Abstract
Entamoeba histolytica, which causes amebic colitis and liver abscess, is considered a major enteric pathogen in residents and travelers to developing countries where the disease is endemic. Interaction of this protozoan parasite with the intestine is mediated through the binding of the trophozoite stage to intestinal mucin and epithelium via a galactose and N-acetyl-d-galactosamine (Gal/GalNAc) lectin comprised of a disulfide linked heavy (ca. 180 kDa) and light chain (ca. 35 kDa) and a noncovalently bound intermediate subunit (ca. 150 kDa). Our efforts to develop a vaccine against this pathogen have focused on an internal 578 amino acid fragment, designated LecA, located within the cysteine-rich region of the heavy chain subunit because: (i) it is a major target of adherence-blocking antibodies of seropositive individuals and (ii) vaccination with his-tagged LecA provides protection in animal models. We developed a purification process for preparing highly purified non-tagged LecA using a codon-optimized gene expressed in Escherichia coli. The process consisted of: (i) cell lysis, collection and washing of inclusion bodies; (ii) solubilization and refolding of denatured LecA; and (iii) a polishing gel filtration step. The purified fragment existed primarily as a random coil with β-sheet structure, contained low endotoxin and nucleic acid, was highly immunoreactive, and elicited antibodies that recognized native lectin and that inhibited in vitro adherence of trophozoites to CHO cells. Immunization of CBA mice with LecA resulted in significant protection against cecal colitis. Our procedure yields sufficient amounts of highly purified LecA for future studies on stability, immunogenicity, and protection with protein-adjuvant formulations.
Keywords: Entamoeba histolytica, Lectin, Amebiasis, Diarrhea, LecA vaccine
1. Introduction
Amebic colitis and liver abscess are due to infection with the enteric protozoan parasite Entamoeba histolytica. This parasite has recently been separated using modern diagnostic techniques from the nonpathogenic parasite E. dispar, which is more common and identical in appearance to E. histolytica [11]. The World Health Organization (WHO) estimates that approximately 50 million people worldwide suffer from invasive amebic infection each year, resulting in 40–100 thousand deaths annually [28,46]. Carefully conducted serologic studies in Mexico, where amebiasis is endemic, demonstrated antibodies to E. histolytica in 8.4% of the population [7]. In the urban slum of Fortaleza, Brazil, 25% of the people tested carried antibody to E. histolytica and prevalence of anti-amebic antibodies in children aged 6–14 was 40% [5]. A prospective study of preschool children in a slum of Dhaka, Bangladesh demonstrated new E. histolytica infection in 39% of children >1 year, with 10% having an E. histolytica infection associated with diarrhea and 3% with dysentery [17]. Amebiasis also is the second most common cause of diarrhea in returning travelers [14]. Recently the Global Enteric Multicenter Study (GEMS) has demonstrated E. histolytica infection to be among the top 15 pathogens resulting in severe diarrhea in infants and children in Africa and Asia [23]. Thus, there is an increasing recognition of the burden of infection due to this protozoan parasite.
E. histolytica invades tissue and causes clinical disease following ingestion of the infectious cyst form of the parasite from fecally contaminated food or water [2,9,34,35,42]. Excystation of the amebic trophozoites occurs in the intestinal lumen. Trophozoites adhere to the colonic mucus and epithelial cells through interaction of a Gal/GalNAc-specific lectin [29,31]. The trophozoite kills host epithelial and immune cells in a process that requires the Gal/GalNAc lectin. E. histolytica resists the host’s immune response and survives to cause extra-intestinal infection such as amebic liver abscess.
Several different E. histolytica proteins have been studied as potential vaccines. These include the serine-rich protein, a 29-kDa cysteine-rich antigen, and the Gal/GalNAc-specific lectin [37–41]. The Gal/GalNAc-specific lectin has been examined in the greatest detail and results support its evaluation as a potential vaccine candidate [4,21,22,36,37]. The LecA domain encompasses the critical neutralizing antibody epitopes for amebic adherence, killing and endocytosis of host cells. In children an IgA antibody response against LecA is associated with immunity. Preliminary studies have demonstrated that the LecA alum-absorbed parenteral vaccine provides protection from amebic colitis in a murine model. Protection provided by the vaccine in mice correlates with the frequency of antigen-specific CD4+ T cells that produce intracellular IFN-γ, whereas in children both fecal IgA and IFN-γ, are associated with protection.
The Gal/GalNAc lectin is a 260 kDa heterotrimer of highly conserved disulfide-linked heavy (Hgl) and light (Lgl) subunits non-covalently associated with an intermediate subunit (Igl)[3,10,13,27,30,32,33,43,45]. The carbohydrate recognition domain (CRD) is a cysteine-rich region within Hgl recognized by adherence-inhibitory MAb [25,26]. Native lectin can be purified from E. histolytica cultures, but not in amounts sufficient as a vaccine candidate. We have focused on a region located within Hgl designated “LecA” (aa 578–1154) as a vaccine candidate because (i) it is a major target of the cell mediated and humoral immune response in seropositive individuals and (ii) vaccination with a his-tagged version provides protection in animal models [16,19]. We now describe a new scalable purification process for nontagged LecA and demonstrate LecA-mediated protection in a recently developed mouse model of amebic colitis that more accurately mirrors amebic colitis in humans rather than the liver abscess model that was used in past studies.
