookworm Necator americanus in the golden hamster, Mesocricetus auratus. This animal model was used to test a battery of hookworm (N. americanus and Ancylostoma caninum) recombinant antigens as potential vaccine antigens. Hamsters immunized a leading vaccine candidate N. americanus-Ancylostoma secreted protein 2 (Na-ASP-2) and challenged with N. americanus infective larvae (L3), resulted in 30-46.2% worm reduction over the course of three vaccine trials, relative to adjuvant controls. In addition, significant reduction of worm burdens was also observed in the hamsters immunized with adult hookworm antigens A. caninum aspartic protease 1 (Ac-APR-1); A. caninum-glutathione-S transferase 1 (Ac-GST-1) and Necator cysteine proteases 2 (Na-CP-2) (44.4%, 50.6%, and 29.3%, respectively). Our data on the worm burden reductions afforded by these hookworm antigens approximate the level of protection reported previously from dogs challenged with A. caninum L3, and provide additional evidence to support these hookworm antigens as vaccine candidates for human hookworm infection. The hamster model of N. americanus provides useful information for the selection of antigens to be tested in downstream vaccine development.


Subject(s)
Antigens, Helminth/immunology , Necator americanus/immunology , Necatoriasis/prevention & control , Vaccines, Synthetic , Amino Acid Sequence , Animals , Antigens, Helminth/genetics , Antigens, Surface/genetics , Antigens, Surface/immunology , Base Sequence , Cloning, Molecular , Cricetinae , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Disease Models, Animal , Humans , Larva/immunology , Male , Mesocricetus , Molecular Sequence Data , Open Reading Frames/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Vaccines, Synthetic/standards
7.
BMC Struct Biol ; 7: 42, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17594497

ABSTRACT

BACKGROUND: Human hookworm infection is a major cause of anemia and malnutrition of adults and children in the developing world. As part of on-going efforts to control hookworm infection, The Human Hookworm Vaccine Initiative has identified candidate vaccine antigens from the infective L3 larval stages and adult stages of the parasite. Adult stage antigens include the cytosolic glutathione-S-transferases (GSTs). Nematode GSTs facilitate the inactivation and degradation of a variety of electrophilic substrates (drugs) via the nucleophilic addition of reduced glutathione. Parasite GSTs also play significant roles in multi-drug resistance and the modulation of host-immune defense mechanisms. RESULTS: The crystal structures of Na-GST-1 and Na-GST-2, two major GSTs from Necator americanus the main human hookworm parasite, have been solved at the resolution limits of 2.4 A and 1.9 A respectively. The structure of Na-GST-1 was refined to R-factor 18.9% (R-free 28.3%) while that of Na-GST-2 was refined to R-factor 17.1% (R-free 21.7%). Glutathione usurped during the fermentation process in bound in the glutathione binding site (G-site) of each monomer of Na-GST-2. Na-GST-1 is uncomplexed and its G-site is abrogated by Gln 50. These first structures of human hookworm parasite GSTs could aid the design of novel hookworm drugs. CONCLUSION: The 3-dimensional structures of Na-GST-1 and Na-GST-2 show two views of human hookworm GSTs. While the GST-complex structure of Na-GST-2 reveals a typical GST G-site that of Na-GST-1 suggests that there is some conformational flexibility required in order to bind the substrate GST. In addition, the overall binding cavities for both are larger, more open, as well as more accessible to diverse ligands than those of GSTs from organisms that have other major detoxifying mechanisms. The results from this study could aid in the design of novel drugs and vaccine antigens.


Subject(s)
Ancylostomatoidea/enzymology , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Amino Acid Sequence , Ancylostomatoidea/genetics , Animals , Crystallography, X-Ray , Dimerization , Drug Design , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Humans , Models, Molecular , Molecular Sequence Data , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Structural Homology, Protein
8.
Mol Biochem Parasitol ; 152(2): 132-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17280728