2. Materials and methods
2.1. Reagents, his tagged LecA, and protein determination
Chemicals were purchased from Sigma–Aldrich (St. Louis, MO) and Fisher Scientific (Waltham, MA) unless otherwise noted. Purification processes were done with ACS or higher grade chemicals to minimize metal ion contamination. Media preparations were prepared using non-animal based Veggie™ reagents from EMD4 Biosciences (Gibbstown, NJ). His-tagged LecA was prepared as previously described [19]. The Thermo Scientific/Pierce Modified Lowry Assay (Rockford, IL) was used to determine protein concentration [24].
2.2. LecA cloning and expression
The lecA clone was codon-optimized and synthesized by DNA2.0 (Menlo Park, CA) in the vector pJexpress401 that contains the kanamycin resistance (Kanr) gene and a T5 promoter for gene expression. Expression was done in Escherichia coli HMS174 (EMD4Biosciences, Gibbstown, NJ). Transformation was performed following the manufacturer’s recommendations. 2-L shaking flasks containing 1 L of 2 × YT media + kan (50 μg/mL) were inoculated from an overnight culture, and incubated at 37 °C with shaking. Induction was initiated at OD600 of 0.6–0.8 by the addition of isopropyl-β-d-thiogalactopyranoside to a final concentration of 1 mM and continued for 3 h. Cells were collected by centrifugation, with a typical yield per 1-L of 5.25 ± 0.3 g wet cell weight. Cells were stored frozen at −20 °C.
2.3. Purification
Cell lysis and subsequent washing steps were modified from previously described procedures [8]. Cell pellets were thawed on ice and suspended in Inclusion Body Wash Buffer (IBWB) consisting of 2%CHAPS, 20 mM Tris–HCl, 10 mM EDTA, pH 8.0, at 5 mL per gram wet cell weight. Lysis was performed with sonication and inclusion bodies (IB) were collected by centrifugation. Following lysis, the IB preparation was washed twice with IBWB and twice with an excess of 25% isopropyl alcohol (IPA). IB were solubilized with 7 M guanidine-HCl, 0.15 M reduced glutathione, 0.1 M Tris–HCl, 2 mM EDTA, and 25 mM DTT at pH 8.0, and gently stirred at 4 °C.
Following solubilization, the preparation was clarified by centrifugation. To facilitate refolding, the IB preparation was added dropwise with gentle stirring to a 50-fold volume of 50 mM Tris–HCl, pH 8.0 containing 0.5 M L-arginine and 0.6 mM oxidized glutathione. Refolding was completed overnight at 4 °C without stirring. The refolded sample was brought to room temperature, diluted 1:2 in 1X PBS, pH 7.4, and concentrated 2-fold using Vivaflow 50 crossflow cassettes (30,000 cutoff, Sartorius Stedim North America, Bohemina, NY). The concentration and dilution process was repeated an additional three times. Following the final buffer exchange, the sample was concentrated 6-fold, filtered through a 0.45 μm membrane filter, and concentrated 10-fold using Amicon Ultra-15 Centrifugal filter units (10,000 cutoff, Millipore, Burlington, MA).
The concentrate was applied to a Superdex S-200 XK26/60 column (prep grade, GE Healthcare, Piscataway, NJ) equilibrated with PBS and eluted in the downward flow mode at a flow rate of 2.5 mL/min. Fractions were assayed for A280 and A254, and analyzed for protein by SDS-PAGE. LecA peak fractions were pooled and stored at 2–8 °C.
2.4. Isoelectric focusing and electrophoresis
LecA was applied to the first dimension ZOOM® strip (Life Technologies, Carlsbad, CA) and electrofocusing was performed at 200 V for 1 h, 450 V for 45 min, 750 V for 45 min, and 2000 V for 1.5 h. The ZOOM® strip was then cast into a NuPAGE® 4–12% Bis-Tris ZOOM® gel and sealed with 0.5% agarose. Staining was done with Bio-Safe™ Coomassie stain. Commercially available NuPAGE® 4–12% Bis–Tris gels were used for SDS-PAGE. LecA was run using NuPAGE® MOPS SDS buffer kit according to the manufacturer’s instructions. For Western blots, 7F4 MAb, 3F4 MAb, polyclonal antibody against CRD, and anti-E. coli antibodies (Abcam®, Cambridge, MA) were used. Native-PAGE was performed using the Native PAGE™ Novex® Bis-Tris 4–12% acrylamide gel system. Silver staining was performed using the SilverQuest™ kit.