ABSTRACT

Ac-MTP-2 is an astacin-like metalloprotease secreted by adult Ancylostoma caninum hookworms. Ac-mtp-2 cDNA was cloned by immunoscreening a cDNA library with antisera prepared against adult A. caninum excretory/secretory (ES) products. The full-length Ac-mtp-2 contains 850 bp cDNA encoding a 233 amino acid open reading frame (ORF) with 32% amino acid identity to Ce-NSP-4, a pharyngeal cell-derived secreted metalloprotease of the nematode Caenorhabditis elegans. The predicted ORF contained a conserved Met-turn sequence (SXMHY), but only a partial zinc-binding signature sequence (GXXXEHXRXER instead of HEXXHXXGXXHEXXRXDR) found in other astacins. However, by both gelatin gel electrophoresis and azocasein digestion, the recombinant Ac-MTP-2 exhibited proteolytic activity that was inhibited by the zinc chelator 1,10-phenanthroline and Ac-TMP, a putative tissue inhibitor of metalloprotease that was previously shown to be a highly abundant component of adult A. caninum ES products. By RT-PCR, Western blot Ac-MTP-2 was found only expressed in adult hookworms and secreted in the adult ES products. Immunolocalization with antisera shows that Ac-MTP-2 is located to the esophageal glands (confirming its role as a secretory protein), as well as to the parasite uterus. It is hypothesized that Ac-MTP-2 functions in the extracorporeal digestion of the intestinal mucosal plug lodged in the buccal capsule of the adult parasite.


Subject(s)
Ancylostoma/enzymology , Helminth Proteins/genetics , Metalloproteases/genetics , Amino Acid Sequence , Ancylostoma/metabolism , Animals , Cloning, Molecular , DNA, Complementary/metabolism , Fluorescent Antibody Technique , Helminth Proteins/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloproteases/metabolism , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment
9.
PLoS Med ; 2(10): e295, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16231975

ABSTRACT

BACKGROUND: Hookworms infect 730 million people in developing countries where they are a leading cause of intestinal blood loss and iron-deficiency anemia. At the site of attachment to the host, adult hookworms ingest blood and lyse the erythrocytes to release hemoglobin. The parasites subsequently digest hemoglobin in their intestines using a cascade of proteolysis that begins with the Ancylostoma caninum aspartic protease 1, APR-1. METHODS AND FINDINGS: We show that vaccination of dogs with recombinant Ac-APR-1 induced antibody and cellular responses and resulted in significantly reduced hookworm burdens (p = 0.056) and fecal egg counts (p = 0.018) in vaccinated dogs compared to control dogs after challenge with infective larvae of A. caninum. Most importantly, vaccinated dogs were protected against blood loss (p = 0.049) and most did not develop anemia, the major pathologic sequela of hookworm disease. IgG from vaccinated animals decreased the catalytic activity of the recombinant enzyme in vitro and the antibody bound in situ to the intestines of worms recovered from vaccinated dogs, implying that the vaccine interferes with the parasite's ability to digest blood. CONCLUSION: To the best of our knowledge, this is the first report of a recombinant vaccine from a hematophagous parasite that significantly reduces both parasite load and blood loss, and it supports the development of APR-1 as a human hookworm vaccine.


Subject(s)
Aspartic Acid Endopeptidases/pharmacology , Aspartic Acid Endopeptidases/therapeutic use , Hookworm Infections/drug therapy , Hookworm Infections/veterinary , Vaccination/veterinary , Amino Acid Sequence , Ancylostoma/pathogenicity , Anemia/etiology , Anemia/prevention & control , Animals , Antibody Formation , Dogs , Hemorrhage/etiology , Hemorrhage/prevention & control , Hookworm Infections/pathology , Immunoglobulin G/analysis , Male , Molecular Sequence Data , Recombinant Proteins
10.
Infect Immun ; 73(10): 6903-11, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16177370

ABSTRACT

We report the cloning and expression of Ac-GST-1, a novel glutathione S-transferase from the adult hookworm Ancylostoma caninum, and its possible role in parasite blood feeding and as a vaccine target. The predicted Ac-GST-1 open reading frame contains 207 amino acids (mass, 24 kDa) and exhibited up to 65% amino acid identity with other nematode GSTs. mRNA encoding Ac-GST-1 was detected in adults, eggs, and larval stages, but the protein was detected only in adult hookworm somatic extracts and excretory/secretory products. Using antiserum to the recombinant protein, Ac-GST-1 was immunolocalized to the parasite hypodermis and muscle tissue and weakly to the intestine. Recombinant Ac-GST-1 was enzymatically active, as determined by conjugation of glutathione to a model substrate, and exhibited a novel high-affinity binding site for hematin. The possible role of Ac-GST-1 in parasite heme detoxification during hemoglobin digestion or heme uptake prompted interest in evaluating it as a potential vaccine antigen. Vaccination of dogs with Ac-GST-1 resulted in a 39.4% reduction in the mean worm burden and 32.3% reduction in egg counts compared to control dogs following larval challenge, although the reductions were not statistically significant. However, hamsters vaccinated with Ac-GST-1 exhibited statistically significant worm reduction (53.7%) following challenge with heterologous Necator americanus larvae. These studies suggest that Ac-GST-1 is a possible drug and vaccine target for hookworm infection.