2.5. Characterization studies
The apparent molecular mass was determined by size exclusion chromatography on a Superdex 200 XK26/60 column [1]. Circular dichroism (CD) spectra were measured using an Aviv model 400 spectropolarimeter (Lakewood, NJ) in 0.1 cm cuvettes. Endotoxin levels were determined using the FDA-licensed Endosafe PTS™ cartridges and reader from Charles River Laboratories (Charleston, SC). Results were confirmed by PTS kinetic LAL analysis and kinetic chromogenic testing at Charles River Laboratories. Double-stranded DNA and RNA were quantified using the Qubit™ Fluorometer and their respective kits (Life Technologies). Sequencing analysis was performed at the Protein Sciences Facility at the University of Illinois (Urbana–Champaign). For N-terminal sequence analysis, proteins were transferred to PVDF membranes, and sequencing was performed on a Procise 494 HT Perkin-Elmer instrument using Edman chemistry. Results were analyzed using the NCBI-eukaryotes database. Immunoreactivity with 7F4 MAb, 3F4 MAb, 1G7, 3D12, and anti-CRD antibody (Fig. 1) was determined by endpoint titration with the E. HISTOLYTICA II ELISA (TechLab, Inc., Blacksburg, VA). Analysis of best fit curves was determined by simple linear regression. All MAbs have been described previously [25,26].
2.6. Reaction of anti-LecA antiserum with native lectin and LecA
New Zealand white rabbits (female, 5 lb) were injected subcutaneously with 200 μg LecA emulsified in Complete Freund Adjuvant followed two weeks later by boosts at 1-week intervals with 100 μg LecA emulsified in Incomplete Freund Adjuvant. All animal work was approved by the Animal Care Committee at Cocalico Biologicals, Inc. (Reamstown, PA). Reactivity of anti-LecA antiserum was determined by endpoint titration using microwells coated with immobilized native lectin or LecA.
2.7. CHO cell adherence assay
Chinese hamster ovary (CHO) cells were grown and maintained in α-MEM medium, and E. histolytica strain HM1:IMSS trophozoites were grown as previously described [12]. The adherence assay was performed as described by Ravdin and Guerrant [35]. E. histolytica trophozoites (1 × 104) were preincubated with rabbit preimmune or test serum on ice for 1 h. Trophozoites and CHO cells were then mixed, centrifuged together and incubated on ice for 90 min. Just prior to microscopic analysis, the tubes were vortexed briefly and cells counted on a haemocytometer. Adherence was measured as the number of trophozoites having at least three adherent CHO cells and reported as percent rosettes formation. At least 100 amebae were counted in each tube and each sample was done in triplicate.
2.8. Immunizations, antigen detection, and E. histolytica challenge
Four week old male CBA/J mice were obtained from the Jackson Laboratory and maintained under specific-pathogen-free conditions at the University of Virginia. Fifteen mice per group were immunized subcutaneously at weeks 0, 2 and 4 with adjuvant EM014 alone (control group), EM014 + LecA or EM014 + his-LecA (20 μg antigen per mouse per immunization) in a 100 μl final volume [20]. Each mouse received a total of three immunizations at 2-week intervals (weeks 0, 2 and 4). All the mice were challenged intracecally three weeks after the final boost with E. histolytica strain HM1: IMSS which had been sequentially passed through the mouse cecum. Mice were challenged with 2 × 106 log phase trophozoites in 150 μl, injected intracecally after laparotomy [18]. The animals were sacrificed one week after the challenge and cecal contents obtained. Typically, 200 μl of cecal contents were used for amebic determination with the TechLab E. histolytica II kit, and the remaining cecal contents were cultured in TYI-S-33 medium for five days at 37 °C.
2.9. Statistical analysis
All statistical analysis involving protection studies was done using GraphPad Prism software. Chi square test was used to compare values for protected and unprotected groups, while Mann–Whitney test was used to compare fecal antigen load values. Significance for the adherence assay was calculated using Student’s t-test.
3. Results
3.1. LecA expression, purification, and characterization
The engineered organism grown in animal-free Veggie™ yeast extract and peptone gave expression levels comparable to media containing beef products, with >80% of the expressed LecA present as Inclusion Bodies (IB) (data not shown). More than 80% of contaminants were removed by simple washing of IB. Guanidine–HCl effectively solubilized washed IB, with refolding being done using a previously described arginine-assisted process [18]. The refolding process involved dilution of the denatured protein into arginine-containing buffer followed by concentration/dilution steps to incorporate NaCl. Analysis by immunoassay confirmed that refolding had occurred properly. Size exclusion of the refolded, solubilized concentrate removed DNA, small nucleic acids, and aggregated proteins (Fig. 2). The main peak contained LecA and two minor peaks of low MWr polypeptides. Most of the endotoxin eluted prior to the primary LecA peak.