Subject(s)
Ancylostoma/immunology , Ancylostomiasis/prevention & control , Carrier Proteins/immunology , Glutathione Transferase/immunology , Hemeproteins/immunology , Vaccines , Amino Acid Sequence , Ancylostoma/enzymology , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , Cloning, Molecular , Cricetinae , Dogs , Glutathione Transferase/analysis , Glutathione Transferase/genetics , Heme-Binding Proteins , Hemeproteins/analysis , Hemeproteins/genetics , Larva/enzymology , Molecular Sequence Data , RNA, Messenger/analysis , RNA, Messenger/metabolism , Vaccines/genetics , Vaccines/immunology
11.
Vaccine ; 23(39): 4754-64, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16054275

ABSTRACT

The ASP-2 protein secreted by infective larvae of the human hookworm, Necator americanus, is under development as a recombinant vaccine. Recombinant Na-ASP-2 was expressed in Pichia pastoris, and the purified protein was characterized. At the 60 L scale, the 21.3 kDa recombinant protein was produced at a yield of 0.4 g/L. When formulated with Alhydrogel and injected into rats to determine immunological potency, three 50 microg doses of the formulated recombinant protein elicited geometric mean antibody titers up to 1:234,881. Rat anti-Na-ASP-2 antibody recognized larval-derived ASP-2 and also inhibited larval migration through skin in vitro. The processes developed and tested for the high yield production of recombinant Na-ASP-2 provide a foundation for clinical vaccine development.


Subject(s)
Helminth Proteins/immunology , Necator americanus/immunology , Pichia/genetics , Vaccines, Synthetic/immunology , Animals , Clinical Trials as Topic , Helminth Proteins/isolation & purification , Humans , Larva/immunology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Vaccination
12.
FASEB J ; 19(12): 1743-5, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16037096

ABSTRACT

The development of a vaccine would provide an important new tool for the control of human hookworm infection. On the basis of successful vaccination of laboratory animals with living irradiated, third-stage hookworm larvae (L3), we examined the antibody responses of individuals from hookworm endemic areas of Brazil and China against the most abundant L3 secreted antigens, the ancylostoma secreted proteins, ASP-1 and ASP-2. Logistic regression was used to investigate the effects of antibody isotype responses to ASPs on the risk of an individual harboring heavy hookworm infection. A significant protective association was observed between increasing anti-ASP-2 IgE levels and the risk of heavy hookworm infection. To confirm that ASP-2 is a protective antigen, laboratory dogs were immunized with recombinant ASP-2 formulated with the GlaxoSmithKline Adjuvant, AS03. Sera obtained from the immunized dogs exhibited high geometric mean antibody titers, immunoprecipitated native ASP-2 from L3 extracts and localized the site of ASP-2 expression to the glandular esophagus and body channels exiting to the cuticle. The sera also exhibited an increased ability to inhibit migration of L3 through tissue in vitro relative to sera from AS03-injected controls. Upon L3 challenge, the ASP-2 vaccinated dogs exhibited significant reductions in fecal egg counts and intestinal hookworm burden. These findings provide strong support for the development of an effective recombinant vaccine against hookworm infection in humans.


Subject(s)
Ancylostoma/pathogenicity , Antigens, Helminth/chemistry , Helminth Proteins/chemistry , Hookworm Infections/immunology , Hookworm Infections/prevention & control , Necator americanus/pathogenicity , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Dogs , Enzyme-Linked Immunosorbent Assay , Esophagus/metabolism , Female , Gastrointestinal Tract/pathology , Hookworm Infections/parasitology , Humans , Immunohistochemistry , Immunoprecipitation , Male , Middle Aged , Recombinant Proteins/chemistry , Regression Analysis , Time Factors , Vaccines
13.
Vaccine ; 23(24): 3123-30, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15837211

ABSTRACT

Syrian Golden hamsters were vaccinated with the recombinant fusion proteins Ay-ASP-2 and Ay-MTP-1 from the infective larvae of the hookworm Ancylostoma ceylanicum. Vaccines comprised each antigen alone or the combination of the two proteins. All vaccinated group developed high antibody titers (>1:40,000); coadministration of a second antigen did not significantly affect the magnitude of the antibody response. Following challenge, hamsters vaccinated with each single antigen exhibited reductions in worm burden (32% and 28% to Ay-ASP-2 and Ay-MTP-1, respectively) and fecal egg counts (56% and 43%, respectively). A vaccine cocktail, containing both antigens further reduced worm burden (36%) and fecal egg counts (59%) (p<0.001). Moreover, vaccination with the antigen cocktail significantly improved hemoglobin values (p=0.01) and body weights (p=0.001) compared to what achieved with either each antigen or adjuvant alone. Taken together, these data suggest that combination of two or more antigens may present an effective vaccine development strategy to improve protection and/or disease symptoms in affected individuals.