SDS-PAGE and silver staining showed a major band, confirmed as LecA by N-terminal sequencing and tryptic digest analysis, which had an estimated molecular mass of 70 kDa, in line with the predicted value (Fig. 3). The major band constituted >95% of the Coomassie-stained material. Several minor contaminants, representing RNA polymerase, transcription repair coupling factor, and lac repressor, represented <5% of the total protein. The apparent molecular mass, which was determined with 5 mM dithiothreitol in the sample and 0.5 mM dithiothreitol in the column buffer, was roughly twice the size observed by SDS-PAGE. Monoclonal antibodies and polyclonal antibodies specific for LecA and CRD gave identical patterns by Western blot analysis (Fig. 3). In addition to the conformational mAbs 3F4 and 7F4 already discussed, mAbs with a linear epitope (mAb 1G7) and another conformational epitope (mAb 3D12) gave comparable ELISA results for both native Lectin and LecA (data not shown). Immunoblot analysis with anti-E. coli antiserum did not reveal E. coli contaminants; however, sequence analyses revealed the three minor E. coli contaminants listed above. Analytical native gel electrophoresis showed all three lots to be identical. All three lots had low levels of endotoxin (≤0.1 EU/μg), both by in-house and external testing, and DNA and RNA levels ≤ 82 ng/mg and <20 ng/mg, respectively (Table 1). Simple linear regression analysis showed that immunoreactivity with nontagged LecA was higher than with his-tagged LecA, with values closer to those obtained with native lectin. LecA that had been heat-treated (boiling for 2 min) was no longer immunoreactive. Anti-LecA antiserum reacted with LecA at a titer of 50,000, and with native lectin at a titer of 100,000. LecA stored at 2–8 °C for 18 months or lyophilized retained the same immunoreactivity level and exhibited the same CD spectra as freshly prepared LecA.
Table 1.
Lot # | Total proteina (mg) | Endotoxinb (EU/μg) | DNAc (ng/mg) | RNAc (ng/mg) | Analytical sensitivityd (ng detected) |
Linear regression analysisd |
---|---|---|---|---|---|---|
071111RSW | 27.0 | 0.01 | 51 | <20 | 0.14 | 0.71 |
071111LAB | 12.9 | 0.01 | 82 | <20 | 0.11 | 0.77 |
071411LAB | 24.7 | 0.02 | 61 | <20 | 0.31 | 0.74 |
Protein was determined using the Thermo Scientific/Pierce Modified Lowry Assay (Rockford, IL).
Endotoxin levels were determined using the FDA-licensed Endosafe PTS™ cartridges and reader from Charles River Laboratories.
Double-stranded (ds) DNA and RNA were quantified using the Qubit™ Fluorimeter and Life Technologies kits.
Analytical sensitivity and linear regression analysis was determined using a modified quantitative E. HISTOLYTICA II kit.
3.2. Anti-LecA antiserum prevents parasite adherence to monolayer cells
We determined whether anti-LecA antibodies recognized the parasite in vitro and blocked the parasite-host cell interaction, using the CHO cell adherence assay. Amebic cells were pretreated with preimmune or test rabbit anti-LecA antiserum, mixed with CHO cells, and the ability of amebae to adhere to CHO cells was quantitated. Preincubation with antiserum significantly decreased adherence from 24.7 ± 3.9% to 7.3 ± 2% (at 1:10 dilution, p < 0.005) and from 31 ± 6 to 15.3 ± 3.5% (at 1:100 dilution, p < 0.005) in comparison to the sham serum (Fig. 4). These results showed that anti-LecA antiserum recognized the amebic surface lectin and interfered with the ability of the parasite to bind CHO cells. Addition of galactose blocked the binding of trophozoites to CHO cells as expected.
3.3. Immunization with LecA confers protection
CBA/J mice were immunized with adjuvant EM014 alone (control group), EM014 + LecA or EM014 + his tagged LecA as described under the methods section. Mice were challenged with 2 × 106 log phase trophozoites at week 7. On week 8, amebic infection in the ceca from sacrificed mice was evaluated by ELISA and by culture in TYI-S-33 media. Vaccination with LecA resulted in a significant decrease in amebic load. The mean OD value for the vaccinated mice was 1.17 ± 0.5 whereas that for the control mice was 5.4 1.1 (p < 0.05, Fig. 5A). 60% mice in the LecA vaccinated group showed ± values below the baseline (OD ≤ 0.05) as compared to only 20% in the control group, indicating very low levels of antigen in the vaccinated mice. Culture results reflected the ELISA observations as only 20% were infected in the LecA-immunized group compared to 60% mice in the control group (p < 0.05, Fig. 5B). The protection, measured by the standard methods of antigen load and culture positivity, were comparable between untagged and His-tagged LecA formulations.
4. Discussion
We previously showed that his-tagged LecA afforded protection in a mouse model of amebic colitis [16]. However, there were difficulties in achieving his-tagged LecA preparations of a consistent degree of purity, and there were concerns about the potential long-term effect of anti-his-tag antibodies elicited by vaccines with his-tagged protein preparations. Therefore, we developed a scalable purification process that yielded sufficient amounts of highly purified non-tagged LecA for characterization, immunogenicity, and protection studies. Our results showed that LecA was produced primarily as IB, regardless of the E. coli host, vector, or growth conditions tested. We initially suspected that IB resulted from disulfide linkages due to the unusually high cysteine content in this cysteine-rich portion of the heavy chain subunit, but were unable to prevent their formation with reducing/denaturing agents. By using guanidine-HCl and arginine in a redox environment with reduced and oxidized glutathione in the refolding process, we were able to solubilize and refold the protein in a reproducible fashion. Refolding may be optimized by using low protein concentrations, but for the purposes of our scalable purification process, this was not optimal because of the amount of protein purified. L-arginine has previously been shown to increase repulsive rather than attraction interaction between proteins [18]. This effect, in turn, improves the refolding process by increasing the yield of soluble protein. Thus, L-arginine incorporated into the refolding step for LecA made our process more scalable. Although the formation of IB made purification more challenging, it allowed us to remove most of the contaminating lysate proteins by washing and collection by centrifugation of IB.
Size exclusion enabled us to obtain LecA preparations of consistent purity with reduced levels of endotoxin. Levels of <20 EU/mL have been recommended for recombinant subunit vaccines, although this concentration still is unclear since endotoxin levels often are specific to a licensed vaccine, and generalized guidance statements only refer to the U.S. Food and Drug Administration endotoxin limit of 5 EU/kg/h for intravenous drugs [15,44]. For our studies, we can now produce LecA with more accurately defined endotoxin levels that fall under guideline levels and which is protective. This is important since, as noted by Brito and Singh [6], immunogenicity may decrease as endotoxin is removed during a purification process.
By incorporating a gentle L-arginine refolding process, we were able to improve folding and decrease aggregation, as demonstrated by immunoassay analysis and comparison with native lectin and his-tagged lectin, both of which are protective in animal models [16,19]. Linear regression analysis showed that refolded LecA exhibited slope values higher than those observed with his-tagged LecA, and that when heat-treated, LecA was no longer reactive. These findings indicated proper recognition and binding of antibodies to the protein, further supporting proper refolding. The monoclonal antibodies used in our evaluation have been shown previously to bind to conformationally dependent epitopes [25]. All bind effectively to his-tagged LecA and nontagged LecA in a manner consistent with that observed with native lectin, indicating proper refolding of LecA. These observations, and the demonstration that (i) anti-LecA antiserum inhibited the binding of CHO cells to trophozoites, (ii) immunization with LecA reduced antigen load in a mouse model of amebic colitis, and (iii) immunization with LecA protected mice from infection, showed that non-tagged LecA was as effective as his-tagged LecA and native lectin, and that key antigenic properties of lectin were preserved [16,19].
Over 90% of human amebiasis is due to amebic colitis, and not the liver abscess for which animal models have long been available. In this study, we used the recently developed CBA mouse model of amebic colitis in which mice are infected by injection of amebic trophozoites into the cecum. Chronic E. histolytica infection with ulcerative colitis develops in 60–90% of infected C3H mice and reproducibly 80% of CBA mice; histopathologically the lesions in the mouse colon closely resemble human amebic colitis. Without intervention, E. histolytica infection and amebic colitis persist for months. We now have demonstrated that LecA which is not his tagged avoids potential problems that may occur with his tagged vaccines, that LecA can now be produced in a scalable manner, and importantly, that LecA prevents amebic infection and colitis in a highly relevant mouse model of amebiasis.
As a purified recombinant antigen, LecA requires the use of an adjuvant formulation in order to enhance immunogenicity and protective efficacy. EM014 consists of GLA-SE (a synthetic TLR4 ligand formulated in a stable oil-in-water emulsion) combined with the synthetic TLR9 ligand CpG 1826. EM014 induces a Th1-type immune response, resulting in the highest levels of splenocyte IFN-γ production and protective efficacy against amebiasis when compared to a range of other adjuvant formulations, including Freund’s adjuvant [16,44]. EM014 represents an adjuvant formulation with a potential path forward as a component of a vaccine product, since both GLA-SE and CpGs have been evaluated in completed and ongoing clinical trials with recombinant vaccine antigens (www.clinicaltrials.gov).
The high molecular mass of LecA, more than twice the predicted size or size determined by SDS-PAGE, supported the circular dichroism results showing that at the molecular level, about half of the molecule exists primarily in an unfolded state. Therefore, one possibility is that LecA exists with a large Stokes radius accompanied by a high percentage of random coil structure and β-sheet structure. Alternatively, it is possible that in the native configuration, LecA exists as a dimer, which would also explain the high mass. Titration with Ellman’s reagent detected all predicted 31 sulfhydryl groups in denatured LecA even though none was accessible in nondenatured LecA. Thus, the odd number of sulfhydryl groups may indicate that at least one of the sulfhydryl groups is accessible in the native molecule, and participates in dimer formation. Size exclusion profiles did not change in the presence of dithiothreitol, suggesting that if such a disulfide linkage exists, it is very stable. In any event, our data support the idea that sulfhydryl groups in LecA are engaged in intra-molecular interactions. The purified peptide is stable since we did not observe any further unfolding, as determined by circular dichroism spectra or loss of immunoreactivity following storage at 18 months or after lyophilization.