Subject(s)
Ancylostoma/immunology , Ancylostomiasis/prevention & control , Antigens, Helminth/immunology , Helminth Proteins/immunology , Hookworm Infections/prevention & control , Metalloproteases/immunology , Amino Acid Sequence , Ancylostomiasis/immunology , Ancylostomiasis/parasitology , Animals , Antibodies, Helminth/analysis , Antibodies, Helminth/biosynthesis , Body Weight , Cloning, Molecular , Cricetinae , Feces/parasitology , Hemoglobins/metabolism , Hookworm Infections/immunology , Hookworm Infections/parasitology , Larva/immunology , Mesocricetus , Metalloproteases/isolation & purification , Molecular Sequence Data , Parasite Egg Count , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
14.
Int J Parasitol ; 35(3): 303-13, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15722082

ABSTRACT

Hookworm infection persists as a public health problem in developing nations. Vaccine-based strategies offer the best chance of long-term control. Aspartyl protease inhibitors from parasitic nematodes are highly immunogenic, and have been suggested as potential vaccine antigens. An aspartyl protease inhibitor, API-1, was cloned and characterised from the hookworms Ancylostoma caninum and Ancylostoma ceylanicum. Using sequence from the hookworm expressed sequence tag project, specific primers were designed and used to amplify Ac-api-1 from A. caninum infective L3 cDNA by PCR. Amplicons from the 5' and 3' ends were cloned, sequenced, and combined to create an 874-bp full-length composite sequence of the Ac-api-1 gene. The A. ceylanicum api-1 cDNA of 878 bp was cloned from L3 cDNA using the A. caninum primers. The amino acid sequences of hookworm orthologues were nearly identical, and database searching indicated they belonged to the aspin family, a group of nematode specific aspartyl protease inhibitors that includes the Ascaris pepsin inhibitor PI-3. Ac-api-1 mRNA was detected by reverse transcriptase PCR in eggs, L1, L3 and adult life cycle stages. A polyclonal antiserum against Escherichia coli expressed recombinant Ac-API-1 detected the protein in adult A. caninum excretory/secretory products, but not in those from activated infective larvae. Immunolocalisation experiments using the antiserum indicated that Ac-API-1 is present primarily in the pseudocoelomic fluid in adult hookworms. Soluble, yeast-expressed Ac-API-1 failed to inhibit pepsin or a hookworm gut aspartyl protease in vitro, but inhibited approximately 30% of the proteolytic activity of adult excretory/secretory products. The pseudocoleomic location, presence in all life cycle stages, lack of inhibitory activity against pepsin, and inhibitory activity against excretory/secretory products suggest that Ac-API-1 inhibits an unidentified, putative aspartyl protease secreted by adult hookworms, and may be released as an enzyme-inhibitor complex. The highly immunogenic properties of nematode aspins suggest that Ac-API-1 represents a promising target for a recombinant hookworm vaccine.


Subject(s)
Ancylostoma/enzymology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Protease Inhibitors/analysis , Amino Acid Sequence , Ancylostoma/genetics , Ancylostoma/growth & development , Ancylostoma/immunology , Animals , Antigens, Helminth/analysis , Cloning, Molecular/methods , DNA, Complementary/genetics , DNA, Helminth/genetics , Life Cycle Stages , Molecular Sequence Data , Protease Inhibitors/chemistry , Protease Inhibitors/immunology , Protein Structure, Secondary , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment
15.
J Mol Biol ; 346(3): 801-14, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15713464