The process that we developed yielded >20 mg of highly purified LecA protein per liter of culture, the purified preparations have acceptably low endotoxin levels, the LecA elicits a protective response in mice, and the purification process is scalable for cGMP. The availability of highly purified LecA will allow us to proceed with antigen dose/potency studies, long term stability testing, further analysis of peptide secondary structure, and development of additional acceptance criteria for quality control. The Gal/GalNAc lectin is the best studied antigen of E. histolytica, and currently LecA is the most logical choice as a vaccine candidate. We now have a process to produce sufficient amounts of a properly folded recombinant peptide for new detailed studies on this antigen and its consideration as a vaccine. This is a significant step toward generating a human amebic vaccine.
Acknowledgments
This study was funded by NIH Grant 1R43 AI085938 (Alum-Absorbed Subunit Vaccine to Prevent Intestinal Amebiasis), NIH Grant U01-AI070384 (Cooperative Research Partnership for an Amebic Colitis Vaccine), and by TechLab, Inc. (Blacksburg, VA). We thank Michael Flaherty and Eric Rigel (TechLab, Inc.) for performing isoelectric focusing and native gel electrophoresis, Blake Stott and Joel Herbein (TechLab, Inc.) for providing native Gal/GalNAc lectin from E. histolytica, and Brian Hall for analyzing residual immunoreactivity of heat-treated LecA. D. Lyerly is co-founder and co-owner of TechLab, Inc. L. Barroso and K. Pedersen are employees of TechLab, Inc.
References
- [1].Andrews P. Estimation of the molecular weights of proteins by Sephadex gel-filtration. Biochem J. 1944;91:222–33. doi: 10.1042/bj0910222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Beaver PC, Blanchard JL, Seibold HR. Invasive amebiasis in naturally infected new world and old world monkeys with and without clinical disease. Am J Trop Med Hyg. 1988;39:343–52. doi: 10.4269/ajtmh.1988.39.343. [DOI] [PubMed] [Google Scholar]
- [3].Beck DL, Tanyuksel M, Mackey A, Haque R, Trapaidze N, Pearson WR, et al. Sequence conservation of the Gal/GalNAc lectin from clinical isolates. Exp Parasitol. 2002;101:157–63. doi: 10.1016/s0014-4894(02)00113-3. [DOI] [PubMed] [Google Scholar]
- [4].Beving D, Soong C, Ravdin J. Oral immunization with a recombinant cysteine-rich section of the Entamoeba histolytica galactose-inhibitable lectin elicits an intestinal secretory immunoglobulin A response that has in vitro adherence inhibition activity. Infect Immun. 1996;64:1473–6. doi: 10.1128/iai.64.4.1473-1476.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Braga LL, Lima AAM, Sears CL, Newman RD, Wuhib T, Paiva CA, et al. Seroepidemiology of Entamoeba histolytica in a slum in Northeastern Brazil. Am J Trop Med Hyg. 1996;55:693–7. doi: 10.4269/ajtmh.1996.55.693. [DOI] [PubMed] [Google Scholar]
- [6].Brito LA, Singh M. Acceptable levels of endotoxin in vaccine formulations during preclinical research. J Pharm Sci. 2010;100:34–7. doi: 10.1002/jps.22267. [DOI] [PubMed] [Google Scholar]
- [7].Caballero-Salcedo A, Viveros-Rogel M, Salvatierra B, Tapia-Conyer R, Sepulveda-Amor J, Gutierrez G, et al. Seroepidemiology of amebiasis in Mexico. Am J Trop Med Hyg. 1994;50:412–9. doi: 10.4269/ajtmh.1994.50.412. [DOI] [PubMed] [Google Scholar]
- [8].Cash HL, Whitham CV, Hooper LV. Refolding, purification, and characterization of human and murine RegIII proteins expressed in Escherichia coli. Prot Express Purif. 2006;48:151–9. doi: 10.1016/j.pep.2006.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Chadee K, Meerovitch E. Entamoeba histolytica: early progressive pathology in the cecum of the gerbil (Meriones unguiculatus) Am J Trop Med Hyg. 1985;34:283–91. doi: 10.4269/ajtmh.1985.34.283. [DOI] [PubMed] [Google Scholar]
- [10].Cheng XJ, Hughes MA, Houston CD, Loftus B, Gilchrist CA, Lockhart LA, et al. The 150 kDa Gal/GalNAc lectin co-receptor of Entamoeba histolytica is a member of a gene family containing multiple CXXC sequence motifs. Infect Immun. 2001;69:5892–8. doi: 10.1128/IAI.69.9.5892-5898.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Diamond LS, Clark CG. A redescription of Entamoeba histolytica Schaudinn 1903 (emended Walker 1911) separating it from Entamoeba dispar (Brumpt 1925) J Eukary Microbiol. 1993;40:340–4. doi: 10.1111/j.1550-7408.1993.tb04926.x. [DOI] [PubMed] [Google Scholar]
- [12].Diamond LS, Harlow DR, Cunnick CC. A new medium for the axenic cultivation of Entamoeba histolytica and other Entamoeba. Trans R Soc Trop Med Hyg. 1978;72:431–2. doi: 10.1016/0035-9203(78)90144-x. [DOI] [PubMed] [Google Scholar]
- [13].Dodson JM, Lenkowski PW, Jr, Eubanks AC, Jackson TFHG, Napodano J, Lyerly DM, et al. Infection and immunity mediated by the carbohydrate recognition domain of the Entamoeba histolytica Gal/GalNAc lectin. J Infect Dis. 1999;179:460–6. doi: 10.1086/314610. [DOI] [PubMed] [Google Scholar]
- [14].Freedman DO, Weld LH, Kozarsky PE, Fisk T, Robins R, von Sonnenburg F, et al. Spectrum of disease and relation to place of exposure among ill returned travelers. N Engl J Med. 2006;354:119. doi: 10.1056/NEJMoa051331. [DOI] [PubMed] [Google Scholar]
- [15].Guideline on Validation of the Limulus Amebocyte Lysate Test as an End-Product Endotoxin test for Human and Animal Parenteral Drugs, Biological Products, and Medical Devices. U.S. Dept. of Health and Human Services, FDA; Dec, 1987. [Google Scholar]
- [16].Guo XL, Barroso L, Becker L, Lyerly DM, Vedvick TS, Reed SG, et al. Protection against intestinal amebiasis by a recombinant vaccine is transferable by T cells and mediated by gamma interferon. Infect Immun. 2009;77:3909–18. doi: 10.1128/IAI.00487-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Haque R, Ali IKM, Sack RB, Ramakrishnan G, Farr BM, Petri WA., Jr Amebiasis and mucosal IgA antibody against the Entamoeba histolytica adherence lectin in Bangladeshi children. J Infect Dis. 2001;183:1787–93. doi: 10.1086/320740. [DOI] [PubMed] [Google Scholar]
- [18].Ho J, Middelberg A, Ramage P, Kocher H. The likelihood of aggregation during protein renaturation can be assessed using the second virial coefficient. Protein Sci. 2003;12:708–16. doi: 10.1110/ps.0233703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Houpt E, Barroso L, Lockhart LA, Wright R, Lyerly D, Petri r WA. Prevention of intestinal amebiasis by vaccination with the Entamoeba histolytica Gal/GalNac lectin. Vaccine. 2004;22:611–7. doi: 10.1016/j.vaccine.2003.09.003. [DOI] [PubMed] [Google Scholar]
- [20].Houpt ER, Glembocki DJ, Obrig TG, Moskaluk CA, Lockhart LA, Wright RL, et al. The mouse model of amebic colitis reveals mouse strain susceptibility to infection and exacerbation of disease by CD4+ T cells. J Immunol. 2001;169:4496–503. doi: 10.4049/jimmunol.169.8.4496. [DOI] [PubMed] [Google Scholar]
- [21].Ivory C, Chadee K. Intranasal immunization with Gal-inhibitable lectin plus an adjuvant of CpG oligodeoxynucleotides protects against Entamoeba histolytica challenge. Infect Immun. 2007;75:4917–22. doi: 10.1128/IAI.00725-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Ivory C, Keller K, Chadee K. CpG-Oligodeoxynucleotide is a potent adjuvant with an Entamoeba histolytica Gal-inhibitable lectin vaccine against amoebic liver abscess in gerbils. Infect Immun. 2006;74:528–36. doi: 10.1128/IAI.74.1.528-536.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Kotloff K, Nataro J, Blackwelder W, Nasrin D, Rarag T, Panchalingam S, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case–control study. Lancet. 2013 May 13; doi: 10.1016/S0140-6736(13)60844-2. [DOI] [PubMed] [Google Scholar]
- [24].Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265–75. [PubMed] [Google Scholar]
- [25].Mann B, Lockhart L. Molecular analysis of the Gal/GalNAc adhesion of Entamoeba histolytica. J Euk Microbiol. 1998;45:13S–6S. doi: 10.1111/j.1550-7408.1998.tb04518.x. [DOI] [PubMed] [Google Scholar]
- [26].Mann BJ, Chung CY, Dodson JM, Ashley LS, Braga LL, Snodgrass TL. Neutralizing monoclonal antibody epitopes of the Entamoeba histolytica galactose adhesion map to the cysteine-rich extracellular domain of the 170-kilodalton subunit. Infect Immun. 1993;61:1772–8. doi: 10.1128/iai.61.5.1772-1778.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Mann BJ, Torian BE, Vedvick TS, Petri WA., Jr Sequence of the cysteine-rich heavy subunit of the galactose lectin of Entamoeba histolytica. Proc Natl Acad Sci USA. 1991;88:3248–52. doi: 10.1073/pnas.88.8.3248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Petri WA., Jr Recent advances in amebiasis. Crit Rev Clin Lab Sci. 1996;33:1–37. doi: 10.3109/10408369609101485. [DOI] [PubMed] [Google Scholar]