ABSTRACT

Human hookworm infection is a major cause of anemia and malnutrition of adults and children in the developing world. As part of on-going efforts to control hookworm infection, The Human Hookworm Vaccine Initiative has identified candidate vaccine antigens from the infective L3 larval stages of the parasite, including a family of pathogenesis-related (PR) proteins known as the Ancylostoma-secreted proteins (ASPs). A novel crystal structure of Na-ASP-2, a PR-1 protein secreted by infective larvae of the human hookworm Necator americanus, has been solved to resolution limits of 1.68 A and to an R-factor of 17% using the recombinant protein expressed in and secreted by Pichia pastoris. The overall fold of Na-ASP-2 is a three-layer alphabetaalpha sandwich flanked by an N-terminal loop and a short, cysteine-rich C terminus. Our structure reveals a large central cavity that is flanked by His129 and Glu106, two residues that are well conserved in all parasitic nematode L3 ASPs. Na-ASP-2 has structural and charge similarities to chemokines, which suggests that Na-ASP-2 may be an extra-cellular ligand of an unknown receptor. Na-ASP-2 is a useful homology model for NIF, a natural antagonistic ligand of CR3 receptor. From these modeling studies, possible binding modes were predicted. In addition, this first structure of a PR-1 protein from parasitic helminths may shed light on the molecular basis of host-parasite interactions.


Subject(s)
Helminth Proteins/chemistry , Helminth Proteins/immunology , Necator americanus/immunology , Necatoriasis/prevention & control , Amino Acid Sequence , Animals , Antigens, Helminth/chemistry , Antigens, Helminth/genetics , Base Sequence , Binding Sites , Chemotactic Factors/chemistry , Cloning, Molecular , Crystallography, X-Ray , DNA, Helminth/genetics , Helminth Proteins/genetics , Humans , Ligands , Macrophage-1 Antigen/metabolism , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Molecular Weight , Necator americanus/genetics , Necator americanus/pathogenicity , Necatoriasis/immunology , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Peptide Hydrolases/immunology , Protein Conformation , Sequence Homology, Amino Acid , Static Electricity , Vaccines/chemistry , Vaccines/genetics
16.
Biochemistry ; 43(22): 6959-67, 2004 Jun 08.
Article in English | MEDLINE | ID: mdl-15170333

ABSTRACT

Beta-amyloid (Abeta) is a major pathological determinant of Alzheimer's disease (AD). Both active and passive immunization studies have shown that antibodies against Abeta are effective in decreasing cerebral Abeta levels, reducing Abeta accumulation, and attenuating cognitive deficits in animal models of AD. However, the therapeutic potential of these antibodies in human AD patients is limited because of adverse inflammatory reactions and cerebral hemorrhaging associated with the treatments. Here we show that single chain variable fragments (scFv's) represent an attractive alternative to more conventional antibody-based therapeutics to reduce Abeta toxicity. The binding affinities and binding epitopes of two different scFv's to Abeta were characterized using a surface plasmon resonance (SPR) biosensor. An scFv binding the 17-28 region of Abeta effectively inhibited in vitro aggregation of Abeta as determined by thioflavin T (ThT) fluorescence staining and atomic force microscopy (AFM) analysis, while an scFv binding the carboxyl-terminal region of Abeta (residues 29-40) did not inhibit aggregation. The scFv to the 17-28 region when co-incubated with Abeta not only decreased aggregation but also eliminated any toxic effects of aggregated Abeta on the human neuroblastoma cell line, SH-SY5Y. The ability of scFv's to inhibit both aggregation and cytotoxicity of Abeta indicates that scFv's have potential therapeutic value for treating AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/immunology , Immunoglobulin Variable Region/pharmacology , Neuroblastoma/metabolism , Neurons/drug effects , Plaque, Amyloid/drug effects , Benzothiazoles , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/isolation & purification , Microscopy, Atomic Force , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Peptide Library , Surface Plasmon Resonance , Thiazoles , Tumor Cells, Cultured
17.
J Infect Dis ; 189(10): 1952-61, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15122534

ABSTRACT

We expressed a catalytically active cysteine protease, Ac-CP-2, from the blood-feeding stage of the canine hookworm Ancylostoma caninum and vaccinated dogs with the purified protease. Dogs acquired high-titer, antigen-specific antibody responses, and adult hookworms recovered from the intestines of vaccinated dogs were significantly smaller than hookworms from control dogs. There was also a marked decrease in fecal egg counts and the number of female hookworms in vaccinated dogs. Ac-CP-2 is expressed by the parasite in the brush-border membrane of its alimentary canal, and anti-Ac-CP-2 antibodies were bound to the gut of hookworms from vaccinated dogs, which suggests that these antibodies were ingested by the parasites with their blood meal. IgG from vaccinated dogs decreased proteolytic activity against a peptide substrate by 73%, which implies that neutralizing antibodies were induced by vaccination. These results indicate that cysteine proteases involved in parasite nutrition are promising candidates as vaccines against hookworm disease.


Subject(s)
Ancylostoma/immunology , Ancylostomiasis/veterinary , Cysteine Endopeptidases/immunology , Dog Diseases/parasitology , Protozoan Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Ancylostoma/genetics , Ancylostomiasis/immunology , Ancylostomiasis/parasitology , Ancylostomiasis/prevention & control , Animals , Antibodies, Protozoan/blood , Dog Diseases/immunology , Dog Diseases/prevention & control , Dogs , Feces/parasitology , Female , Intestines , Male , Parasite Egg Count/veterinary , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sex Ratio , Vaccination , Vaccines, Synthetic/immunology
18.
J Infect Dis ; 189(5): 919-29, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14976610

ABSTRACT

cDNAs encoding 2 Ancylostoma-secreted proteins (ASPs), Ancylostoma ceylanicum (Ay)-ASP-1 and Ay-ASP-2, were cloned from infective third-stage larvae (L3) of the hookworm A. ceylanicum and were expressed as soluble recombinant fusion proteins secreted by the yeast Pichia pastoris. The recombinant fusion proteins were purified, adjuvant formulated, and injected intramuscularly into hamsters. Hamsters vaccinated either by oral vaccination with irradiated L3 (irL3) or by injections of the adjuvants alone served as positive and negative controls, respectively. Anti-ASP-1 and anti-ASP-2 antibody titers exceeded 1 : 100000. Each vaccinated hamster was challenged orally with 100 L3. Two groups of vaccinated hamsters (i.e., those vaccinated with either irL3 or ASP-2 formulated with Quil A) exhibited significant reductions in adult hookworm burdens, compared with control hamsters. The hookworms recovered from the hamsters vaccinated with ASP-2 plus Quil A were reduced in length. Splenomegaly, which was observed in control hamsters, was not seen in hamsters vaccinated with either irL3 or ASP-2 formulated with Quil A. These results indicate that ASP-2 is a promising molecule for the development of a hookworm vaccine.


Subject(s)
Ancylostoma/genetics , Helminth Proteins/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Ancylostoma/growth & development , Animals , Base Sequence , Cloning, Molecular , Conserved Sequence , DNA Primers , DNA, Complementary/genetics , Larva , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid
19.
Int J Parasitol ; 33(11): 1245-58, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-13678639

ABSTRACT

Hookworm infection is one of the most important parasitic infections of humans, possibly outranked only by malaria as a cause of misery and suffering. An estimated 1.2 billion people are infected with hookworm in areas of rural poverty in the tropics and subtropics. Epidemiological data collected in China, Southeast Asia and Brazil indicate that, unlike other soil-transmitted helminth infections, the highest hookworm burdens typically occur in adult populations, including the elderly. Emerging data on the host cellular immune responses of chronically infected populations suggest that hookworms induce a state of host anergy and immune hyporesponsiveness. These features account for the high rates of hookworm reinfection following treatment with anthelminthic drugs and therefore, the failure of anthelminthics to control hookworm. Despite the inability of the human host to develop naturally acquired immune responses to hookworm, there is evidence for the feasibility of developing a vaccine based on the successes of immunising laboratory animals with either attenuated larval vaccines or antigens extracted from the alimentary canal of adult blood-feeding stages. The major antigens associated with each of these larval and adult hookworm vaccines have been cloned and expressed in prokaryotic and eukaryotic systems. However, only eukaryotic expression systems (e.g., yeast, baculovirus, and insect cells) produce recombinant proteins that immunologically resemble the corresponding native antigens. A challenge for vaccinologists is to formulate selected eukaryotic antigens with appropriate adjuvants in order to elicit high antibody titres. In some cases, antigen-specific IgE responses are required to mediate protection. Another challenge will be to produce anti-hookworm vaccine antigens at high yield low cost suitable for immunising large impoverished populations living in the developing nations of the tropics.


Subject(s)
Antigens, Helminth/isolation & purification , Hookworm Infections/prevention & control , Vaccines, Synthetic , Adolescent , Adult , Aged , Ancylostomiasis/immunology , Ancylostomiasis/prevention & control , Animals , Brazil/epidemiology , Child , China/epidemiology , Chronic Disease , Hookworm Infections/epidemiology , Hookworm Infections/immunology , Humans , Larva , Middle Aged , Necatoriasis/immunology , Necatoriasis/prevention & control , Prevalence , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL
...