- [29].Petri WA, Smith RD, Schlesinger PH, Ravdin JI. Isolation of the galactose-binding lectin which mediates the in vitro adherence of Entamoeba histolytica. J Clin Invest. 1987;80:1238–44. doi: 10.1172/JCI113198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Petri WA, Jr, Chapman MD, Snodgrass T, Mann BJ, Broman J, Ravdin JI. Subunit structure of the galactose and N-acetyl-d-galactosamine inhibitable adherence lectin of Entamoeba histolytica. J Biol Chem. 1989;264:3007–12. [PubMed] [Google Scholar]
- [31].Petri WA, Jr, Mann BJ, Haque R. The bittersweet interface of parasite and host; lectin-carbohydrate interactions during human invasion by the parasite Entamoeba histolytica. Ann Rev Microbiol. 2002;56:39–64. doi: 10.1146/annurev.micro.56.012302.160959. [DOI] [PubMed] [Google Scholar]
- [32].Pillai DR, Wan PSK, Yau YCW, Ravdin JI, Kain KC. The cysteine-rich region of the Entamoeba histolytica adherence lectin (170-kilodalton subunit) is sufficient for high affinity Gal/GalNAc-specific binding in vitro. Infect Immun. 1999;67:3836–41. doi: 10.1128/iai.67.8.3836-3841.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Ramakrishnan G, Ragland BD, Purdy JE, Mann BJ. Physical mapping and expression of gene families encoding the N-acetyl-d-galactosamine adherence lectin of Entamoeba histolytica. Mol Microbiol. 1996;19:91–100. doi: 10.1046/j.1365-2958.1996.356885.x. [DOI] [PubMed] [Google Scholar]
- [34].Ravdin JI. Amebiasis now. Am J Trop Med Hyg. 1989;41:40–8. doi: 10.4269/ajtmh.1989.41.40. [DOI] [PubMed] [Google Scholar]
- [35].Ravdin JI, Guerrant RL. Role of adherence of cytopathogenic mechanisms of Entamoeba histolytica Study with mammalian tissue culture cells and human erythrocytes. J Clin Invest. 1981;68:1305–13. doi: 10.1172/JCI110377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Ravdin J, Shain D, Kelsall B. Antigenicity, immunogenicity and vaccine efficacy of the galactose-specific adherence protein of Entamoeba histolytica. Vaccine. 1993;11:241–6. doi: 10.1016/0264-410x(93)90024-r. [DOI] [PubMed] [Google Scholar]
- [37].Ryan E, Butterton J, Zhang T, Baker M, Stanley S, Calderwood S. Oral immunization with attenuated vaccine strains of Vibrio cholera expressing a dodecapeptide repeat of the serine-rich Entamoeba histolytica protein fused to the cholera toxin B subunit induces systemic and mucosal antiamebic and Anti-V. cholera antibody responses in mice. Infect Immun. 1997;65:3118–25. doi: 10.1128/iai.65.8.3118-3125.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Soong C, Kain K, Abd-Alla M, Jackson T, Ravdin J. A recombinant cysteine-rich section of the Entamoeba histolytica galactose-inhibitable lectin is efficacious as a subunit vaccine in the gerbil model of amebic liver abscess. J Infect Dis. 1995;171:645–51. doi: 10.1093/infdis/171.3.645. [DOI] [PubMed] [Google Scholar]
- [39].Soong C, Torian B, Abd-Alla M, Jackson T, Gatharim V, Ravdin J. Protection of gerbils from amebic liver abscess by immunization with recombinant Entamoeba histolytica 29-kilodalton antigen. Infect Immun. 1995;63:472–7. doi: 10.1128/iai.63.2.472-477.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Stanley S. Progress towards development of a vaccine for amebiasis. Clin Microbiol Rev. 1997;10:637–49. doi: 10.1128/cmr.10.4.637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Sultan F, Jin L, Jobling M, Holmes R, Stanley S. Mucosal immunogenicity of a holotoxin-like molecule containing the serine-rich Entamoeba histolytica protein (SREHP) fused to the A2 domain of cholera toxin. Infect Immun. 1998;66:462–8. doi: 10.1128/iai.66.2.462-468.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Takeuchi A, Phillips BP. Electron microscope studies of experimental Entamoeba histolytica infection in the guinea pig. Am J Trop Med Hyg. 1975;24:34–48. doi: 10.4269/ajtmh.1975.24.34. [DOI] [PubMed] [Google Scholar]
- [43].Tannich E, Ebert F, Horstmann RD. Primary structure of the 170 kDa surface lectin of pathogenic Entamoeba histolytica. Proc Natl Acad Sci USA. 1991;88:1849–53. doi: 10.1073/pnas.88.5.1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Vedvick TS, Carter L, Moulton G, Goto Y, Bertholet S, Reed SG, et al. An improved manufacturing process for a recombinant polyprotein vaccine. BioPharm Int. 2008 Jan 17; [Google Scholar]
- [45].Vines RR, Ramakrishnan G, Rogers J, Lockhart L, Mann BJ, Petri WA., Jr Regulation of adherence and virulence by the Entamoeba histolytica lectin cytoplasmic domain, which contains the β2 integrin motif. Mol Biol Cell. 1998;9:2069–79. doi: 10.1091/mbc.9.8.2069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].WHO Amoebiasis. WHO. Week Epidemiol Record. 1997;72:97–100. [Google Scholar]